Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Dec 1.
Published in final edited form as: Curr Opin HIV AIDS. 2009 Jul;4(4):266–273. doi: 10.1097/COH.0b013e32832c91dd

HIV-prevention science at a crossroads: advances in reducing sexual risk

Sten H Vermund a, Katherine L Allen a, Quarraisha Abdool Karim b,c
PMCID: PMC2995247  NIHMSID: NIHMS253545  PMID: 19532063

Abstract

Purpose of review

We review the current state of evidence-based prevention strategies for reducing sexual transmission of HIV. The combined programmatic and scientific efforts through 2008 to reduce sexual transmission of HIV have failed to reduce substantially the global pandemic.

Recent findings

Prevention interventions to reduce HIV infection target behavioral, biomedical, and structural risk factors. Some of these prevention strategies have been evaluated in randomized clinical trials (RCTs) with HIV seroincidence endpoints. When RCTs are not feasible, a variety of observational and quasiexperimental research approaches can provide insight as to program effectiveness of specific strategies. Only five RCTs have demonstrated a notable decrease in sexually acquired HIV incidence. These include the Mwanza study of syndromic management of sexually transmitted diseases and three male circumcision trials in East Africa; a microbicide trial reported in 2009 shows substantial promise for the efficacy of PRO 2000 (0.5% gel).

Summary

The combined programmatic and scientific efforts to reduce sexual transmission of HIV have made incremental progress. New prevention tools are needed to stem the continued spread of HIV, though microbicides and vaccines will take many more years to develop, test, and deploy. Combination strategies of existing modalities should be tested to evaluate the potential for more proximate prevention benefits.

Keywords: circumcision, HIV, microbicide, microbicide, prevention, sexual behavior, sexually transmitted disease

Introduction

The spread of the human immunodeficiency virus (HIV) represents the worst global pandemic since the bubonic plague in 14th century Europe and Asia. Between 1981 and 2007, 64.9 million people are estimated to have been infected with HIV of whom about half have died [1]. In 2007 alone, 2.5 million persons were infected with HIV –90% of whom live in Sub-Saharan Africa, Asia, and Eastern Europe [2]. About 80% of all newly infected persons acquired infection through sexual contact [3,4]. The combined programmatic and scientific efforts to reduce sexual transmission of HIV have failed to address the pandemic effectively.

Diversity in how the epidemic manifests itself is a key challenge. The virus has considerable genetic variability, inhibiting the development of an effective HIV vaccine. The diversity of infected populations reflects the diverse modes of transmission – heterosexual, homosexual, needle-related, blood and blood product-related, and perinatal. Diversity in the route of transmission and in the burden of disease presents a mosaic of dynamic subepidemics, each driven by a variety of behavioral, structural and biological risk factors. In this review, we emphasize evidence-based prevention efforts targeting sexual transmission, particularly those published in 2007 and 2008.

HIV prevention: a failure of implementation

A failure of vision and/or political commitment to implement what we know works has limited the success of HIV-prevention efforts. UNAIDS estimated that in 2005, fewer than one in five people could access any known HIV-prevention strategies [5]. When prevention interventions are accessible and coupled with behavioral changes substantial decreased in HIV incidence can occur. The ‘ABC’ of delaying sexual debut in youth (abstinence), reducing the number of sexual partners and reducing concurrent sexual relationships (be faithful), and correct, consistent condom use reduced HIV seroincidence in Uganda, Thailand, and perhaps elsewhere [610]. As background HIV prevalence rises within populations, there is a poor correlation between personal risk factors and probability of HIV acquisition. The probability of exposure per sexual encounter, related to background prevalence, becomes the single biggest risk factor for getting infected in generalized epidemics and within at-risk subgroups.

This epidemic has been further exacerbated by ignoring the complex array of societal and contextual factors that increase a person’s risk for HIV infection. Focusing exclusively on individual and personal risk factors and dependence on intrinsically driven behaviors will not adequately address current epidemic trajectories. Community stigma inhibits community education, testing for HIV, and access to prevention and care. Sex and power dynamics including survival strategies and economic pressures are critical extrinsic factors that diminish individual autonomy and control of HIV-risk reduction. To reduce sexual risk inherently requires, at a minimum, engaging the sexual dyad, whether heterosexual or homosexual. Exclusion of women from the formal economy, as well as societal expectations of subservience and procreation fuel vulnerability of women through the adoption of a variety of survival strategies. This vulnerability can manifest in sex work to feed one’s children, sexual liaisons to enable school continuation, remaining in abusive marital relations, sexual exploitation in the work-place, forced marriage, and exposure to life-threatening HIV/AIDS stigma [1114]. These are best described in Africa and Asia, but may apply anywhere in the world.

Preventing HIV infection: randomized controlled trials

Observational studies have generated a substantial body of literature on a range of biomedical, behavioral, and structural interventions associated with lowered HIV-risk behaviors and/or HIV incidence [1517]. However, prevention interventions have only rarely proved efficacious when tested in randomized clinical trials (RCTs) with HIV endpoints. Furthermore, there has been a poor correlation between behavior change and reduction in HIV-incidence rates. Between 2005 and 2007, several important, innovative Phase III HIV prevention RCTs were in progress (Table 1) [1839].

Table 1.

Summary of recent clinical trials of HIV prevention

Author Intervention Population Outcome Strength of association (95% CI) Comments
Selected prevention trials in progress with HIV endpoints
HPTN 043 Protocol chair: T.J. Coates Expanded VCT and bridging to care South Africa; Tanzania; Zimbabwe; Thailand Seroprevalence surveys (to estimate HIV incidence) In progress Project Accept, community- RCT in progress to test impact of expanded HIV testing and community mobilization on HIV incidence
HPTN 052 Protocol chair: S. Cohen Reduce partner transmission via early ART Malawi; South Africa; Zimbabwe; India; Thailand; Botswana Seroconversion in discordant couples In progress Individual randomization of discordant couples to assess whether or not early treatment of the HIV+ person reduces HIV infections in partner
Control of STDs to reduce HIV acquisition
Wawer et al. [18] Periodic mass therapy for STI Rakai, Uganda HIV incidence RR = 0.97 (0.81–1.16) Mature HIV epidemic, relatively low rates of curable STIs, high HSV-2 rates, mass periodic STD therapy
Kamali et al. [19] STD syndromic management Masaka, Uganda HIV incidence RR = 0.91 (0.56–1.47) Mature HIV epidemic, relatively low rates of curable STIs, most infections occurred between regular partners
Grosskurth et al. [20] STD syndromic management Mwanza, Tanzania HIV incidence RR = 0.58 (0.42–0.79) Syndromic management worked to reduce HIV incidence. High curable STI rates among core transmitters.
Celum et al. [21••] HSV-2 suppression South Africa; Zimbabwe; Zambia; Peru; USA HIV incidence RR = 1.16 (0.83–1.62) The larger of the two HSV-2 suppression studies, it suggests that nose of acyclovir given to participants may not have been sufficient to reduce HIV susceptibility resulting from HSV-2 infection.
Watson-Jones et al. [22] Herpes simplex virus type 2 (HSV-2) suppression Tanzania HIV incidence RR = 1.08 (0.64–1.83) HSV-2 suppression trial showed no benefit for HIV prevention.
Adult male circumcision
Auvert et al. [23] Adult male circumcision Orange Farm, South Africa HIV incidence RR = 0.49 (0.28–0.84) All three trials with remarkably similar results, the strongest effect size of any RCTs for prevention with HIV end points. Male circumcision seen as a powerful tool to reduce HIV transmission; its utility in public health practice now being assessed.
Gray et al. [24] Adult male circumcision Rakai, Uganda HIV incidence RR = 0.47 (0.28–0.78)
Bailey et al. [25] Adult male circumcision Kisumu, Kenya HIV incidence RR = 0.40 (0.24–0.68)
Behavior change-based interventions
Gregson et al. [26] Integrated community and clinical HIV control Manicaland, Zimbabwe HIV incidence RR = 1.27 (0.92–1.75) No notable impact on HIV seroincidence.
Jewkes et al. [27••] Stepping Stones behavioral intervention South Africa HIV incidence RR = 0.95 (0.67–1.35) A significant decrease in HSV- 2 acquisition was observed as well as a decrease in intimate partner violence suggesting that study impacted HIV-risk behaviors but not HIV transmission.
Latkin et al. [28] Network–oriented peer education Thailand; USA Behavioral endpoints No significant decrease in risk behaviors (sexual or injection) Incidence rates were not as high as expected in either arm of the study. It was underpowered to determine the effect of peer education on HIV incidence.
Cowan et al. [29] Multicomponent behavioral intervention in adolescents Zimbabwe HIV incidence In progress Baseline data suggest that the intervention was implemented prior to onset of sexual activity of adolescents.
Pronyk et al. [30] Microfinance-based structural Intervention South Africa HIV incidence, unprotected sex, intimate partner violence RR = 1.06 (0.66–1.69), RR = 0.89 (0.66–1.19), RR = 1.02 (0.85–1.23) The study lacked the precision necessary to adequately measure effect size. The number of villages included in the study was determined by feasibility of implementing the intervention, and was not necessarily large enough to detect a small effect size.
Padian et al. [31] Diaphragm South Africa, Zimbabwe HIV incidence RR = 1.05 (0.84–1.32) Condoms were used extensively in the control arm and not in the intervention arm. This suggests that diaphragm was as efficacious as condoms at preventing HIV acquisition given that risk for acquisition was approximately equal.
Microbicide trials
Halpern et al. [32] Microbicide (Cellulose sulfate gel) Nigeria HIV Incidence HR = 0.8 (0.3–1.8) This trial was suspended early due to data from a parallel trial, which suggested that cellulose sulfate increased risk for HIV acquisition.
Abdool Karim et al. [41] Microbicide (0.5%PRO 2000 gel) Malawi, South Africa; Zambia; Zimbabwe; United States HIV incidence HR = 0.7 (0.4–1.0) 30% efficacy suggested borderline statistical significance.
Feldblum et al. [33] Microbicide (0.1% SAVVY gel) Nigeria HIV incidence HR = 1.7 (0.9–3.5) Trial was halted due to lower-than-expected HIV incidence among participants. There was not adequate power to detect effect of SAVVY gel.
Peterson et al. [34] Microbicide (0.1% SAVVY gel) Ghana HIV incidence HR = 0.88 (0.33–2.27) Trial was halted due to lower-than-expected HIV incidence among participants. There was not adequate power to detect effect of SAVVY gel.
Skoler-Karpoff et al. [35] Microbicide (Carraguard) South Africa HIV incidence HR = 0.87 (0.69–1.09) Low levels of reported adherence prevented the trial from having the power needed to detect efficacy of Carraguard.
Van Damme et al. [36] Microbicide (6% cellulose sulfate gel) Benin; South Africa; India; Uganda HIV incidence HR = 1.61 (0.86–3.01) Safety concerns for this product.
Vaccine trials
Buchbinder et al. [37••] Cell-mediated immunity vaccine North America; South America; Caribbean; Australia HIV incidence HR = 1.2 (0.6–2.2) This trial was halted early because protective efficacy was not observed in males in this cohort. Efficacy assess ment could not be determined in women because of low HIV acquisition rates.
Flynn et al. [38] Recombinant glycoprotein vaccine (VaxGen; bivalent subtype B/B rgp120) North America HIV incidence Vaccine efficacy: 0.06 (317–24) No vaccine benefits or harms noted.
Pitisuttithum et al. [39] Recombinant glycoprotein vaccine (VaxGen; bivalent subtype B/E rgp120) Thailand HIV incidence Vaccine efficacy: 0.1 (330.8–23.8) No vaccine benefits or harms noted.
*

This included: peer education, condom distribution, and syndromic management for sexually transmitted infections.

The study was terminated early due to insufficient incidence in study sites. Behavioral data were collected for secondary outcome measures and was used in final analysis. VCT, voluntary counseling and testing.

By 2007, as the trials reached completion or were stopped early for reasons of harm or futility, negative sentiments about HIV prevention science started to emerge. This situation was most notable in the vaccine and microbicide fields. Just months after a 2008 Science [40] article that discussed some of the current failures in the microbicide field and questioned the future of the microbicides, a promising microbicide result for PRO 2000 (0.5%) gel (discussed later) was presented at the 16th Conference on Retroviruses and Opportunistic Infections [41]. This good news notwithstanding, most recent prevention trials (with the important exception of the male circumcision trials) have not demonstrated efficacy in preventing HIV infection [27••,29,30,33,42].

The number of recently completed but unsuccessful prevention trials mask important progress in the efforts to reduce HIV infection. Building on observations in couples from Quinn et al. [43] and Fideli et al. [44] on the close correlation between viral load and HIV transmission, new opportunities for HIV prevention have emerged through expanding access to testing and knowledge of HIV status, linked to antiretroviral therapy (ART) for infected persons. This ‘test and treat’ concept has potential as an intervention for decreasing viral load in communities and thus reducing the probability of transmission at a population level. The use of ART as a prevention tool has already proven effective in prevention of mother-to-child HIV transmission (PMTCT) – women who are given ART during pregnancy and postpartum have a significantly decreased risk for transmission of virus to their newborn [4547]. However, use of ART, as a prevention tool requires patients to know their HIV-positive status, have access to care, and to adhere to medications necessary to lower viral loads and minimize antiviral drug resistance. With six drug classes now in use (http://www.fda.gov/oashi/aids/virals.html, accessed 30 March 2009), several of which are in affordable generic formulations, advances in the field of antiviral chemotherapy have made effective viral load reduction even more feasible.

Vaccines

The ‘Step’ study of the MRK Ad5 gag/pol/nef HIV-1 vaccine did not indicate vaccine benefit in adenovirus-seronegative persons and showed higher HIV serocon-version in adenovirus-seropositive persons [37••]. Deep tissue HIV invasion may occur in just 2 weeks or less, demonstrating how hard it will be to use a human immunologic barrier to block HIV, given the current state of knowledge [48].

Physical barriers

Although the effectiveness of male condoms is exceedingly well documented despite the lack of a RCT [49], condom usage rates are still low in most high-risk persons globally. The Zimbabwean MIRA trial did not suggest the female diaphragm to be more successful in preventing HIV compared with condom use alone [31]. Efficacy of female condoms remains unknown and unstudied in clinical trials; effectiveness data are also sparse.

Chemical barriers (topical microbicides)

Cellulose sulfate gel, 1.0% C31G (SAVVY), Carraguard, and Buffer-gel products failed to prevent HIV transmission in prevention RCTs [3236,41,50]. However, PRO 2000 (0.5%) gel, a nonspecific entry/fusion inhibitor, was 30% effective (0.05 <P ≤ 0.10 against placebo gel or no gel groups, P <0.05 against placebo and no gel groups combined) in reducing HIV incidence [41]. Microbicide trials with antiretroviral active agents (e.g., tenofovir) are in progress [51].

Oral prophylactic antiretroviral therapy

Animal data suggest that ART can be given before HIV exposure and continued for several weeks after exposure to reduce risk of HIV transmission [5255]. Preexposure prophylaxis (PrEP), some with postexposure (PEP) components are in progress worldwide in both men who have sex with men (MSM) and at-risk heterosexuals. Perhaps the most promising product (as of this 2009 writing) is tenofovir combined with emtricitabine (TDF/FTC or Truvada) due to its lower likelihood of emerging drug resistance, high genital tract concentrations, and long half-life [56,57]. If effective, implementation will be challenging, especially in more resource-limited settings where even HIV-infected persons are underserved with antiretroviral services. Nonetheless, if drug prices fall, decision analysis models suggest it could be a cost-effective intervention [58].

Male circumcision

Randomized clinical trials in South Africa, Uganda, and Kenya demonstrated approximately 50% seroincidence decline in circumcised heterosexual men [2325]. This has galvanized an effort to expand such services in high prevalence venues, given their high cost efficiency [5961]. As scale-up occurs, mitigation of harm from suboptimal surgery will be a challenge [6264]. Benefits of circumcision to MSM are unknown, and would only be expected to benefit men who are exclusively (or almost exclusively) insertive in their sexual acts [65,66].

Control of sexually transmitted infections

The suppression of herpes simplex virus type-2 infection with chronic acyclovir did not reduce HIV seroconversions in either MSM in the Americas or with high-risk heterosexual women in Africa [21••,22]. This was disappointing given the very strong correlation of HSV-2 infection with risk of HIV acquisition reported in observational studies; many in the field expected HSV-2 suppression to mimic adult male circumcision successes [67]. In addition to these more recent trials, previous community-RCTs have examined the effect of improved STI treatment on HIV transmission [1820,26]. The results of these studies were mixed suggesting that treatment of bacterial STI may be more effective in preventing HIV infection in settings with low-to-emerging HIV epidemics or mature epidemics with high incidence and where there is a high background prevalence of bacterial STI [6871].

Behavior change and youth

Work is in progress on an array of approaches and strategies [15]. A meta-analysis suggested no efficacy of abstinence-only-based behavior change in preventing HIV, but promise was shown for more comprehensive ‘abstinence-plus’ programs [7274]. A community RCT in Tanzania did not demonstrate reduced HIV incidence in adolescent girls, despite improving knowledge, attitudes, and reducing some sexual risk behaviors in boys [42]. The Stepping Stones initiative in rural South Africa was a behavioral intervention that did not reduce incidence of HIV in 15–26 year olds, but had a significant impact on reducing HSV-2 incidence and intimate partner violence [27••]. However, this study did not demonstrate a change in HIV rates, reinforcing that there are no good surrogate markers for HIV incidence itself [27••]. A RCT of risk reduction among Zimbabwean adolescents is in progress, having recruited a very high proportion of youth into the intervention vs. control groups before they became sexually active [29].

Structural intervention

Structural interventions are policy changes that encourage risk reduction. Raising cigarette taxes to discourage youth from smoking and banning smoking in public indoors areas are two familiar examples. The Microfinance for AIDS and Gender Equity (IMAGE) study combined a microfinance program with training in sex–power relationships and HIV-risk reduction in rural South Africa. Reductions in levels of intimate-partner violence were significant in the intervention arm along with reductions in HIV-risk behaviors [75], but HIV rates did not differ [30]. Although HIV results were disappointing, the RCT illustrates how multi-component interventions will be used to try to break the cycle of HIV transmission.

In summary, 28 years into the pandemic only five randomized controlled trials have demonstrated a decrease in HIV-incidence rates. The Mwanza trial demonstrated a 42% reduction in HIV seroincidence in persons living in communities with immediate availability of syndromic management of bacterial sexually transmitted diseases (STDs) [20]. This promising finding was not observed in the context of three subsequent phase III RCTS of STD intervention studies in the Rakai, Masaka, Manicaland regions [18,19,26]. A microbicide has shown promise in the HPTN035 study of PRO 2000 0.5% gel [41]. The protective effect of medical male circumcision in preventing female–male transmission of HIV has been confirmed consistently in three independently conducted phase III RCTs [2325]. The translation of these impressive finding has been limited by political, cultural/social and programmatic challenges [59,61,62,7681]. This leaves us with a limited arsenal of proven HIV-prevention strategies, though circumstantial evidence of male condom efficacy and salutary benefits of delayed sexual debut and partner reduction remain compelling.

Methodological challenges

Notwithstanding the scientific challenges in preventing HIV infection, the conduct of RCTs in HIV prevention has numerous additional challenges in design, selecting appropriate outcome measures for both efficacy and effectiveness studies including the lack of proxy markers for protection [82,83]. A key factor contributing to small effect size of interventions is the ethical obligation to provide risk reduction education in comparison or control populations, even in settings where there remains limited access to prevention in the real world. For example in the MIRA RCT, female diaphragms and condoms were compared against condoms alone [31]. Although women using diaphragms had the same transmission rates as those whose partners used condoms, it was interpreted widely that diaphragms were ineffective. A more correct interpretation was that they might have been equally efficacious as condoms, as condom use in the diaphragm group was uncommon, despite encouragements for their use.

The prevention community has also been slow to embrace combination prevention efficacy trials despite what we have learned from therapeutic trials or strategies in the PMTCT field about the importance of using more comprehensive intervention strategies targeting all potential routes of viral entry and/or replication. Instead, HIV-prevention efforts have adopted the approach of incremental/attributable risk reduction. Although these unitary strategies for HIV prevention are alluring in theory (lower cost, ease of implementation); research investments (from the USA National Institute of Allergy and Infectious Diseases, the United Kingdom Medical Research Council, and the Bill and Melinda Gates Foundation, for example) have disproportionately emphasized ‘magic bullets’ for HIV prevention (i.e., prophylactic vaccines, PrEP, and microbicides) compared with more complex multicomponent interventions. The latter could have epidemic impact of more immediacy and effectiveness [37••,40,84]. The authors of this review believe that future trials must combine interventions, each with small effects but with the potential for synergy if combined. Prevention science strategies and trial conduct needs to take into account the myriad of biological, behavioral, and structural factors that increase a person’s risk for infection at multiple levels. Rarely does an individual at risk of getting infected with HIV have a single intrinsic factor that might lead to infection.

Conclusion

A diverse biomedical and biobehavioral portfolio of research is essential for us to make progress in HIV control. Building on the tools provided by basic, clinical, and social sciences, we need to move new products and strategies into rigorous field testing. Implementation of single modality interventions will not solve the problem. History has taught us through PEPFAR and the Global Fund to Fight AIDS, tuberculosis and malaria that ‘where there is a will there is a way’. These therapeutic implementation successes have also highlighted the numerous challenges in the context of failing healthcare delivery systems, inadequate infrastructure and limited human resources to attain access, coverage and acceptability levels needed to make a difference to current epidemic trajectories [85,86,87].

Vaccines and microbicides are still seen as the holy grails of prevention as reflected in budgetary allocations. Although these approaches will remain an important part of our medium-to long-term strategy, we must invest in strategies with impact in the shorter term. Combinations of interventions based on existing tools such as the delaying sexual debut, partner fidelity, use of condom barriers (the abstinence, be faithful, correct condom use ‘ABC’ mantra), and male circumcision will be needed to complement whatever biomedical interventions are made available and are used by the target communities. We believe that the lessons learned suggest that multicomponent, multilevel interventions with careful monitoring and evaluation in implementation will be more likely to succeed than unitary biomedical approaches.

Acknowledgments

Our review research was supported by the HIV Prevention Trials Network (HPTN), sponsored by the National Institute of Allergy and Infectious Diseases, National Institute of Child Health and Human Development, National Institute on Drug Abuse, National Institute of Mental Health, and Office of AIDS Research, of the National Institutes of Health, the United States Department of Health and Human Services (U01 AI068619).

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

• of special interest

•• of outstanding interest

Additional references related to this topic can also be found in the Current World Literature section in this issue (pp. 337–338).

  • 1. [Accessed 5 January 2009];USAID Health: HIV/AIDS. World Wide Web URL: http://www.usaid.gov/our_work/global_health/aids/News/aidsfaq.html.
  • 2.UNAIDS. AIDS epidemic update: December 2007. Geneva: UNAIDS; 2007. [Google Scholar]
  • 3.UNAIDS. 2008 Report on the global AIDS epidemic. Geneva: UNAIDS; 2008. [Google Scholar]
  • 4.WHO, UNAIDS, UNICEF. Progress Report 2008. Geneva, Switzerland: WHO, UNAIDS, UNICEF; 2008. Towards Universal Access: scaling up priority HIV/ AIDS interventions in the health sector. [Google Scholar]
  • 5.WHO, UNAIDS, UNICEF. Towards Universal Access: Scaling up priority HIV/ AIDS interventions in the health sector. Geneva: WHO; 2008. [Google Scholar]
  • 6.Celentano DD, Nelson KE, Lyles CM, et al. Decreasing incidence of HIV and sexually transmitted diseases in young Thai men: evidence for success of the HIV/AIDS control and prevention program. AIDS. 1998;12:F29–F36. doi: 10.1097/00002030-199805000-00004. [DOI] [PubMed] [Google Scholar]
  • 7.Hallett TB, Aberle-Grasse J, Bello G, et al. Declines in HIV prevalence can be associated with changing sexual behaviour in Uganda, urban Kenya, Zimbabwe, and urban Haiti. Sex Transm Infect. 2006;82 (Suppl 1):i1–i8. doi: 10.1136/sti.2005.016014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kilian AH, Gregson S, Ndyanabangi B, et al. Reductions in risk behaviour provide the most consistent explanation for declining HIV-1 prevalence in Uganda. AIDS. 1999;13:391–398. doi: 10.1097/00002030-199902250-00012. [DOI] [PubMed] [Google Scholar]
  • 9.Nelson KE, Celentano DD, Eiumtrakol S, et al. Changes in sexual behavior and a decline in HIV infection among young men in Thailand. N Engl J Med. 1996;335:297–303. doi: 10.1056/NEJM199608013350501. [DOI] [PubMed] [Google Scholar]
  • 10.Rojanapithayakorn W, Hanenberg R. The 100% condom program in Thailand. AIDS. 1996;10:1–7. doi: 10.1097/00002030-199601000-00001. [DOI] [PubMed] [Google Scholar]
  • 11.Dunkle KL, Jewkes RK, Brown HC, et al. Gender-based violence, relationship power, and risk of HIV infection in women attending antenatal clinics in South Africa. Lancet. 2004;363:1415–1421. doi: 10.1016/S0140-6736(04)16098-4. [DOI] [PubMed] [Google Scholar]
  • 12.Fenton L. Preventing HIV/AIDS through poverty reduction: the only sustainable solution? Lancet. 2004;364:1186–1187. doi: 10.1016/S0140-6736(04)17109-2. [DOI] [PubMed] [Google Scholar]
  • 13.Gupta GR. How men’s power over women fuels the HIV epidemic. BMJ. 2002;324:183–184. doi: 10.1136/bmj.324.7331.183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Parker RG, Easton D, Klein CH. Structural barriers and facilitators in HIV prevention: a review of international research. AIDS. 2000;14 (Suppl 1):S22–S32. doi: 10.1097/00002030-200006001-00004. [DOI] [PubMed] [Google Scholar]
  • 15.Coates TJ, Richter L, Caceres C. Behavioural strategies to reduce HIV transmission: how to make them work better. Lancet. 2008;372:669–684. doi: 10.1016/S0140-6736(08)60886-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gupta GR, Parkhurst JO, Ogden JA, et al. Structural approaches to HIV prevention. Lancet. 2008;372:764–775. doi: 10.1016/S0140-6736(08)60887-9. [DOI] [PubMed] [Google Scholar]
  • 17.Padian NS, Buvé A, Balkus J, et al. Biomedical interventions to prevent HIV infection: evidence, challenges, and way forward. Lancet. 2008;372:585–599. doi: 10.1016/S0140-6736(08)60885-5. [DOI] [PubMed] [Google Scholar]
  • 18.Wawer MJ, Sewankambo NK, Serwadda D, et al. Control of sexually transmitted diseases for AIDS prevention in Uganda: a randomised community trial. Rakai Project Study Group. Lancet. 1999;353:525–535. doi: 10.1016/s0140-6736(98)06439-3. [DOI] [PubMed] [Google Scholar]
  • 19.Kamali A, Quigley M, Nakiyingi J, et al. Syndromic management of sexually-transmitted infections and behaviour change interventions on transmission of HIV-1 in rural Uganda: a community randomised trial. Lancet. 2003;361:645–652. doi: 10.1016/s0140-6736(03)12598-6. [DOI] [PubMed] [Google Scholar]
  • 20.Grosskurth H, Mosha F, Todd J, et al. Impact of improved treatment of sexually transmitted diseases on HIV infection in rural Tanzania: randomised controlled trial. Lancet. 1995;346:530–536. doi: 10.1016/s0140-6736(95)91380-7. [DOI] [PubMed] [Google Scholar]
  • 21••.Celum C, Wald A, Hughes J, et al. Effectofaciclovir on HIV-1 acquisition inherpes simplex virus 2 seropositive women and men who have sex with men: a randomised, double-blind, placebo-controlled trial. Lancet. 2008;371:2109–2119. doi: 10.1016/S0140-6736(08)60920-4. Daily acyclovir did not reduce HIV seroincidence among persons with HSV-seropositive status. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22•.Watson-Jones D, Weiss HA, Rusizoka M, et al. Effect of herpes simplex suppression on incidence of HIV among women in Tanzania. N Engl J Med. 2008;358:1560–1571. doi: 10.1056/NEJMoa0800260. This smaller study also demonstrated no benefit of HSV-2 suppression in prevention HIV infection. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Auvert B, Taljaard D, Lagarde E, et al. Randomized, controlled intervention trial of male circumcision for reduction of HIV infection risk: the ANRS 1265 Trial. PLoS Med. 2005;2:e298–e1298. doi: 10.1371/journal.pmed.0020298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gray RH, Kigozi G, Serwadda D, et al. Male circumcision for HIV prevention in men in Rakai, Uganda: a randomised trial. Lancet. 2007;369:657–666. doi: 10.1016/S0140-6736(07)60313-4. [DOI] [PubMed] [Google Scholar]
  • 25.Bailey RC, Moses S, Parker CB, et al. Male circumcision for HIV prevention in young men in Kisumu, Kenya: a randomised controlled trial. Lancet. 2007;369:643–656. doi: 10.1016/S0140-6736(07)60312-2. [DOI] [PubMed] [Google Scholar]
  • 26.Gregson S, Adamson S, Papaya S, et al. Impact and process evaluation of integrated community and clinic-based HIV-1 control: a cluster-randomised trial in eastern Zimbabwe. PLoS Med. 2007;4:e102–e1102. doi: 10.1371/journal.pmed.0040102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27••.Jewkes R, Nduna M, Levin J, et al. Impact of Stepping Stones on incidence of HIV and HSV-2 and sexual behaviour in rural South Africa: cluster randomised controlled trial. BMJ. 2008;337:a506–a1506. doi: 10.1136/bmj.a506. Although this behavioral RCT reduced HSV-2 rates and sexual risk, there was no impact on HIV seroincidence. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Latkin CA, Donnell D, Metzger D, et al. The efficacy of a network intervention to reduce HIV risk behaviors among drug users and risk partners in Chiang Mai, Thailand and Philadelphia, USA. Soc Sci Med. 2009;68:740–748. doi: 10.1016/j.socscimed.2008.11.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Cowan FM, Pascoe SJ, Langhaug LF, et al. The Regai Dzive Shiri Project: a cluster randomised controlled trial to determine the effectiveness of a multi-component community-based HIV prevention intervention for rural youth in Zimbabwe: study design and baseline results. Trop Med Int Health. 2008;13:1235–1244. doi: 10.1111/j.1365-3156.2008.02137.x. [DOI] [PubMed] [Google Scholar]
  • 30.Pronyk PM, Hargreaves JR, Kim JC, et al. Effect of a structural intervention for the prevention of intimate-partner violence and HIV in rural South Africa: a cluster randomized trial. Lancet. 2006;368:1973–1983. doi: 10.1016/S0140-6736(06)69744-4. [DOI] [PubMed] [Google Scholar]
  • 31.Padian NS, van der Straten A, Ramjee G, et al. Diaphragm and lubricant gel for prevention of HIV acquisition in southern African women: a randomised controlled trial. Lancet. 2007;370:251–261. doi: 10.1016/S0140-6736(07)60950-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Halpern V, Ogunsola F, Obunge O, et al. Effectiveness of cellulose sulfate vaginal gel for the prevention of HIV infection: results of a Phase III trial in Nigeria. PLoS ONE. 2008;3:e3784. doi: 10.1371/journal.pone.0003784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Feldblum PJ, Adeiga A, Bakare R, et al. SAVVY vaginal gel (C31G) for prevention of HIV infection: a randomized controlled trial in Nigeria. PLoS ONE. 2008;3:e1474. doi: 10.1371/journal.pone.0001474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Peterson L, Nanda K, Opoku BK, et al. SAVVY (C31G) gel for prevention of HIV infection in women: a Phase 3, double-blind, randomized, placebo-controlled trial in Ghana. PLoS ONE. 2007;2:e1312. doi: 10.1371/journal.pone.0001312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Skoler-Karpoff S, Ramjee G, Ahmed K, et al. Efficacy of Carraguard for prevention of HIV infection in women in South Africa: a randomised, double-blind, placebo-controlled trial. Lancet. 2008;372:1977–1987. doi: 10.1016/S0140-6736(08)61842-5. [DOI] [PubMed] [Google Scholar]
  • 36.Van Damme L, Govinden R, Mirembe FM, et al. Lack of effectiveness of cellulose sulfate gel for the prevention of vaginal HIV transmission. N Engl J Med. 2008;359:463–472. doi: 10.1056/NEJMoa0707957. [DOI] [PubMed] [Google Scholar]
  • 37••.Buchbinder SP, Mehrotra DV, Duerr A, et al. Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet. 2008;372:1881–1893. doi: 10.1016/S0140-6736(08)61591-3. The surprising result of this promising HIV vaccine approach was that were was no benefit for adenovirus seronegative persons, but seroincidence was elevated compare to placebo among adenovirus seropositives. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Flynn NM, Forthal DN, Harro CD, et al. Placebo-controlled phase 3 trial of a recombinant glycoprotein 120 vaccine to prevent HIV-1 infection. J Infect Dis. 2005;191:654–665. doi: 10.1086/428404. [DOI] [PubMed] [Google Scholar]
  • 39.Pitisuttithum P, Gilbert P, Gurwith M, et al. Randomized, double-blind, placebo-controlled efficacy trial of a bivalent recombinant glycoprotein 120 HIV-1 vaccine among injection drug users in Bangkok, Thailand. J Infect Dis. 2006;194:1661–1671. doi: 10.1086/508748. [DOI] [PubMed] [Google Scholar]
  • 40.Grant RM, Hamer D, Hope T, et al. Whither or wither microbicides? Science. 2008;321:532–534. doi: 10.1126/science.1160355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41•.Abdool Karim S, Coletti A, Richardson B, et al. Safety and effectiveness of vaginal microbicides buffer gel and 0.5% PRO 2000/5 gel for the prevention of HIV infection in women: results of the HPTN 035 Trial. CROI; Montreal, Canada: 2009. This is the first evidence of effectiveness of vaginal microbicide to prevention HIV transmission. The full manuscript will be published in 2009. [Google Scholar]
  • 42.Ross DA, Changalucha J, Obasi AI, et al. Biological and behavioural impact of an adolescent sexual health intervention in Tanzania: a community-randomized trial. AIDS. 2007;21:1943–1955. doi: 10.1097/QAD.0b013e3282ed3cf5. [DOI] [PubMed] [Google Scholar]
  • 43.Quinn TC, Wawer MJ, Sewankambo N, et al. Viral load and heterosexual transmission of human immunodeficiency virus type 1. Rakai Project Study Group. N Engl J Med. 2000;342:921–929. doi: 10.1056/NEJM200003303421303. [DOI] [PubMed] [Google Scholar]
  • 44.Fideli US, Allen SA, Musonda R, et al. Virologic and immunologic determinants of heterosexual transmission of human immunodeficiency virus type 1 in Africa. AIDS Res Hum Retroviruses. 2001;17:901–910. doi: 10.1089/088922201750290023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Garcia PM, Kalish LA, Pitt J, et al. Maternal levels of plasma human immunodeficiency virus type 1 RNA and the risk of perinatal transmission. Women and Infants Transmission Study Group. N Engl J Med. 1999;341:394–402. doi: 10.1056/NEJM199908053410602. [DOI] [PubMed] [Google Scholar]
  • 46.Mayaux MJ, Dussaix E, Isopet J, et al. Maternal virus load during pregnancy and mother-to-child transmission of human immunodeficiency virus type 1: the French perinatal cohort studies. SEROGEST Cohort Group. J Infect Dis. 1997;175:172–175. doi: 10.1093/infdis/175.1.172. [DOI] [PubMed] [Google Scholar]
  • 47.Mofenson LM, Lambert JS, Stiehm ER, et al. Risk factors for perinatal transmission of human immunodeficiency virus type 1 in women treated with zidovudine. Pediatric AIDS Clinical Trials Group Study 185 Team. N Engl J Med. 1999;341:385–393. doi: 10.1056/NEJM199908053410601. [DOI] [PubMed] [Google Scholar]
  • 48.Robb ML. Failure of the Merck HIV vaccine: an uncertain step forward. Lancet. 2008;372:1857–1858. doi: 10.1016/S0140-6736(08)61593-7. [DOI] [PubMed] [Google Scholar]
  • 49.Holmes KK, Levine R, Weaver M. Effectiveness of condoms in preventing sexually transmitted infections. Bull World Health Organ. 2004;82:454–461. [PMC free article] [PubMed] [Google Scholar]
  • 50.Cates W, Feldblum P. HIV prevention research: the ecstasy and the agony. Lancet. 2008;372:1932–1933. doi: 10.1016/S0140-6736(08)61824-3. [DOI] [PubMed] [Google Scholar]
  • 51.Balzarini J, Van Damme L. Microbicide drug candidates to prevent HIV infection. Lancet. 2007;369:787–797. doi: 10.1016/S0140-6736(07)60202-5. [DOI] [PubMed] [Google Scholar]
  • 52.Cranage M, Sharpe S, Herrera C, et al. Prevention of SIV rectal transmission and priming of T cell responses in macaques after local preexposure application of tenofovir gel. PLoS Med. 2008;5:e157. doi: 10.1371/journal.pmed.0050157. discussion e157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Denton PW, Estes JD, Sun Z, et al. Antiretroviral preexposure prophylaxis prevents vaginal transmission of HIV-1 in humanized BLT mice. PLoS Med. 2008;5:e16. doi: 10.1371/journal.pmed.0050016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Garcia-Lerma JG, Otten RA, Qari SH, et al. Prevention of rectal SHIV transmission in macaques by daily or intermittent prophylaxis with emtricita-bine and tenofovir. PLoS Med. 2008;5:e28. doi: 10.1371/journal.pmed.0050028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Subbarao S, Otten RA, Ramos A, et al. Chemoprophylaxis with tenofovir disoproxil fumarate provided partial protection against infection with simian human immunodeficiency virus in macaques given multiple virus challenges. J Infect Dis. 2006;194:904–911. doi: 10.1086/507306. [DOI] [PubMed] [Google Scholar]
  • 56.Dumond JB, Yeh RF, Patterson KB, et al. Antiretroviral drug exposure in the female genital tract: implications for oral pre and postexposure prophylaxis. AIDS. 2007;21:1899–1907. doi: 10.1097/QAD.0b013e328270385a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Paxton LA, Hope T, Jaffe HW. Preexposure prophylaxis for HIV infection: what if it works? Lancet. 2007;370:89–93. doi: 10.1016/S0140-6736(07)61053-8. [DOI] [PubMed] [Google Scholar]
  • 58.Paltiel AD, Freedberg KA, Scott CA, et al. HIV preexposure prophylaxis in the United States: impact on lifetime infection risk, clinical outcomes, and cost-effectiveness. Clin Infect Dis. 2009;48:806–815. doi: 10.1086/597095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Auvert B, Marseille E, Korenromp EL, et al. Estimating the resources needed and savings anticipated from roll-out of adult male circumcision in Sub-Saharan Africa. PLoS ONE. 2008;3:e2679. doi: 10.1371/journal.pone.0002679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kahn JG, Marseille E, Auvert B. Cost-effectiveness of male circumcision for HIV prevention in a South African setting. PLoS Med. 2006;3:e517. doi: 10.1371/journal.pmed.0030517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Williams BG, Lloyd-Smith JO, Gouws E, et al. The potential impact of male circumcision on HIV in Sub-Saharan Africa. PLoS Med. 2006;3:e262. doi: 10.1371/journal.pmed.0030262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bailey RC, Egesah O, Rosenberg S. Male circumcision for HIV prevention: a prospective study of complications in clinical and traditional settings in Bungoma, Kenya. Bull World Health Organ. 2008;86:669–677. doi: 10.2471/BLT.08.051482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Bailey RC, Plummer FA, Moses S. Male circumcision and HIV prevention: current knowledge and future research directions. Lancet Infect Dis. 2001;1:223–231. doi: 10.1016/S1473-3099(01)00117-7. [DOI] [PubMed] [Google Scholar]
  • 64.Mattson CL, Muga R, Poulussen R, et al. Feasibility of medical male circumcision in Nyanza Province, Kenya. East Afr Med J. 2004;81:230–235. doi: 10.4314/eamj.v81i5.9165. [DOI] [PubMed] [Google Scholar]
  • 65.Millett GA, Flores SA, Marks G, et al. Circumcision status and risk of HIV and sexually transmitted infections among men who have sex with men: a meta-analysis. JAMA. 2008;300:1674–1684. doi: 10.1001/jama.300.14.1674. [DOI] [PubMed] [Google Scholar]
  • 66.Vermund SH, Qian HZ. Circumcision and HIV prevention among men who have sex with men: no final word. JAMA. 2008;300:1698–1700. doi: 10.1001/jama.300.14.1698. [DOI] [PubMed] [Google Scholar]
  • 67.Abu-Raddad LJ, Magaret AS, Celum C, et al. Genital herpes has played a more important role than any other sexually transmitted infection in driving HIV prevalence in Africa. PLoS ONE. 2008;3:e2230. doi: 10.1371/journal.pone.0002230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Freeman EE, Orroth KK, White RG, et al. Proportion of new HIV infections attributable to herpes simplex 2 increases over time: simulations of the changing role of sexually transmitted infections in sub-Saharan African HIV epidemics. Sex Transm Infect. 2007;83 (Suppl 1):i17–i24. doi: 10.1136/sti.2006.023549. [DOI] [PubMed] [Google Scholar]
  • 69.Grosskurth H, Gray R, Hayes R, et al. Control of sexually transmitted diseases for HIV-1 prevention: understanding the implications of the Mwanza and Rakai trials. Lancet. 2000;355:1981–1987. doi: 10.1016/S0140-6736(00)02336-9. [DOI] [PubMed] [Google Scholar]
  • 70.White RG, Orroth KK, Glynn JR, et al. Treating curable sexually transmitted infections to prevent HIV in Africa: still an effective control strategy? J Acquir Immune Defic Syndr. 2008;47:346–353. doi: 10.1097/QAI.0b013e318160d56a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.White RG, Orroth KK, Korenromp EL, et al. Can population differences explain the contrasting results of the Mwanza, Rakai, and Masaka HIV/sexually transmitted disease intervention trials? A modeling study. J Acquir Immune Defic Syndr. 2004;37:1500–1513. doi: 10.1097/01.qai.0000127062.94627.31. [DOI] [PubMed] [Google Scholar]
  • 72.Underhill K, Montgomery P, Operario D. Sexual abstinence only programmes to prevent HIV infection in high income countries: systematic review. BMJ. 2007;335:248. doi: 10.1136/bmj.39245.446586.BE. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Underhill K, Montgomery P, Operario D. Abstinence-plus programs for HIV infection prevention in high-income countries. Cochrane Database Syst Rev. 2008:CD007006. doi: 10.1002/14651858.CD007006. [DOI] [PubMed] [Google Scholar]
  • 74.Underhill K, Operario D, Montgomery P. Abstinence-only programs for HIV infection prevention in high-income countries. Cochrane Database Syst Rev. 2007:CD005421. doi: 10.1002/14651858.CD005421.pub2. [DOI] [PubMed] [Google Scholar]
  • 75.Pronyk PM, Kim JC, Abramsky T, et al. A combined microfinance and training intervention can reduce HIV risk behaviour in young female participants. AIDS. 2008;22:1659–1665. doi: 10.1097/QAD.0b013e328307a040. [DOI] [PubMed] [Google Scholar]
  • 76.Gray RH, Li X, Kigozi G, et al. The impact of male circumcision on HIV incidence and cost per infection prevented: a stochastic simulation model from Rakai, Uganda. AIDS. 2007;21:845–850. doi: 10.1097/QAD.0b013e3280187544. [DOI] [PubMed] [Google Scholar]
  • 77.Gray RH, Wawer MJ, Polis CB, et al. Male circumcision and prevention of HIV and sexually transmitted infections. Curr Infect Dis Rep. 2008;10:121–127. doi: 10.1007/s11908-008-0022-y. [DOI] [PubMed] [Google Scholar]
  • 78.Kigozi G, Gray RH, Wawer MJ, et al. The safety of adult male circumcision in HIV-infected and uninfected men in Rakai, Uganda. PLoS Med. 2008;5:e116. doi: 10.1371/journal.pmed.0050116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Krieger JN, Bailey RC, Opeya J, et al. Adult male circumcision: results of a standardized procedure in Kisumu District, Kenya. BJU Int. 2005;96:1109–1113. doi: 10.1111/j.1464-410X.2005.05810.x. [DOI] [PubMed] [Google Scholar]
  • 80.Shaffer DN, Bautista CT, Sateren WB, et al. The protective effect of circumcision on HIV incidence in rural low-risk men circumcised predominantly by traditional circumcisers in Kenya: two-year follow-up of the Kericho HIV Cohort Study. J Acquir Immune Defic Syndr. 2007;45:371–379. doi: 10.1097/QAI.0b013e318095a3da. [DOI] [PubMed] [Google Scholar]
  • 81.Siegfried N, Muller M, Volmink J, Deeks J, Egger M, Low N, Weiss H, Walker S, Williamson P. Male circumcision for prevention of heterosexual acquisition of HIV in men. Cochrane Database Syst Rev. 2003:CD003362. doi: 10.1002/14651858.CD003362. [DOI] [PubMed] [Google Scholar]
  • 82•.Lagakos SW, Gable AR. Challenges to HIV prevention: seeking effective measures in the absence of a vaccine. N Engl J Med. 2008;358:1543–1545. doi: 10.1056/NEJMp0802028. A helpful review of key prevention trials challenges, emphasizing PrEP and microbicide trials. [DOI] [PubMed] [Google Scholar]
  • 83.Lagakos SW, Gable AR. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, D.C: National Academies Press; 2008. [Google Scholar]
  • 84.Corey L, McElrath MJ, Kublin JG. Poststep modifications for research on HIV vaccines. AIDS. 2009;23:3–8. doi: 10.1097/QAD.0b013e32830e6d6d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.De Cock KM, Gilks CF, Lo YR, Guerma T. Can antiretroviral therapy eliminate HIV transmission? Lancet. 2009;373:7–9. doi: 10.1016/S0140-6736(08)61732-8. [DOI] [PubMed] [Google Scholar]
  • 86.Garnett GP, Baggaley RF. Treating our way out of the HIV pandemic: could we, would we, should we? Lancet. 2009;373:9–11. doi: 10.1016/S0140-6736(08)61698-0. [DOI] [PubMed] [Google Scholar]
  • 87•.Granich RM, Gilks CF, Dye C, et al. Universal voluntary HIV testing with immediate antiretroviral therapy as a strategy for elimination of HIV transmission: a mathematical model. Lancet. 2009;373:48–57. doi: 10.1016/S0140-6736(08)61697-9. This fanciful model examined the effectiveness of both increased HIV testing and early initiation of ART. Although the model assumptions are unrealistic, it is a stimulating examination of the potential for ‘test and treat’ initiatives as a HIV prevention strategy. [DOI] [PubMed] [Google Scholar]

RESOURCES