Next Article in Journal
Characterization of the Virome in Mosquitoes Across Distinct Habitats in the Yucatán Peninsula, Mexico
Previous Article in Journal
Metagenomic Next-Generation Sequencing for the Diagnosis of Infectious Uveitis: A Comprehensive Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biomedical Interventions for HIV Prevention and Control: Beyond Vaccination

1
School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
2
Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus, Sun Yat-sen University, Shenzhen 518107, China
3
Key Laboratory of Public Health Safety of the Ministry of Education, School of Public Health, Fudan University, Shanghai 200032, China
4
Key Laboratory of Tropical Disease Control, Sun Yat-sen University, Ministry of Education, Guangzhou 514400, China
5
State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
*
Authors to whom correspondence should be addressed.
Viruses 2025, 17(6), 756; https://doi.org/10.3390/v17060756
Submission received: 20 March 2025 / Revised: 15 May 2025 / Accepted: 22 May 2025 / Published: 26 May 2025
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)

Abstract

:
The global HIV epidemic remains persistent, mostly because of neither a drug for its cure nor a vaccine for its prevention. An HIV vaccine is thought of as the most cost-effective biomedical intervention to eventually terminate the HIV epidemic, but it is not clinically available yet due to technical hurdles. However, beyond vaccination, increasing alternative and innovative biomedical interventions have been developed for the prevention and control of HIV infections. Herein, we discuss the challenges encountered in the innovation of biomedical interventions against HIV infections, and summarize the landscape and latest advances of these biomedical interventions to intercept the risk of HIV acquisition and transmission, aiming to provide valuable clues for exploring more out-of-the-box solutions to prevent HIV infections, thereby contributing to realizing the 2030 goal of ending the AIDS epidemic.

Graphical Abstract

1. Introduction

More than 40 years have passed since the discovery of the human immunodeficiency virus (HIV) and acquired immunodeficiency syndrome (AIDS), yet neither a drug for its cure nor a vaccine for its prevention are available [1,2]. Despite advances in behavioral interventions, such as needle exchange programs, condom promotion, and partner education, the HIV epidemic remains a persistent global health challenge. As reported by the Joint United Nations Programme on HIV/AIDS (UNAIDS), there are currently 39.9 million people living with HIV (PLWH), and more than 40 million have died from AIDS-related complications [3]. In 2023, 1.3 million new infections were reported, underscoring the urgent need for more effective strategies [3]. With continuous advancements in HIV testing technologies and the increased availability of antiretroviral therapy (ART), 86% of people living with HIV (PLWH) were aware of their HIV status, and 30.7 million individuals (77% of PLWH) were receiving ART by the end of 2023.
However, the UNAIDS “90-90-90” global targets, proposed in 2014, have not yet been accomplished. These targets aimed to ensure the following by 2020: (1) 90% of PLWHs know their status, (2) 90% of diagnosed PLWHs receive sustained ART, and (3) 90% of PLWHs on ART treatment achieve viral suppression. Subsequently, the “95-95-95” targets for 2025—which demand 95% success rates for diagnosis, treatment initiation, and viral suppression—now also face significant challenges. The promotion of ART coverage—combined with biomedical interventions like pre-exposure prophylaxis (PrEP) and post-exposure prophylaxis (PEP)—can effectively reduce HIV transmission [4]. Due to the persistence of latent viral reservoirs in host cells, despite long-term treatment [5], and a rapid viral rebound after ART interruption [6], the current ART drugs alone cannot eliminate HIV infections, but ART treatment can render an epidemiologically irrelevant transmission risk under sustained virological suppression based on the Undetectable = Untransmittable (U=U) principle. Nevertheless, long-term use of ART drugs is associated with adverse events, such as cardiovascular disease and neurocognitive disorders [7,8,9]. Thus, ART therapy, rather offering an HIV cure, has transformed AIDS into a kind of chronic disease, which prompts new medical and social challenges [10,11]. In summary, the development of innovative biomedical interventions will be crucial for achieving the 2030 goal of ending the AIDS epidemic. While an effective HIV-preventive vaccine remains elusive due to technical hurdles, research efforts have increasingly shifted toward exploring alternative strategies. Herein, we provide an overview of the current landscape of HIV biomedical interventions beyond the HIV vaccine, highlighting their potential to mitigate the HIV epidemic (Figure 1). We aim to provide insights on exploring out-of-the-box solutions against HIV infections, and thus contribute to efforts for ending the AIDS epidemic.

2. Challenges for the Development of Biomedical Interventions Against HIV Acquisition and Transmission

To establish a foundation for evidence-based HIV biomedical interventions, it is necessary to have a comprehensive understanding of the difficulties and challenges in developing new strategies against HIV acquisition and transmission. Therefore, we have summarized these challenges encountered in the innovation of biomedical interventions against HIV infections (Figure 2).

2.1. High Genetic Variability

The extensive genetic variation in the HIV genome, driven by frequent mutations and recombination, represents a significant challenge in controlling the global HIV pandemic. As a retrovirus with a single-stranded RNA genome, HIV exhibits a mutation rate of 5 to 10 bases per replication cycle, attributable to the absence of proofreading by reverse transcriptase [12]. Additional errors introduced during transcription by RNA polymerase II [13] further contribute to the genetic diversity of HIV, particularly in the envelope (env) gene. This gene encodes proteins that are essential for viral entry into host cells. The envelope protein, a primary target of neutralizing antibodies, demonstrates substantial variability, which poses a significant obstacle to the development of broadly effective vaccines and antibodies [14]. Additionally, HIV undergoes frequent recombination, which significantly complicates efforts to prevent the transmission of this virus. HIV possesses two single-stranded, positive-sense RNA genomes. However, these two RNA strands are not identical copies and do not undergo conventional meiosis or homologous pairing during genetic recombination. Consequently, HIV cannot be classified as a true diploid, but is instead termed a “pseudodiploid” virus. The pseudodiploid nature of HIV arises may through the following two key mechanisms: (1) cellular co-infection by multiple viral particles, and (2) packaging of heterologous RNA genomes into individual virions [15,16]. This dual-RNA architecture enables a single infected cell to harbor multiple genomic templates. During reverse transcription, the viral polymerase undergoes frequent template switching between these RNA strands, generating recombinant proviral DNA with enhanced genetic diversity. This molecular recombination serves as a major driver of HIV’s evolutionary adaptability, facilitating immune evasion and drug resistance development [17]. During reverse transcription, reverse transcriptase can switch selectively between the two RNA template strands, producing a chimeric DNA molecule and resulting in genetic recombination, with an average frequency of up to five recombinant events per proviral genome synthesis [18].
The high recombination rates in HIV genes have contributed to the emergence of diverse subtypes, recombinant forms, and unique strains, exacerbating the global pandemic [19,20]. HIV can be divided into HIV-1 and HIV-2. HIV-1 and HIV-2 originate from different types of SIV in different monkey species. HIV-1 is closely related to SIV in chimpanzees (SIVcpz) [21], whereas HIV-2 is more closely related to SIV in white-browed monkeys (SIVsm) [22]. Compared to HIV-1, HIV-2 exhibits a significantly lower mutation rate, reduced pathogenicity, and diminished transmissibility. As a result, HIV-2 infections represent only a minor fraction of global HIV cases. Geographically, HIV-2 remains largely restricted to West Africa, likely due to a combination of epidemiological factors and limited spread beyond the region (if not specified, HIV in the following text refers to HIV-1). HIV-1 has evolved into four major groups, which are as follows: M, O, N, and P, with Group M further subdivided into eleven subtypes [23]. These four groups emerged from four independent zoonotic transmissions, with group M being the predominant and globally prevalent form of HIV-1 [24]. Amino acid differences within subtypes can range from 8 to 30%, and between subtypes, from 17 to 42%, greatly complicating the development of vaccines and other biomedical interventions [25].
Importantly, mutation and recombination in HIV are not independent, but interact synergistically. Some studies have shown that mutation rates are significantly higher in recombinant regions, with approximately 15–20% of mutations associated with genetic recombination [26]. These genetic variations complicate the design of interventions, necessitating broad-spectrum strategies to address the diversity of circulating strains. Under selective pressures from host immune responses and antiretroviral drugs, HIV evolves rapidly, making it highly adaptable to the host, and thus leading to significant challenges for prevention and control. The genetic diversity of HIV arises not only from the error-prone nature of reverse transcriptase and frequent recombination events, but also through the action of host-encoded mutagenic enzymes. The APOBEC3 family, particularly APOBEC3G and APOBEC3F, plays a prominent role in this process. When HIV fails to express functional Vif protein, these cytidine deaminases are packaged into virions and subsequently induce G-to-A hypermutations by deaminating cytosines in the minus-strand cDNA during reverse transcription [27]. While extensive APOBEC-mediated hypermutation generally proves to be lethal to the virus, sublethal levels of editing may allow viral variants to evade immune detection and become established in the population, thereby driving viral evolution and contributing to drug resistance [28]. Additional host factors, including ADARs (adenosine deaminases acting on RNA), may further modify HIV RNA through editing, although their exact role in shaping viral diversity requires further elucidation [29]. Consequently, biomedical interventions must account for the complexity of subtype variability, necessitating broad-spectrum strategies that encompass multiple subtypes.

2.2. Highly Glycosylated Antigens

Glycan arrays that densely surround the surface of the envelope antigen represent another distinctive feature of HIV. Glycosylation, a common post-translational modification of proteins, is closely linked to the proper folding and conformational changes of viral proteins [30]. Glycosylation can influence viral infectivity by facilitating viral attachment, cell fusion, and migration, and also plays a role in viral replication, including assembly and budding [31]. HIV gp120 glycoprotein has a highly glycosylated surface and is among the most heavily glycosylated non-synthetic proteins. A single envelope protein trimer contains more than 90 N-linked glycans, which comprise over half of its mass [32]. Compared to the binding of antibodies to protein antigens, the affinity of antibodies for carbohydrate-based antigens is relatively low [33], which impedes cross-linking and receptor-binding of B-cell receptors (BCRs). Regarding the source of glycans, HIV does not encode the gene product responsible for glycan synthesis. Instead, HIV utilizes host cell’s cellular enzymes and raw materials to glycosylate the surface of viral antigens. The antigen surface, covered by the host’s “self” glycans, enables HIV to develop immune tolerance to the functional antibodies and evade antibody recognition [34]. Furthermore, due to the homology between HIV glycans and host glycans, antibodies generated against the viral N-linked glycans may cross-react with the host’s N-linked glycans. Structurally, the highly glycosylated structure of the HIV antigen confers unique properties that significantly complicate HIV prevention. On one hand, this highly glycosylated structure can harbor key conserved antigenic epitopes of HIV, reducing the overall antigenicity of gp120 and making it exceptionally challenging for the immune system to generate the corresponding broadly neutralizing antibodies [35]. On the other hand, the conformational plasticity of glycosylated antigens enables them to undergo “conformational masking”, allowing them to escape recognition by neutralizing antibodies and thus protecting their vulnerable regions from immune attack [36]. For instance, although the receptor-binding region of gp120 lacks glycosylated modifications, the glycan located near the CD4 receptor-binding site creates a masking effect that renders it resistant to antibodies targeting this epitope, thereby evading antibody-mediated neutralization [37].

2.3. Latent Viral Reservoirs

After successfully invading host cells, HIV can synthesize viral DNA through the action of reverse transcriptase and integrate it into the host genome via the enzyme integrase. During this process, HIV can persist for extended periods in the form of pre-integrated viral DNA, particularly in long-lived cells, such as resting CD4+ memory T cells, forming what is known as a latent viral reservoir [38,39]. Conventional ART drugs are effective in targeting and eliminating free virions undergoing active replication. However, they have minimal impact on latent HIV reservoirs in a quiescent state. Even in patients receiving long-term ART with undetectable plasma viral loads, latent viral reservoirs persist [5]. While conventional ART effectively suppresses the plasma viral load to undetectable levels and prevents productive virions from productively infecting cells, viral rebound can occur rapidly following antigenic activation or treatment interruption [40]. Even intensification with ART has not been successful in eradicating the viral reservoir and preventing antiretroviral-free viral rebound [41]. Thus, eradicating the latent HIV reservoir has become a key scientific challenge in the development of vaccines and curative treatments for HIV [42,43,44]. Current strategies aimed at eliminating the latent HIV reservoir include several innovative approaches, which are as follows: (1) Early initiation of ART, in which HIV reservoirs can form and expand rapidly within days of infection. By promptly identifying newly infected individuals and initiating ART as early as possible, the initial establishment and expansion of viral reservoirs can be minimized [45]. (2) The “Shock and Kill” strategy, which is an approach that aims to reactivate latent HIV-infected cells, thereby allowing the expression of viral proteins, which can then be targeted and eliminated by the immune system [46]. Some clinical trials, such as the RIVER study, have explored the feasibility of this strategy, showing promising results in clearing latent reservoirs [47]. A recent study demonstrated that its HSV-vectored therapeutic vaccine was able to reduce the size of latent reservoirs [48]. (3) The “Block and Lock” strategy, which is a strategy that focuses on preventing the reactivation of latent HIV by locking it in an inactive state through the use of specific compounds that inhibit the transcriptional machinery necessary for HIV activation [49]. In summary, while significant progress has been made in controlling active HIV infection with ART, the challenge of eliminating latent reservoirs remains a critical barrier to a complete cure. Innovative strategies targeting these reservoirs are essential to achieving a functional or sterilizing cure for HIV. In addition, while viral persistence mechanisms primarily inform HIV cure research, understanding latency reservoirs is critical for optimizing PrEP durability in seronegative high-risk populations [50].

2.4. Lack of Animal Infection Models

Animal models can replicate and mimic the process of viral infection in vivo, providing preliminary evidence for the safety and efficacy of drugs and vaccines, and thus are indispensable for elucidating the mechanisms of pathogenesis, and for the development of therapeutics and vaccines [51,52]. However, due to HIV’s strict host specificity, it can only cause obvious AIDS clinical symptoms in humans. While some primates can be infected with HIV, they do not exhibit typical clinical symptoms. Consequently, there is a lack of appropriate animal models for HIV-related studies. Currently, simian immunodeficiency virus (SIV) or chimeric simian/human immunodeficiency virus (SHIV) infections in non-human primates, such as rhesus macaques, are used to model HIV infections in humans [52,53]. Despite the similarities between SIV and HIV, there are significant genetic and biological differences between the two viruses [54], as well as differences in the immune systems of non-human primates and humans. As a result, the immune responses observed in these models may not fully replicate those observed in HIV patients, complicating the extrapolation of findings from animal studies to human applications in pathogenesis, drug development, and vaccine design. In addition, there are significant differences in the course of disease, as well as pathological changes and clinical manifestations between SIV and HIV, making it insufficient to rely on a single animal model for studying HIV infections. Moreover, the cost and ethical considerations associated with the use of non-human primates are also important factors that must be taken into account. Scientists are actively seeking inexpensive and easily accessible small animal models for HIV research. In recent years, the BLT (bone marrow, liver, and thymus) humanized mouse model has been used to partially replicate HIV infection in vivo, facilitating the study of pathogenesis, drug screening, and gene therapy [55,56,57,58,59]. However, due to the development of graft-versus-host disease (GvHD) and its inability to fully replicate the human immune system [60], there remains a need to develop alternative animal models that are more suitable and effective in the development of biomedical interventions against HIV infections.

2.5. Lack of Correlates of Protection

Once infected with HIV, no one has been found to spontaneously eradicate HIV and completely recover to the pre-infection state, suggesting that the human immune system alone is unable to provide effective immune protection to cure HIV infections. The correlates of protection are potential parameters to assess the protective efficacy conferred by biomedical interventions against HIV, and thus they are critical to determining whether the intervention is effective against HIV infections. Unfortunately, we still lack a comprehensive understanding of which immune components and antigenic targets are pivotal in controlling HIV infection [61,62,63]. This knowledge gap has hindered progress in the development of immunoassays to accurately predict the protective effects of interventions in humans, as well as in innovating immunological tools capable of inducing durable protective immune responses. Due to the high variability of HIV and the complexity of the infection process, it remains challenging to evaluate the protective efficacy of interventions using a single biomarker, further complicating the assessment of preventive effectiveness. Therefore, researchers are exploring a wide range of potential immunoprotective indicators, including the breadth and magnitude of antibody responses (both neutralizing and non-neutralizing), T-cell activity, and cytokine profiles [64,65,66,67,68,69]. For example, the results from an animal experiment indicate that CXCR5+ CD8 T cells expand rapidly during chronic SIV infection and play a key role in controlling SIV progression, demonstrating their potential as indicators of immune protection [70]. Additionally, cyclic GMP-AMPase has been identified as an innate immune sensor for HIV, which is rapidly activated upon HIV infection and induces the production of type-I interferons and a variety of cytokines, thereby exerting a protective effect against HIV [71]. Despite ongoing efforts, no universal and reliable indicators have yet been established to fully measure protection efficacy against HIV. A more comprehensive understanding of the immune system’s response to HIV is urgently needed to identify and validate more effective and reliable immunoprotective indicators. Such insights could significantly improve our ability to assess the immunoprotective effects of various interventions, thereby guiding the development of more effective HIV prevention and treatment strategies.

2.6. Ethical and Biosafety Issues

Clinical trials are an essential step in the development of HIV biomedical interventions. However, because HIV is highly pathogenic and practically incurable, conducting clinical trials for biomedical interventions, particularly in live HIV infection research, carries unavoidable risks. For example, during the studies of an HIV vaccine, researchers found that although vaccination with an attenuated live SIV mutant vaccine can provide effective protection against wild SIV infection, this strategy is still prohibited from further research in the human clinical trials due to potential virus recovery mutations [72]. Studies involving human HIV cohorts have also shown that even highly attenuated strains of HIV can still lead to AIDS-associated symptoms [73]. These findings highlight safety concerns during the development of biomedical interventions against HIV. Significant gaps persist in assessing the safety and efficacy of HIV prevention products for special populations, particularly pregnant and lactating women. These gaps primarily stem from ethical concerns regarding potential risks to infants, which often preclude their inclusion in clinical trials [74]. While randomized, double-blind, placebo-controlled trials (RCTs) remain the gold standard for minimizing bias and ensuring robust results, their application in HIV prevention research requires careful ethical scrutiny. Traditional placebo-controlled designs—where control groups receive no active prophylaxis—are now considered ethically unacceptable in this field [75]. This shift reflects the widespread availability of highly effective antiretroviral-based prevention (e.g., PrEP), which establishes a clear ethical obligation to provide proven protection to all trial participants [76,77]. Consequently, contemporary HIV prevention trials must balance scientific rigor with ethical imperatives, often adopting alternative designs (e.g., active-controlled or superiority trials), which might hinder the development progress to some extent.

3. Advances of Biomedical Interventions for HIV Prevention and Control

HIV prevention and control is the most key component in achieving the goal of eliminating AIDS. Biomedical interventions are crucial for HIV prevention and control, and we herein summarize current biomedical interventions for HIV prevention and control, aiming to provide a reference for the development of innovative strategies against HIV infections (Table 1).

3.1. PrEP

The concept of PrEP was introduced in the mid-2000s as a strategy to reduce HIV transmission in individuals at high risk who are HIV-negative. This approach involves the regular use of antiretroviral drugs to lower the risk of HIV acquisition. The initial PrEP drugs, primarily oral nucleoside reverse transcriptase inhibitors (NRTIs) like Tenofovir/Emtricitabine (TDF/FTC) and Tenofovir Alafenamide/Emtricitabine (TAF/FTC), have demonstrated good safety and efficacy in numerous studies [79]. In 2012, the World Health Organization (WHO) endorsed daily oral TDF/FTC as a PrEP measure for HIV-negative individuals in serodiscordant relationships, marking a significant milestone in HIV prevention. In 2015, the WHO expanded its PrEP recommendations to include men who have sex with men (MSM) and people who inject drugs, further broadening the scope of HIV prevention efforts [93]. Despite these advances, adherence challenges with daily oral PrEP, as observed in trials such as VOICE and KPNC, underscored the need for more user-friendly options [94,95]. Additionally, the short duration of protection and the need for frequent medication can lead to side effects, such as gastrointestinal irritation and nephrotoxicity [96,97]. Consequently, researchers are intensifying their efforts to develop long-acting antiretroviral drugs and alternative routes of drug delivery, beyond daily oral intake, to sustainably inhibit viral replication, prevent the emergence of drug-resistant strains of the virus, and address issues of patient privacy and social stigma. For example, cabotegravir, an integrase strand transfer inhibitor given as an injection every two months, showed higher efficacy than daily TDF/FTC in trials (HPTN 083 and HPTN 084) involving MSM and transgender women [84]. This led the WHO, in 2022, to recommend cabotegravir as a long-acting PrEP option for individuals at a high risk of HIV infection. Nevertheless, two critical considerations persist with long-acting cabotegravir (CAB-LA) implementation, as follows: (1) the emergence of LA-CAB resistance mutations in breakthrough infections [98], and (2) the phenomenon of prolonged viral suppression with subsequent immunological discordance, which is termed long-acting early viral inhibition (LEVI) syndrome, following undiagnosed acute HIV infection at drug initiation [99]. Islatravir (ISL), a new class of nucleoside reverse transcriptase translocation inhibitor, is undergoing evaluation in the Impower 022 and Impower 024 trials to assess its effectiveness across different populations. Of note, lenacapavir, a long-acting capsid inhibitor, demonstrated a strong preventive effect against HIV transmission. A single subcutaneous injection of lenacapavir provided drug exposure for up to six months, leading to its approval for HIV prevention in the European Union in 2022 [100]. Importantly, impressive findings from the Phase III clinical trial (PURPOSE 1) recently revealed that administering lenacapavir subcutaneously every six months resulted in almost 100% efficacy in preventing HIV infections [85], effectively improving the PrEP dosing regimen and the single route of delivery, which is of great significance in meeting the goals of ending AIDS by 2030 [101]. However, it should be noted that the global distribution of newer antiretroviral agents like lenacapavir may face significant limitations in the near future. These constraints primarily stem from manufacturing capacity limitations, which contribute to both supply shortages and prohibitively high costs, thereby restricting accessibility in many regions. Here, we summarize those key Phase III PrEP clinical trials (Table 2).
The tissue distribution profiles of PrEP agents exhibit significant site-specific variations across different mucosal surfaces. Pharmacokinetic studies demonstrate that tenofovir and emtricitabine achieve substantially higher drug concentrations in colorectal tissues compared to vaginal mucosa [105]. This differential distribution results in suboptimal drug exposure at certain anatomical sites, particularly in vaginal tissue where the drug concentration may fail to reach the threshold required for effective prophylaxis. Consequently, the preventive efficacy of PrEP shows marked anatomical variation depending on the exposure site. Alternatively, the dapivirine ring represents a new PrEP-based approach that offers a long-term HIV prevention option for women who are unwilling or unable to take daily oral medication. Unlike oral PrEP, it does not require daily or pericoital use. The dapivirine ring is a silicone ring inserted into the vagina, which provides a sustained, localized release of dapivirine (a non-nucleoside reverse transcriptase inhibitor) over a 28-day period, offering women a controlled, long-lasting, and discreet form of HIV prevention. Several studies have demonstrated that the dapivirine ring has a favorable level of safety and is effective in decreasing the risk of HIV infection, and can have improved efficacy with improved adherence [82,106]. In 2021, WHO endorsed the dapivirine ring as an additional HIV prevention method for women at high risk [107]. However, the dapivirine ring still has several limitations, including the following: (a) it is only suitable for vaginal intercourse and cannot protect against other routes of HIV transmission; (b) its overall protection effect ranges from 27% to 63%, which may not provide sufficient protection; and (c) localized vaginal irritation may reduce adherence to its use. Additionally, research on the rectal dapivirine ring may further expand the scope and application of PrEP.
Long-lasting biomedical delivery devices, whether administered locally or systemically, are also being explored for PrEP, as they can improve the invisibility, effectiveness, and compliance of biomedical interventions [108]. Long-acting antiretroviral implants represent a promising and highly effective HIV intervention technology, providing sustained release of antiretroviral drugs for months or even years after being implanted under the skin. These implants offer a convenient, long-term solution for preventing HIV infection by suppressing viral replication. Several research teams are currently engaged in the development of long-acting antiretroviral implants and investigating their feasibility and safety [108,109,110,111,112]. Long-acting antiretroviral implants achieve sustained drug release through various modalities, such as the following: (1) refillable nanochannel implants that maintain therapeutic tenofovir alafenamide (TAF) concentrations for 83 days; [109] (2) biodegradable or non-biodegradable polymer matrices that provide islatravir (ISL)-based protection for six months [113]; and (3) poly lactic-co-glycolic acid (PLGA) bioerodible matrices that sustain effective dolutegravir (DTG) levels beyond five months [114]. However, challenges remain in stabilizing the drug release rate and mitigating potential adverse effects associated with the implants, such as inflammation [115,116]. Long-acting antiretroviral implants with controlled adverse effects and stably releasing drug concentrations up to the potent dose could contribute greatly to HIV prevention if they are successfully put into clinical use.

3.2. PEP

After HIV exposure via vaginal mucosa, the detection of HIV in regional lymph nodes can be achieved in about 2 days, and it takes 5 days for HIV to be detected in the blood, suggesting that there is a window of opportunity after HIV exposure [117]. PEP is the intervention aimed at blocking HIV infection during this window phase by administering antiretroviral drugs for 28 consecutive days, ideally starting within 24 h, and no later than 72 h after exposure. PEP provides an emergency, remedial HIV prevention option for individuals who have been exposed to HIV through an occupational or nonoccupational event. The evaluation of PEP efficacy presents unique methodological challenges due to its inherently episodic nature as a single-course intervention. The absence of RCTs assessing PEP effectiveness for HIV prevention reflects these difficulties, as the transient and unpredictable nature of exposure events complicates the design of robust clinical studies. Nevertheless, extensive preclinical evidence from animal models has consistently demonstrated the efficacy of PEP in reducing the HIV transmission risk [118,119]. These experimental findings are further corroborated by population-level studies documenting PEP’s effectiveness in real-world settings [87,120,121]. The convergence of these complementary lines of evidence has led to the widespread adoption of PEP in clinical practice, with regulatory approval granted in most national HIV prevention guidelines globally. Coformulation of zidovudine (ZDV) and lamivudine (3TC) was first recommended for occupational PEP [122], and later for nonoccupational PEP [123]. Contemporary guidelines generally recommend replacing zidovudine with tenofovir and using DTG as the preferred third drug for PEP in NRTIs-based dual or triple combination regimens [124,125,126]. As a remedial prophylactic measure, PEP does not provide 100% protection, and thus should not be relied upon as the sole or primary HIV prevention strategy. Reported failed reasons after PEP include delayed initiation, poor or non-adherence, and continued high-risk sexual exposures after PEP administration. It is worth noting that studies have shown that the convenience of PEP may lead at-risk populations to over-rely on it, substituting it for other preventive measures, which increases the risk of HIV transmission [127]. Additionally, the timing of PEP initiation and adherence are critical factors influencing its effectiveness [128]. The main risks associated with PEP include adverse reactions to antiretroviral (ARV) drugs and the potential emergence of drug-resistant strains of HIV. Consequently, several trials have investigated the adverse effects and tolerability of PEP (Table 3). The WHO guidelines advise that recurrent PEP users or individuals with ongoing HIV exposure risks should transition to PrEP for more effective, continuous prevention. Despite the risks of failed protection and the occurrence of adverse events, the use of PEP continues to grow, as it provides a critical emergency intervention following potential HIV exposure.

3.3. Treatment as Prevention (TasP) and U=U Principle

The WHO’s 2023 HIV Control Guidelines points out “Three-tiered framework of HIV control encompassing (1) population-level incidence reduction, (2) suppression of disease progression in seropositive individuals, and (3) functional cure/eradication research”. Through promoting current ART treatment and future functional cure/eradication treatment, AIDS patients (the source of HIV transmission) will be effectively controlled and reduced, so as to reduce and prevent the population-level incidence of HIV infections. TasP is a biomedical prevention strategy to decrease the risk of HIV transmission by controlling the viral load in PLWH through ART. U=U, a core scientific concept in the TasP strategy, explicitly states that as long as an HIV-infected individual has an undetectable viral load, the risk of sexual transmission of HIV is considered to be zero [143]. Numerous studies have shown that the risk of HIV transmission is directly correlated with the viral load in PLWH, and the possibility of HIV transmission can be substantially reduced when the viral load is lowered to very low levels through ART [144,145]. When the viral load reaches undetectable levels, PLWHs no longer exhibit AIDS-related symptoms and are considered non-infectious [143,146]. In fact, there is almost zero risk of sexual transmission of HIV when there are viral loads less than 1000 copies/mL [88]. The current findings indicate that U=U applies at an HIV RNA threshold of 200 copies/mL [147]. Based on the TasP strategy, the 2015 universal ART guidelines recommended that ART be initiated immediately after HIV diagnosis to facilitate viral suppression [148]. The TasP strategy, along with the U=U concept, was formally endorsed by the WHO in 2018, which emphasized that maintaining an undetectable viral load through adherence to ART is key to achieving both TasP and U=U.
In addition to ART for achieving U=U, recent research has explored innovative biological interventions to combat HIV. Among these, therapeutic interfering particles (TIPs)—defective viral particles that replicate conditionally in the presence of wild-type HIV—have emerged as a promising strategy [149]. TIPs act as “parasites”, depleting the resources necessary for wild-type HIV to replicate and proliferate. This competitive interference can continuously reduce the viral load of HIV and exert antiviral effects. Recently, a study has shown that single-dose HIV treatment with TIPs has the potential to limit the viral load and reduce HIV transmission by lowering the viral load to below the transmission threshold defined by the WHO [150], and thus meeting the U=U for HIV prevention. The development and widespread acceptance of these concepts have not only advanced our understanding of HIV prevention, but have also contributed significantly to reducing the stigma faced by PLWH, thereby slowing the global HIV epidemic.

3.4. Testing as Prevention

HIV testing represents a cornerstone of HIV prevention and control strategies, frequently discussed in conjunction with TasP. Despite progress, approximately 14% of people living with HIV remain unaware of their infection status, with many recognizing their infection only at advanced stages. Research indicates that delayed ART initiation due to late diagnosis significantly contributes to increased HIV transmission risk. The use of HIV rapid test kits enables early detection, helping to identify undiagnosed infections. By facilitating prompt ART initiation, these tests play a crucial role in achieving viral suppression, thereby reducing transmission rates. Modeling studies demonstrate that when HIV self-testing kits are introduced as a supplementary testing method among MSM, they could reduce HIV prevalence by 10%, while substantially improving awareness of serostatus within this population [151]. Furthermore, organized large-scale HIV testing initiatives (e.g., crowdsourced testing) in MSM communities have proven to be effective in lowering HIV incidence [152]. These findings underscore the indispensable role of HIV testing in prevention efforts, even though the concept of “testing as prevention” has yet to gain widespread recognition.

3.5. Prevention of Vertical Transmission

Prevention of vertical transmission is a strategy designed to prevent HIV transmission from mother to child, which is the primary route of HIV infection in children. The approach involves a range of intervention measures aimed at interrupting this transmission. Statistics indicate that, in the absence of preventive interventions, the cumulative rate of vertical transmission of HIV during pregnancy, delivery, and breastfeeding can be as high as 45% [153]. This probability can be reduced to 2% after intervention with ART, and can be further reduced (<1%) with a combination of intervention measures [154]. Research has demonstrated that the mother’s HIV viral load is a critical determinant of vertical transmission risk. When the mother’s viral load is undetectable, the likelihood of transmission to the child is virtually eliminated, which is consistent with the concept of U=U. In addition, continued ART during breastfeeding can further reduce the risk of HIV transmission through breast milk [154]. The WHO recommends that HIV-positive mothers in resource-limited settings breastfeed while maintaining effective ART, as the nutritional and immunological benefits for the infant substantially outweigh the low residual transmission risk when the maternal viral load is suppressed. However, it must be emphasized that ART-suppressed breastfeeding still carries a non-zero transmission risk. In settings where safe alternatives are available (including affordable, sustainable formula feeding), replacement feeding remains the preferred option to completely eliminate postnatal HIV transmission. In cases where the mother’s viral load is not adequately controlled at the time of delivery, the cesarean section can still significantly reduce the risk of transmission [155]. Current clinical guidelines recommend intravenous zidovudine administration during intrapartum care for pregnant women with HIV viral loads exceeding 1000 copies/mL at delivery. Furthermore, the duration and intensity of infant antiretroviral prophylaxis should be stratified according to transmission risk, which is primarily determined by the degree of maternal virological control throughout pregnancy and viral load status at delivery. Through the promotion and popularization of interventions to prevent vertical transmission in recent years, the global rate of mother-to-child transmission of HIV has been successfully controlled [3].

3.6. Voluntary Medical Male Circumcision (VMMC)

Several randomized controlled trials examining the effect of circumcision on HIV risk in heterosexual men have demonstrated that circumcised men experience significantly lower rates of HIV infection (approximately 60%) compared to their uncircumcised counterparts [90,91,92]. However, circumcision does not reduce the level of HIV in the genital tract or the likelihood that people living with HIV transmit the virus to their sexual partner. In fact, possible mechanisms for the role of VMMC in reducing HIV infection in men who engage in penetrative sex include the following: (1) Reduction of HIV-susceptible tissue. Large numbers of HIV target cells (such as CD4+ T cells and Langerhans cells) are concentrated on the inner mucosal surface of the foreskin [156,157]. Voluntary medical male circumcision (VMMC) removes this tissue, thereby reducing the surface area vulnerable to HIV infection. (2) Alteration of genital microbial composition and inflammation. VMMC may protect against HIV by modifying the genital microbiome and reducing inflammation. The procedure exposes the glans to a drier environment, decreasing the abundance of anaerobic bacteria and associated inflammation [158,159]. This reduces the likelihood of HIV encountering target immune cells and establishing infection. (3) Reduction in co-infections that facilitate HIV transmission. VMMC can lower the incidence of sexually transmitted infections (STIs) linked to HIV acquisition, such as human papillomavirus (HPV) and herpes simplex virus type 2 (HSV-2) [160]. By mitigating these biological co-factors, circumcision indirectly reduces HIV susceptibility. Long-term follow-up data further support the efficacy of circumcision in consistently reducing the risk of HIV infection [161]. Therefore, UNAIDS recommends VMMC as an HIV prevention measure in areas with a high HIV prevalence rate [162]. VMMC is not currently recommended to prevent HIV transmission in MSM and transgender women. Nonetheless, recent studies showed that VMMC also play a role in reducing the risk of HIV infection among MSM [163,164], possibly because VMMC may be of some effect in exclusively insertive sexual intercourse. Consequently, VMMC has proven to be an effective strategy for HIV prevention in men. Furthermore, while VMMC offers HIV prophylaxis to a certain extent, it does not eliminate the risk of infection, and should therefore be used in conjunction with other preventive measures to maximize its effectiveness.

3.7. Gene Editing Technology

Achieving an HIV cure through the body’s own immune system remains extremely difficult; however, there have been a small number of cases worldwide where HIV patients have been successfully cured. In 2009, the first reported case of an HIV cure through stem cell transplantation occurred in the so-called “Berlin Patient,” a person with both acute myeloid leukemia and HIV-1 infection. This patient was cured following a stem cell transplant from a donor with a 32-nucleotide deletion in the CCR5 gene (CCR5 delta 32), which confers resistance to HIV infection [165]. Since then, additional cases have been reported, including the “London Patient”, “Dusseldorf Patient”, “New York Patient”, “City of Hope Patient”, “Geneva Patient”, and another “Berlin Patient”, all of whom were cured of HIV after receiving stem cell transplants [166,167,168,169]. Furthermore, investigations have shown that some Caucasian individuals with the CCR5 delta32 mutation gene remain uninfected and inherently immune to HIV despite extensive exposure to HIV [170]. These findings highlight the potential for genetic approaches in both HIV prevention and treatment. Unfortunately, due to the invasive nature of stem cell transplantation and the stringent conditions under which it is performed, this approach is not universally applicable, and is primarily reserved for patients with severe co-existing conditions, such as malignant tumor. Therefore, researchers are actively exploring gene editing technologies as a promising and accessible alternative for HIV treatment and prevention.
Gene editing technologies, such as CRISPR-Cas9 and Zinc Finger Nucleases, primarily function by modifying either the host cell or viral genome. These technologies have emerged as a promising avenue for exploring novel biomedical interventions for HIV prevention. For HIV-infected individuals, natural immunity to HIV can potentially be achieved by knocking out or modifying HIV co-receptors, such as CCR5 or CXCR4, in the host cell [171,172]. Results from a clinical trial have demonstrated that transplantation of hematopoietic stem and progenitor cells with CCR5 knockdown into patients with HIV-1 infection and acute lymphoblastic leukemia by CRISPR editing can achieve a long-term reconstitution of the patient’s hematopoietic system without the occurrence of gene editing-related side effects, and can produce partial control of HIV infection during ART cessation. However, the percentage of CCR5-deficient lymphocytes following treatment remains relatively low (approximately 5%), which results in limited efficacy in controlling HIV infection, underscoring the need for further research to refine this approach [173]. For the viral genome, gene editing offers the potential to directly target and excise integrated HIV DNA, including from latent viral reservoirs, thus preventing HIV proliferation, or even achieving a cure [174,175]. HIV-specific engineered recombinases (e.g., Brec1) represent a promising gene editing approach for the targeted excision of integrated proviral DNA from the host genome. The molecule’s precise recognition of HIV long terminal repeats (LTRs) substantially minimizes off-target effects. In vitro studies demonstrated that Brec1 excised >90% of integrated proviral DNA, with no detectable off-target activity. Furthermore, in vivo experiments involving transplantation of Brec1-modified hematopoietic stem/progenitor cells (HSPCs) into humanized mice resulted in significant viral load reduction, indicating a potential for sustained immunological control of HIV infection. Notably, viral vectors have also been used as tools for gene editing techniques. This is done by using viral vectors, such as retroviral vectors (mainly lentiviral vectors), which remove pathogenicity but retain the ability to deliver genes, to insert anti-HIV gene fragments into the host cell so that they can be expressed efficiently and consistently in the host cell, thereby preventing HIV infection [176,177]. However, the application of gene editing technology to humans faces enormous ethical challenges. Furthermore, gene editing technology must overcome challenges such as off-targeting effects, HIV’s high variability, and the development of efficient delivery systems [178,179]. There are also concerns about the potential mutations by gene editing, thus leading to tumorigenesis [180,181]. Therefore, additional clinical trials are required to assess the safety and reliability of gene editing as a viable HIV intervention.

3.8. Passive Infusion of Broadly Neutralizing Antibody

With longitudinal co-evolution between the HIV variants and the B-cell lineages, the broadly neutralizing antibody (bnAbs) can be generated in about 1% of HIV-infected patients after multiple years [182,183,184]. Increasing HIV-specific bnAbs have being isolated, including VRC01, B12, 3BNC60, 3BNC117, 2F5, 4E10, 2G12,PG9, PG16, and CH01, etc. Broadly neutralizing antibodies (bNAbs) can potently inhibit both cell-free and cell-to-cell HIV transmission, effectively reducing viral infection of target cells across diverse HIV isolates. The breadth and potency of this neutralization vary significantly, depending on the bNAb’s epitope specificity and structural characteristics. Passive infusion of bnAbs has achieved not only a therapeutic effect in HIV patients, but also a potential for HIV prevention in humans. Of note, passive infusion of bnAbs also showed a role in the prevention and control of mother-to-child HIV transmission [185,186]. However, monotherapy with a single broadly neutralizing antibody (bNAb) demonstrates limited clinical efficacy, as exemplified by the extensively studied VRC01 antibody. Clinical trial data revealed that intravenous VRC01 administration every eight weeks provided protection solely against VRC01-sensitive HIV strains, while failing to reduce the overall HIV-1 acquisition rates compared to placebo [187], or to prevent viral rebound after ART interruption [188]. Consequently, there is an urgent need to develop next-generation bNAbs with both improved breadth against diverse HIV strains and an extended serum half-life. A promising strategy involves engineering multispecific bNAbs, which incorporate multiple antigen-binding domains into a single antibody scaffold. Preclinical studies demonstrate that such bispecific or trispecific bNAb constructs exhibit significantly enhanced antiviral breadth and neutralization potency in animal models compared to single bNAbs [189,190]. Furthermore, administration of combined bnAbs can further improve the broad-spectrum, durability, and efficacy against different HIV mutants [191,192]. In addition, the passive infusion of bnAbs has been demonstrated with acceptable safety and tolerability in humans [193], and it is expected to play a vaccine-like role in preventing HIV infection in the future.
Vectored immunoprophylaxis is an emerging strategy for HIV prevention, and different vectors (such as AAV) are used to express bnAbs or other anti-HIV factors that provide sustained protection against HIV in the host [194,195,196], making them a promising biomedical intervention for HIV prevention.

4. Conclusions and Prospective

We are currently in the most critical decade for achieving the 2030 goal of ending AIDS epidemic. A prophylactic vaccine is thought of as the most cost-effective biomedical intervention to eventually terminate the HIV epidemic among the general population, but it seems to not be clinically available soon due to technical issues. Though there is still no available HIV vaccine approved for clinical use, increasing biomedical interventions beyond vaccination, including through the use of PrEP, PEP, TasP/U=U, PMTCT, Dapivirine, and VMMC, is emerging for the prevention of HIV infections. Moreover, progress has been made in broadly neutralizing monoclonal antibodies, long-acting antiretroviral implants, immunoprophylaxis, interfering viral particles, and gene editing. These interventions are promising to intercept the risk of HIV acquisition and transmission, but many challenges are encountered during the promotion of these strategies in the real-world populations, such as prevention efficiency, which still needs improvement, adherence issues, and stigmatization due to long-term medication, risk compensation behaviors after intervention, etc. To end AIDS, there should be more out-of-the-box thinking. Overall, before achieving an effective HIV vaccine, ongoing research for developing alternative biomedical interventions offers a new hope for ending AIDS in the near future.

Author Contributions

Project designed and supervised by C.S.; literature collection and original draft preparation by Y.L., Z.W. and M.S.; picture drawing by Y.L.; review and editing by H.Z. and C.S.; research was supervised by C.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (82472267, 82271786), the Science and Technology Planning Project of Guangdong Province, China (2021B1212040017, 2022B1212060001, 2023B1212130001, 2024B1212160001), the National Key Technologies R&D Program (2023YFC2306700), Fujian Province’s Third Batch of Flexible Introduction of High-Level Medical Talent Teams (TD202307), the Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety (ZDSYS20230626091203007), the Sanming Project of Medicine in Shenzhen Nanshan (No. SZSM202103008), and the Key Subject of Nanshan District of Shenzhen for AIDS surveillance and prevention.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no competing interests.

References

  1. Govindan, R.; Stephenson, K.E. HIV Vaccine Development at a Crossroads: New B and T Cell Approaches. Vaccines 2024, 12, 1043. [Google Scholar] [CrossRef] [PubMed]
  2. Siliciano, J.D.; Siliciano, R.F. HIV cure: The daunting scale of the problem. Science 2024, 383, 703–705. [Google Scholar] [CrossRef] [PubMed]
  3. UNAIDs. Global HIV & AIDS Statistics—Fact Sheet. Available online: https://www.unaids.org/en/resources/fact-sheet (accessed on 10 April 2025).
  4. Grinspoon, S.K.; Fitch, K.V.; Zanni, M.V.; Fichtenbaum, C.J.; Umbleja, T.; Aberg, J.A.; Overton, E.T.; Malvestutto, C.D.; Bloomfield, G.S.; Currier, J.S.; et al. Pitavastatin to Prevent Cardiovascular Disease in HIV Infection. N. Engl. J. Med. 2023, 389, 687–699. [Google Scholar] [CrossRef]
  5. McMyn, N.F.; Varriale, J.; Fray, E.J.; Zitzmann, C.; MacLeod, H.; Lai, J.; Singhal, A.; Moskovljevic, M.; Garcia, M.A.; Lopez, B.M.; et al. The latent reservoir of inducible, infectious HIV-1 does not decrease despite decades of antiretroviral therapy. J. Clin. Investig. 2023, 133, e171554. [Google Scholar] [CrossRef]
  6. Colby, D.J.; Trautmann, L.; Pinyakorn, S.; Leyre, L.; Pagliuzza, A.; Kroon, E.; Rolland, M.; Takata, H.; Buranapraditkun, S.; Intasan, J.; et al. Rapid HIV RNA rebound after antiretroviral treatment interruption in persons durably suppressed in Fiebig I acute HIV infection. Nat. Med. 2018, 24, 923–926. [Google Scholar] [CrossRef]
  7. Chanie, G.S.; Belachew, E.A.; Seid, A.M.; Limenh, L.W.; Mitku, M.L.; Beyna, A.T.; Mengesha, A.K.; Melese, M.; Esubalew, D.; Gela, Y.Y.; et al. Patients reported neuropsychiatric adverse events and associated factors among PLHIV patients receiving DTG-based regimen antiretroviral therapy real-life clinical practice in Ethiopia: Multi center crossetional study. BMC Psychiatry 2025, 25, 383. [Google Scholar] [CrossRef]
  8. Corti, N.; Menzaghi, B.; Orofino, G.; Guastavigna, M.; Lagi, F.; Di Biagio, A.; Taramasso, L.; De Socio, G.V.; Molteni, C.; Madeddu, G.; et al. Risk of Cardiovascular Events in People with HIV (PWH) Treated with Integrase Strand-Transfer Inhibitors: The Debate Is Not Over; Results of the SCOLTA Study. Viruses 2024, 16, 613. [Google Scholar] [CrossRef]
  9. Mwebaza, J.; Meya, D.; Musiime, V.; Birungi, C. Prevalence of neuropsychiatric adverse events and associated factors among adult patients on dolutegravir attending Mulago ISS clinic. HIV Med. 2023, 24, 491–501. [Google Scholar] [CrossRef]
  10. Deeks, S.G.; Lewin, S.R.; Havlir, D.V. The end of AIDS: HIV infection as a chronic disease. Lancet 2013, 382, 1525–1533. [Google Scholar] [CrossRef]
  11. Eisinger, R.W.; Fauci, A.S. Ending the HIV/AIDS Pandemic(1). Emerg. Infect. Dis. 2018, 24, 413–416. [Google Scholar] [CrossRef]
  12. Hu, W.S.; Hughes, S.H. HIV-1 reverse transcription. Cold Spring Harb. Perspect. Med. 2012, 2, a006882. [Google Scholar] [CrossRef] [PubMed]
  13. Nikolaitchik, O.A.; Islam, S.; Kitzrow, J.P.; Duchon, A.; Cheng, Z.; Liu, Y.; Rawson, J.M.O.; Shao, W.; Nikolaitchik, M.; Kearney, M.F.; et al. HIV-1 usurps transcription start site heterogeneity of host RNA polymerase II to maximize replication fitness. Proc. Natl. Acad. Sci. USA 2023, 120, e2305103120. [Google Scholar] [CrossRef] [PubMed]
  14. Rudometov, A.P.; Chikaev, A.N.; Rudometova, N.B.; Antonets, D.V.; Lomzov, A.A.; Kaplina, O.N.; Ilyichev, A.A.; Karpenko, L.I. Artificial Anti-HIV-1 Immunogen Comprising Epitopes of Broadly Neutralizing Antibodies 2F5, 10E8, and a Peptide Mimic of VRC01 Discontinuous Epitope. Vaccines 2019, 7, 83. [Google Scholar] [CrossRef]
  15. King, S.R.; Duggal, N.K.; Ndongmo, C.B.; Pacut, C.; Telesnitsky, A. Pseudodiploid genome organization AIDS full-length human immunodeficiency virus type 1 DNA synthesis. J. Virol. 2008, 82, 2376–2384. [Google Scholar] [CrossRef]
  16. Iglesias-Sanchez, M.J.; Lopez-Galindez, C. Each genomic RNA in HIV-1 heterozygous virus generate new virions. Virology 2005, 333, 316–323. [Google Scholar] [CrossRef]
  17. Rawson, J.M.O.; Nikolaitchik, O.A.; Keele, B.F.; Pathak, V.K.; Hu, W.S. Recombination is required for efficient HIV-1 replication and the maintenance of viral genome integrity. Nucleic Acids Res. 2018, 46, 10535–10545. [Google Scholar] [CrossRef]
  18. Onafuwa-Nuga, A.; Telesnitsky, A. The remarkable frequency of human immunodeficiency virus type 1 genetic recombination. Microbiol. Mol. Biol. Rev. MMBR 2009, 73, 451–480. [Google Scholar] [CrossRef]
  19. Zhou, Y.; Cui, M.; Hong, Z.; Huang, S.; Zhou, S.; Lyu, H.; Li, J.; Lin, Y.; Huang, H.; Tang, W.; et al. High Genetic Diversity of HIV-1 and Active Transmission Clusters among Male-to-Male Sexual Contacts (MMSCs) in Zhuhai, China. Viruses 2023, 15, 1947. [Google Scholar] [CrossRef]
  20. Li, M.; Zhou, J.; Zhang, K.; Yuan, Y.; Zhao, J.; Cui, M.; Yin, D.; Wen, Z.; Chen, Z.; Li, L.; et al. Characteristics of genotype, drug resistance, and molecular transmission network among newly diagnosed HIV-1 infections in Shenzhen, China. J. Med. Virol. 2023, 95, e28973. [Google Scholar] [CrossRef]
  21. Etienne, L.; Hahn, B.H.; Sharp, P.M.; Matsen, F.A.; Emerman, M. Gene loss and adaptation to hominids underlie the ancient origin of HIV-1. Cell Host Microbe 2013, 14, 85–92. [Google Scholar] [CrossRef]
  22. Schmitt, K.; Mohan Kumar, D.; Curlin, J.; Remling-Mulder, L.; Stenglein, M.; O’Connor, S.; Marx, P.; Akkina, R. Modeling the evolution of SIV sooty mangabey progenitor virus towards HIV-2 using humanized mice. Virology 2017, 510, 175–184. [Google Scholar] [CrossRef] [PubMed]
  23. Hemelaar, J. The origin and diversity of the HIV-1 pandemic. Trends Mol. Med. 2012, 18, 182–192. [Google Scholar] [CrossRef] [PubMed]
  24. Wertheim, J.O.; Worobey, M. Dating the age of the SIV lineages that gave rise to HIV-1 and HIV-2. PLoS Comput. Biol. 2009, 5, e1000377. [Google Scholar] [CrossRef]
  25. Korber, B.; Gaschen, B.; Yusim, K.; Thakallapally, R.; Kesmir, C.; Detours, V. Evolutionary and immunological implications of contemporary HIV-1 variation. Br. Med. Bull. 2001, 58, 19–42. [Google Scholar] [CrossRef]
  26. Schlub, T.E.; Grimm, A.J.; Smyth, R.P.; Cromer, D.; Chopra, A.; Mallal, S.; Venturi, V.; Waugh, C.; Mak, J.; Davenport, M.P. Fifteen to twenty percent of HIV substitution mutations are associated with recombination. J. Virol. 2014, 88, 3837–3849. [Google Scholar] [CrossRef]
  27. Okada, A.; Iwatani, Y. APOBEC3G-Mediated G-to-A Hypermutation of the HIV-1 Genome: The Missing Link in Antiviral Molecular Mechanisms. Front. Microbiol. 2016, 7, 2027. [Google Scholar] [CrossRef]
  28. Sadler, H.A.; Stenglein, M.D.; Harris, R.S.; Mansky, L.M. APOBEC3G contributes to HIV-1 variation through sublethal mutagenesis. J. Virol. 2010, 84, 7396–7404. [Google Scholar] [CrossRef]
  29. Zhu, T.; Niu, G.; Zhang, Y.; Chen, M.; Li, C.-Y.; Hao, L.; Zhang, Z. Host-mediated RNA editing in viruses. Biol. Direct 2023, 18, 12. [Google Scholar] [CrossRef]
  30. Feng, T.; Zhang, J.; Chen, Z.; Pan, W.; Chen, Z.; Yan, Y.; Dai, J. Glycosylation of viral proteins: Implication in virus-host interaction and virulence. Virulence 2022, 13, 670–683. [Google Scholar] [CrossRef]
  31. Vigerust, D.J.; Shepherd, V.L. Virus glycosylation: Role in virulence and immune interactions. Trends Microbiol. 2007, 15, 211–218. [Google Scholar] [CrossRef]
  32. Sullivan, J.T.; Sulli, C.; Nilo, A.; Yasmeen, A.; Ozorowski, G.; Sanders, R.W.; Ward, A.B.; Klasse, P.J.; Moore, J.P.; Doranz, B.J. High-Throughput Protein Engineering Improves the Antigenicity and Stability of Soluble HIV-1 Envelope Glycoprotein SOSIP Trimers. J. Virol. 2017, 91, 18. [Google Scholar] [CrossRef] [PubMed]
  33. Scharenberg, M.; Jiang, X.; Pang, L.; Navarra, G.; Rabbani, S.; Binder, F.; Schwardt, O.; Ernst, B. Kinetic properties of carbohydrate-lectin interactions: FimH antagonists. ChemMedChem 2014, 9, 78–83. [Google Scholar] [CrossRef] [PubMed]
  34. Scanlan, C.N.; Offer, J.; Zitzmann, N.; Dwek, R.A. Exploiting the defensive sugars of HIV-1 for drug and vaccine design. Nature 2007, 446, 1038–1045. [Google Scholar] [CrossRef] [PubMed]
  35. Seabright, G.E.; Doores, K.J.; Burton, D.R.; Crispin, M. Protein and Glycan Mimicry in HIV Vaccine Design. J. Mol. Biol. 2019, 431, 2223–2247. [Google Scholar] [CrossRef]
  36. Moore, P.L.; Gray, E.S.; Wibmer, C.K.; Bhiman, J.N.; Nonyane, M.; Sheward, D.J.; Hermanus, T.; Bajimaya, S.; Tumba, N.L.; Abrahams, M.R.; et al. Evolution of an HIV glycan-dependent broadly neutralizing antibody epitope through immune escape. Nat. Med. 2012, 18, 1688–1692. [Google Scholar] [CrossRef]
  37. Koch, M.; Pancera, M.; Kwong, P.D.; Kolchinsky, P.; Grundner, C.; Wang, L.; Hendrickson, W.A.; Sodroski, J.; Wyatt, R. Structure-based, targeted deglycosylation of HIV-1 gp120 and effects on neutralization sensitivity and antibody recognition. Virology 2003, 313, 387–400. [Google Scholar] [CrossRef]
  38. Shan, L.; Deng, K.; Gao, H.; Xing, S.; Capoferri, A.A.; Durand, C.M.; Rabi, S.A.; Laird, G.M.; Kim, M.; Hosmane, N.N.; et al. Transcriptional Reprogramming during Effector-to-Memory Transition Renders CD4(+) T Cells Permissive for Latent HIV-1 Infection. Immunity 2017, 47, 766–775.e3. [Google Scholar] [CrossRef]
  39. Eriksson, S.; Graf, E.H.; Dahl, V.; Strain, M.C.; Yukl, S.A.; Lysenko, E.S.; Bosch, R.J.; Lai, J.; Chioma, S.; Emad, F.; et al. Comparative analysis of measures of viral reservoirs in HIV-1 eradication studies. PLoS Pathog. 2013, 9, e1003174. [Google Scholar] [CrossRef]
  40. Gunst, J.D.; Gohil, J.; Li, J.Z.; Bosch, R.J.; White Catherine Seamon, A.; Chun, T.W.; Mothe, B.; Gittens, K.; Praiss, L.; De Scheerder, M.A.; et al. Time to HIV viral rebound and frequency of post-treatment control after analytical interruption of antiretroviral therapy: An individual data-based meta-analysis of 24 prospective studies. Nat. Commun. 2025, 16, 906. [Google Scholar] [CrossRef]
  41. López-Huertas, M.R.; Gutiérrez, C.; Madrid-Elena, N.; Hernández-Novoa, B.; Olalla-Sierra, J.; Plana, M.; Delgado, R.; Rubio, R.; Muñoz-Fernández, M.; Moreno, S. Prolonged administration of maraviroc reactivates latent HIV in vivo but it does not prevent antiretroviral-free viral rebound. Sci. Rep. 2020, 10, 22286. [Google Scholar] [CrossRef]
  42. Castro-Gonzalez, S.; Colomer-Lluch, M.; Serra-Moreno, R. Barriers for HIV Cure: The Latent Reservoir. AIDS Res. Hum. Retroviruses 2018, 34, 739–759. [Google Scholar] [CrossRef] [PubMed]
  43. Margolis, D.M.; Garcia, J.V.; Hazuda, D.J.; Haynes, B.F. Latency reversal and viral clearance to cure HIV-1. Science 2016, 353, aaf6517. [Google Scholar] [CrossRef] [PubMed]
  44. Pankrac, J.; Klein, K.; Mann, J.F.S. Eradication of HIV-1 latent reservoirs through therapeutic vaccination. AIDS Res. Ther. 2017, 14, 45. [Google Scholar] [CrossRef] [PubMed]
  45. Ananworanich, J.; Dubé, K.; Chomont, N. How does the timing of antiretroviral therapy initiation in acute infection affect HIV reservoirs? Curr. Opin. HIV AIDS 2015, 10, 18–28. [Google Scholar] [CrossRef]
  46. Nühn, M.M.; Gumbs, S.B.H.; Buchholtz, N.; Jannink, L.M.; Gharu, L.; de Witte, L.D.; Wensing, A.M.J.; Lewin, S.R.; Nijhuis, M.; Symons, J. Shock and kill within the CNS: A promising HIV eradication approach? J. Leukoc. Biol. 2022, 112, 1297–1315. [Google Scholar] [CrossRef]
  47. Fidler, S.; Stöhr, W.; Pace, M.; Dorrell, L.; Lever, A.; Pett, S.; Kinloch-de Loes, S.; Fox, J.; Clarke, A.; Nelson, M.; et al. Antiretroviral therapy alone versus antiretroviral therapy with a kick and kill approach, on measures of the HIV reservoir in participants with recent HIV infection (the RIVER trial): A phase 2, randomised trial. Lancet 2020, 395, 888–898. [Google Scholar] [CrossRef]
  48. Wen, Z.; Li, P.; Yuan, Y.; Wang, C.; Li, M.; Wang, H.; Shi, M.; He, Y.; Cui, M.; Chen, L.; et al. Purging viral latency by a bifunctional HSV-vectored therapeutic vaccine in chronically SIV-infected macaques. eLife 2024, 13, RP95964. [Google Scholar] [CrossRef]
  49. Vansant, G.; Bruggemans, A.; Janssens, J.; Debyser, Z. Block-And-Lock Strategies to Cure HIV Infection. Viruses 2020, 12, 84. [Google Scholar] [CrossRef]
  50. Johnson, K.A.; Chen, M.J.; Kohn, R.; Sachdev, D.; Bacon, O.; Lee, S.; Cohen, S.E. Acute HIV at the Time of Initiation of Pre-exposure or Post-exposure Prophylaxis: Impact on Drug Resistance and Clinical Outcomes. J. Acquir. Immune Defic. Syndr. (1999) 2021, 87, 818–825. [Google Scholar] [CrossRef]
  51. Hessell, A.J.; Haigwood, N.L. Animal models in HIV-1 protection and therapy. Curr. Opin. HIV AIDS 2015, 10, 170–176. [Google Scholar] [CrossRef]
  52. Ambrose, Z.; KewalRamani, V.N.; Bieniasz, P.D.; Hatziioannou, T. HIV/AIDS: In search of an animal model. Trends Biotechnol. 2007, 25, 333–337. [Google Scholar] [CrossRef] [PubMed]
  53. Klatt, N.R.; Canary, L.A.; Sun, X.; Vinton, C.L.; Funderburg, N.T.; Morcock, D.R.; Quinones, M.; Deming, C.B.; Perkins, M.; Hazuda, D.J.; et al. Probiotic/prebiotic supplementation of antiretrovirals improves gastrointestinal immunity in SIV-infected macaques. J. Clin. Investig. 2013, 123, 903–907. [Google Scholar] [CrossRef] [PubMed]
  54. Sliva, K. Latest animal models for anti-HIV drug discovery. Expert Opin. Drug Discov. 2015, 10, 111–123. [Google Scholar] [CrossRef] [PubMed]
  55. Badamchi-Zadeh, A.; Tartaglia, L.J.; Abbink, P.; Bricault, C.A.; Liu, P.T.; Boyd, M.; Kirilova, M.; Mercado, N.B.; Nanayakkara, O.S.; Vrbanac, V.D.; et al. Therapeutic Efficacy of Vectored PGT121 Gene Delivery in HIV-1-Infected Humanized Mice. J. Virol. 2018, 92, 12. [Google Scholar] [CrossRef]
  56. Dash, P.K.; Kaminski, R.; Bella, R.; Su, H.; Mathews, S.; Ahooyi, T.M.; Chen, C.; Mancuso, P.; Sariyer, R.; Ferrante, P.; et al. Sequential LASER ART and CRISPR Treatments Eliminate HIV-1 in a Subset of Infected Humanized Mice. Nat. Commun. 2019, 10, 2753. [Google Scholar] [CrossRef]
  57. Satija, N.; Patel, F.; Schmidt, G.; Doanman, D.V.; Kapoor, M.; La Porte, A.; Wang, Y.C.; Law, K.M.; Esposito, A.M.; Allette, K.; et al. Tracking HIV persistence across T cell lineages during early ART-treated HIV-1-infection using a reservoir-marking humanized mouse model. Nat. Commun. 2025, 16, 2233. [Google Scholar] [CrossRef]
  58. Barnett, E.; Kaginkar, S.; Schmitt, K.; Remling-Mulder, L.; Akkina, R. A dual-purpose humanized mouse model for testing antiviral strategies against both SIV and HIV. Front. Immunol. 2024, 15, 1491481. [Google Scholar] [CrossRef]
  59. Min, A.K.; Javidfar, B.; Missall, R.; Doanman, D.; Durens, M.; Graziani, M.; Mordelt, A.; Marro, S.G.; de Witte, L.; Chen, B.K.; et al. HIV-1 infection of genetically engineered iPSC-derived central nervous system-engrafted microglia in a humanized mouse model. J. Virol. 2023, 97, e0159523. [Google Scholar] [CrossRef]
  60. Karpel, M.E.; Boutwell, C.L.; Allen, T.M. BLT humanized mice as a small animal model of HIV infection. Curr. Opin. Virol. 2015, 13, 75–80. [Google Scholar] [CrossRef]
  61. Heeney, J.L.; Plotkin, S.A. Immunological correlates of protection from HIV infection and disease. Nat. Immunol. 2006, 7, 1281–1284. [Google Scholar] [CrossRef]
  62. Moog, C. Immune responses that correlate with HIV-1 protection? AIDS 2008, 22, 1461–1462. [Google Scholar] [CrossRef] [PubMed]
  63. Medzhitov, R.; Littman, D. HIV immunology needs a new direction. Nature 2008, 455, 591. [Google Scholar] [CrossRef] [PubMed]
  64. Miyazawa, M.; Lopalco, L.; Mazzotta, F.; Lo Caputo, S.; Veas, F.; Clerici, M. The ‘immunologic advantage’ of HIV-exposed seronegative individuals. AIDS 2009, 23, 161–175. [Google Scholar] [CrossRef] [PubMed]
  65. Gilbert, P.; Wang, M.; Wrin, T.; Petropoulos, C.; Gurwith, M.; Sinangil, F.; D’Souza, P.; Rodriguez-Chavez, I.R.; DeCamp, A.; Giganti, M.; et al. Magnitude and breadth of a nonprotective neutralizing antibody response in an efficacy trial of a candidate HIV-1 gp120 vaccine. J. Infect. Dis. 2010, 202, 595–605. [Google Scholar] [CrossRef]
  66. Tay, M.Z.; Liu, P.; Williams, L.D.; McRaven, M.D.; Sawant, S.; Gurley, T.C.; Xu, T.T.; Dennison, S.M.; Liao, H.X.; Chenine, A.L.; et al. Antibody-Mediated Internalization of Infectious HIV-1 Virions Differs among Antibody Isotypes and Subclasses. PLoS Pathog. 2016, 12, e1005817. [Google Scholar] [CrossRef]
  67. Arunachalam, P.S.; Charles, T.P.; Joag, V.; Bollimpelli, V.S.; Scott, M.K.D.; Wimmers, F.; Burton, S.L.; Labranche, C.C.; Petitdemange, C.; Gangadhara, S.; et al. T cell-inducing vaccine durably prevents mucosal SHIV infection even with lower neutralizing antibody titers. Nat. Med. 2020, 26, 932–940. [Google Scholar] [CrossRef]
  68. Zaunders, J.; van Bockel, D. Innate and Adaptive Immunity in Long-Term Non-Progression in HIV Disease. Front. Immunol. 2013, 4, 95. [Google Scholar] [CrossRef]
  69. Li, Y.; Ni, Y.; He, Q.; Hu, X.; Zhang, Y.; He, X.; Ni, M. Survival Analysis and Immune Differences of HIV Long-Term Non-progressors in Xinjiang China: A 12-Year Prospective Cohort Observation. AIDS Behav. 2024, 28, 3151–3160. [Google Scholar] [CrossRef]
  70. Mylvaganam, G.H.; Rios, D.; Abdelaal, H.M.; Iyer, S.; Tharp, G.; Mavigner, M.; Hicks, S.; Chahroudi, A.; Ahmed, R.; Bosinger, S.E.; et al. Dynamics of SIV-specific CXCR5+ CD8 T cells during chronic SIV infection. Proc. Natl. Acad. Sci. USA 2017, 114, 1976–1981. [Google Scholar] [CrossRef]
  71. Gao, D.; Wu, J.; Wu, Y.T.; Du, F.; Aroh, C.; Yan, N.; Sun, L.; Chen, Z.J. Cyclic GMP-AMP synthase is an innate immune sensor of HIV and other retroviruses. Science 2013, 341, 903–906. [Google Scholar] [CrossRef]
  72. Berkhout, B. Evolution of Live-Attenuated HIV Vaccines. BioPharm Int. 2011, 24. Available online: https://www.biopharminternational.com/view/evolution-live-attenuated-hiv-vaccines (accessed on 10 April 2025).
  73. Verity Erin, E.; Zotos, D.; Wilson, K.; Chatfield, C.; Lawson Victoria, A.; Dwyer Dominic, E.; Cunningham, A.; Learmont, J.; Dyer, W.; Sullivan, J.; et al. Viral Phenotypes and Antibody Responses in Long-Term Survivors Infected with Attenuated Human Immunodeficiency Virus Type 1 Containing Deletions in the nef and Long Terminal Repeat Regions. J. Virol. 2007, 81, 9268–9278. [Google Scholar] [CrossRef] [PubMed]
  74. Slack, C.; Ndebele, P.; Allen, M.; Salzwedel, J. Shifts in UNAIDS ethics guidance and implications for ethics review of preventive HIV vaccine trials. J. Int. AIDS Soc. 2021, 24 (Suppl. S7), e25796. [Google Scholar] [CrossRef] [PubMed]
  75. WHO. Ethical Considerations in HIV Prevention Trials. Available online: https://www.unaids.org/sites/default/files/media_asset/ethical-considerations-hiv-prevention-trials_en.pdf (accessed on 10 April 2025).
  76. Haire, B.; Folayan, M.O.; Hankins, C.; Sugarman, J.; McCormack, S.; Ramjee, G.; Warren, M. Ethical considerations in determining standard of prevention packages for HIV prevention trials: Examining PrEP. Dev. World Bioeth. 2013, 13, 87–94. [Google Scholar] [CrossRef]
  77. Dawson, L.; Garner, S.; Anude, C.; Ndebele, P.; Karuna, S.; Holt, R.; Broder, G.; Handibode, J.; Hammer, S.M.; Sobieszczyk, M.E. Testing the waters: Ethical considerations for including PrEP in a phase IIb HIV vaccine efficacy trial. Clin. Trials 2015, 12, 394–402. [Google Scholar] [CrossRef]
  78. Grant, R.M.; Lama, J.R.; Anderson, P.L.; McMahan, V.; Liu, A.Y.; Vargas, L.; Goicochea, P.; Casapía, M.; Guanira-Carranza, J.V.; Ramirez-Cardich, M.E.; et al. Preexposure chemoprophylaxis for HIV prevention in men who have sex with men. N. Engl. J. Med. 2010, 363, 2587–2599. [Google Scholar] [CrossRef]
  79. Baeten, J.M.; Donnell, D.; Ndase, P.; Mugo, N.R.; Campbell, J.D.; Wangisi, J.; Tappero, J.W.; Bukusi, E.A.; Cohen, C.R.; Katabira, E.; et al. Antiretroviral prophylaxis for HIV prevention in heterosexual men and women. N. Engl. J. Med. 2012, 367, 399–410. [Google Scholar] [CrossRef]
  80. Choopanya, K.; Martin, M.; Suntharasamai, P.; Sangkum, U.; Mock, P.A.; Leethochawalit, M.; Chiamwongpaet, S.; Kitisin, P.; Natrujirote, P.; Kittimunkong, S.; et al. Antiretroviral prophylaxis for HIV infection in injecting drug users in Bangkok, Thailand (the Bangkok Tenofovir Study): A randomised, double-blind, placebo-controlled phase 3 trial. Lancet 2013, 381, 2083–2090. [Google Scholar] [CrossRef]
  81. Stalter, R.M.; Dong, T.Q.; Hendrix, C.W.; Palanee-Phillips, T.; van der Straten, A.; Hillier, S.L.; Kiweewa, F.M.; Mgodi, N.M.; Marzinke, M.A.; Bekker, L.G.; et al. Assessing Per-Sex-Act HIV-1 Risk Reduction Among Women Using the Dapivirine Vaginal Ring. J. Infect. Dis. 2024, 229, 1158–1165. [Google Scholar] [CrossRef]
  82. Baeten, J.M.; Palanee-Phillips, T.; Brown, E.R.; Schwartz, K.; Soto-Torres, L.E.; Govender, V.; Mgodi, N.M.; Matovu Kiweewa, F.; Nair, G.; Mhlanga, F.; et al. Use of a Vaginal Ring Containing Dapivirine for HIV-1 Prevention in Women. N. Engl. J. Med. 2016, 375, 2121–2132. [Google Scholar] [CrossRef]
  83. Delany-Moretlwe, S.; Hughes, J.P.; Bock, P.; Ouma, S.G.; Hunidzarira, P.; Kalonji, D.; Kayange, N.; Makhema, J.; Mandima, P.; Mathew, C.; et al. Cabotegravir for the prevention of HIV-1 in women: Results from HPTN 084, a phase 3, randomised clinical trial. Lancet 2022, 399, 1779–1789. [Google Scholar] [CrossRef] [PubMed]
  84. Landovitz, R.J.; Donnell, D.; Clement, M.E.; Hanscom, B.; Cottle, L.; Coelho, L.; Cabello, R.; Chariyalertsak, S.; Dunne, E.F.; Frank, I.; et al. Cabotegravir for HIV Prevention in Cisgender Men and Transgender Women. N. Engl. J. Med. 2021, 385, 595–608. [Google Scholar] [CrossRef] [PubMed]
  85. Bekker, L.G.; Das, M.; Abdool Karim, Q.; Ahmed, K.; Batting, J.; Brumskine, W.; Gill, K.; Harkoo, I.; Jaggernath, M.; Kigozi, G.; et al. Twice-Yearly Lenacapavir or Daily F/TAF for HIV Prevention in Cisgender Women. N. Engl. J. Med. 2024. [Google Scholar] [CrossRef] [PubMed]
  86. Kelley, C.F.; Acevedo-Quiñones, M.; Agwu, A.L.; Avihingsanon, A.; Benson, P.; Blumenthal, J.; Brinson, C.; Brites, C.; Cahn, P.; Cantos, V.D.; et al. Twice-Yearly Lenacapavir for HIV Prevention in Men and Gender-Diverse Persons. N. Engl. J. Med. 2024. [Google Scholar] [CrossRef]
  87. Young, T.N.; Arens, F.J.; Kennedy, G.E.; Laurie, J.W.; Rutherford, G. Antiretroviral post-exposure prophylaxis (PEP) for occupational HIV exposure. Cochrane Database Syst. Rev. 2007, 2007, Cd002835. [Google Scholar] [CrossRef]
  88. Broyles, L.N.; Luo, R.; Boeras, D.; Vojnov, L. The risk of sexual transmission of HIV in individuals with low-level HIV viraemia: A systematic review. Lancet 2023, 402, 464–471. [Google Scholar] [CrossRef]
  89. Tudor Car, L.; van-Velthoven, M.H.; Brusamento, S.; Elmoniry, H.; Car, J.; Majeed, A.; Atun, R. Integrating prevention of mother-to-child HIV transmission (PMTCT) programmes with other health services for preventing HIV infection and improving HIV outcomes in developing countries. Cochrane Database Syst. Rev. 2011, Cd008741. [Google Scholar] [CrossRef]
  90. Auvert, B.; Taljaard, D.; Lagarde, E.; Sobngwi-Tambekou, J.; Sitta, R.; Puren, A. Randomized, controlled intervention trial of male circumcision for reduction of HIV infection risk: The ANRS 1265 Trial. PLoS Med. 2005, 2, e298. [Google Scholar] [CrossRef]
  91. Bailey, R.C.; Moses, S.; Parker, C.B.; Agot, K.; Maclean, I.; Krieger, J.N.; Williams, C.F.; Campbell, R.T.; Ndinya-Achola, J.O. Male circumcision for HIV prevention in young men in Kisumu, Kenya: A randomised controlled trial. Lancet 2007, 369, 643–656. [Google Scholar] [CrossRef]
  92. Gray, R.H.; Kigozi, G.; Serwadda, D.; Makumbi, F.; Watya, S.; Nalugoda, F.; Kiwanuka, N.; Moulton, L.H.; Chaudhary, M.A.; Chen, M.Z.; et al. Male circumcision for HIV prevention in men in Rakai, Uganda: A randomised trial. Lancet 2007, 369, 657–666. [Google Scholar] [CrossRef]
  93. Molina, J.M.; Capitant, C.; Spire, B.; Pialoux, G.; Cotte, L.; Charreau, I.; Tremblay, C.; Le Gall, J.M.; Cua, E.; Pasquet, A.; et al. On-Demand Preexposure Prophylaxis in Men at High Risk for HIV-1 Infection. N. Engl. J. Med. 2015, 373, 2237–2246. [Google Scholar] [CrossRef] [PubMed]
  94. Marrazzo, J.M.; Ramjee, G.; Richardson, B.A.; Gomez, K.; Mgodi, N.; Nair, G.; Palanee, T.; Nakabiito, C.; van der Straten, A.; Noguchi, L.; et al. Tenofovir-based preexposure prophylaxis for HIV infection among African women. N. Engl. J. Med. 2015, 372, 509–518. [Google Scholar] [CrossRef] [PubMed]
  95. Rodger, A.J.; Cambiano, V.; Bruun, T.; Vernazza, P.; Collins, S.; Degen, O.; Corbelli, G.M.; Estrada, V.; Geretti, A.M.; Beloukas, A.; et al. Risk of HIV transmission through condomless sex in serodifferent gay couples with the HIV-positive partner taking suppressive antiretroviral therapy (PARTNER): Final results of a multicentre, prospective, observational study. Lancet 2019, 393, 2428–2438. [Google Scholar] [CrossRef] [PubMed]
  96. Gandhi, M.; Glidden, D.V.; Mayer, K.; Schechter, M.; Buchbinder, S.; Grinsztejn, B.; Hosek, S.; Casapia, M.; Guanira, J.; Bekker, L.G.; et al. Association of age, baseline kidney function, and medication exposure with declines in creatinine clearance on pre-exposure prophylaxis: An observational cohort study. Lancet HIV 2016, 3, e521–e528. [Google Scholar] [CrossRef]
  97. Solomon, M.M.; Lama, J.R.; Glidden, D.V.; Mulligan, K.; McMahan, V.; Liu, A.Y.; Guanira, J.V.; Veloso, V.G.; Mayer, K.H.; Chariyalertsak, S.; et al. Changes in renal function associated with oral emtricitabine/tenofovir disoproxil fumarate use for HIV pre-exposure prophylaxis. AIDS 2014, 28, 851–859. [Google Scholar] [CrossRef]
  98. Parikh, U.M.; Mellors, J.W. How could HIV-1 drug resistance impact preexposure prophylaxis for HIV prevention? Curr. Opin. HIV AIDS 2022, 17, 213–221. [Google Scholar] [CrossRef]
  99. Landovitz, R.J.; Delany-Moretlwe, S.; Fogel, J.M.; Marzinke, M.A.; Piwowar-Manning, E.; Richardson, P.; Halvas, E.K.; Mellors, J.W.; Persaud, D.; Kofron, R.; et al. Features of HIV Infection in the Context of Long-Acting Cabotegravir Preexposure Prophylaxis. N. Engl. J. Med. 2024, 391, 1253–1256. [Google Scholar] [CrossRef]
  100. Segal-Maurer, S.; DeJesus, E.; Stellbrink, H.J.; Castagna, A.; Richmond, G.J.; Sinclair, G.I.; Siripassorn, K.; Ruane, P.J.; Berhe, M.; Wang, H.; et al. Capsid Inhibition with Lenacapavir in Multidrug-Resistant HIV-1 Infection. N. Engl. J. Med. 2022, 386, 1793–1803. [Google Scholar] [CrossRef]
  101. Wen, Z.; Shi, M.; Sun, C. Treatment as the Best Prevention: Twice-Yearly Lenacapavir, a Game Changer in Ending the AIDS Epidemic. Viruses 2024, 16, 1368. [Google Scholar] [CrossRef]
  102. Thigpen, M.C.; Kebaabetswe, P.M.; Paxton, L.A.; Smith, D.K.; Rose, C.E.; Segolodi, T.M.; Henderson, F.L.; Pathak, S.R.; Soud, F.A.; Chillag, K.L.; et al. Antiretroviral preexposure prophylaxis for heterosexual HIV transmission in Botswana. N. Engl. J. Med. 2012, 367, 423–434. [Google Scholar] [CrossRef]
  103. McCormack, S.; Dunn, D.T.; Desai, M.; Dolling, D.I.; Gafos, M.; Gilson, R.; Sullivan, A.K.; Clarke, A.; Reeves, I.; Schembri, G.; et al. Pre-exposure prophylaxis to prevent the acquisition of HIV-1 infection (PROUD): Effectiveness results from the pilot phase of a pragmatic open-label randomised trial. Lancet 2016, 387, 53–60. [Google Scholar] [CrossRef] [PubMed]
  104. Mayer, K.H.; Molina, J.M.; Thompson, M.A.; Anderson, P.L.; Mounzer, K.C.; De Wet, J.J.; DeJesus, E.; Jessen, H.; Grant, R.M.; Ruane, P.J.; et al. Emtricitabine and tenofovir alafenamide vs emtricitabine and tenofovir disoproxil fumarate for HIV pre-exposure prophylaxis (DISCOVER): Primary results from a randomised, double-blind, multicentre, active-controlled, phase 3, non-inferiority trial. Lancet 2020, 396, 239–254. [Google Scholar] [CrossRef] [PubMed]
  105. Cottrell, M.L.; Yang, K.H.; Prince, H.M.; Sykes, C.; White, N.; Malone, S.; Dellon, E.S.; Madanick, R.D.; Shaheen, N.J.; Hudgens, M.G.; 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. [Google Scholar] [CrossRef] [PubMed]
  106. Nel, A.; van Niekerk, N.; Kapiga, S.; Bekker, L.G.; Gama, C.; Gill, K.; Kamali, A.; Kotze, P.; Louw, C.; Mabude, Z.; et al. Safety and Efficacy of a Dapivirine Vaginal Ring for HIV Prevention in Women. N. Engl. J. Med. 2016, 375, 2133–2143. [Google Scholar] [CrossRef]
  107. WHO. WHO Recommends the Dapivirine Vaginal Ring as a New HIV Prevention Option for Women at Substantial Risk of HIV Infection. Available online: https://www.who.int/news/item/26-01-2021-who-recommends-the-dapivirine-vaginal-ring-as-a-new-choice-for-hiv-prevention-for-women-at-substantial-risk-of-hiv-infection (accessed on 10 April 2025).
  108. Krogstad, E.A.; Montgomery, E.T.; Atujuna, M.; Minnis, A.M.; O’Rourke, S.; Ahmed, K.; Bekker, L.G.; van der Straten, A. Design of an Implant for Long-Acting HIV Pre-Exposure Prophylaxis: Input from South African Health Care Providers. AIDS Patient Care STDs 2019, 33, 157–166. [Google Scholar] [CrossRef]
  109. Chua, C.Y.X.; Jain, P.; Ballerini, A.; Bruno, G.; Hood, R.L.; Gupte, M.; Gao, S.; Di Trani, N.; Susnjar, A.; Shelton, K.; et al. Transcutaneously refillable nanofluidic implant achieves sustained level of tenofovir diphosphate for HIV pre-exposure prophylaxis. J. Control. Release 2018, 286, 315–325. [Google Scholar] [CrossRef]
  110. Johnson, L.M.; Krovi, S.A.; Li, L.; Girouard, N.; Demkovich, Z.R.; Myers, D.; Creelman, B.; van der Straten, A. Characterization of a Reservoir-Style Implant for Sustained Release of Tenofovir Alafenamide (TAF) for HIV Pre-Exposure Prophylaxis (PrEP). Pharmaceutics 2019, 11, 315. [Google Scholar] [CrossRef]
  111. Gunawardana, M.; Remedios-Chan, M.; Sanchez, D.; Fanter, R.; Webster, S.; Webster, P.; Moss, J.A.; Trinh, M.; Beliveau, M.; Ramirez, C.M.; et al. Preclinical Considerations for Long-acting Delivery of Tenofovir Alafenamide from Subdermal Implants for HIV Pre-exposure Prophylaxis. Pharm. Res. 2023, 40, 1657–1672. [Google Scholar] [CrossRef]
  112. Li, L.; Areson, C.; van der Straten, A.; Johnson, L.M. Effects of Polymer Blending on the Performance of a Subcutaneous Biodegradable Implant for HIV Pre-Exposure Prophylaxis (PrEP). Int. J. Mol. Sci. 2021, 22, 6529. [Google Scholar] [CrossRef]
  113. Barrett, S.E.; Teller, R.S.; Forster, S.P.; Li, L.; Mackey, M.A.; Skomski, D.; Yang, Z.; Fillgrove, K.L.; Doto, G.J.; Wood, S.L.; et al. Extended-Duration MK-8591-Eluting Implant as a Candidate for HIV Treatment and Prevention. Antimicrob. Agents Chemother. 2018, 62, e01058-18. [Google Scholar] [CrossRef]
  114. Kovarova, M.; Benhabbour, S.R.; Massud, I.; Spagnuolo, R.A.; Skinner, B.; Baker, C.E.; Sykes, C.; Mollan, K.R.; Kashuba, A.D.M.; García-Lerma, J.G.; et al. Ultra-long-acting removable drug delivery system for HIV treatment and prevention. Nat. Commun. 2018, 9, 4156. [Google Scholar] [CrossRef]
  115. Su, J.T.; Simpson, S.M.; Sung, S.; Tfaily, E.B.; Veazey, R.; Marzinke, M.; Qiu, J.; Watrous, D.; Widanapathirana, L.; Pearson, E.; et al. A Subcutaneous Implant of Tenofovir Alafenamide Fumarate Causes Local Inflammation and Tissue Necrosis in Rabbits and Macaques. Antimicrob. Agents Chemother. 2020, 64, e01893-19. [Google Scholar] [CrossRef] [PubMed]
  116. Gatto, G.J.; Krovi, A.; Li, L.; Massud, I.; Holder, A.; Gary, J.; Mills, P.; Mitchell, J.; Luecke, E.; Demkovich, Z.R.; et al. Comparative Pharmacokinetics and Local Tolerance of Tenofovir Alafenamide (TAF) From Subcutaneous Implant in Rabbits, Dogs, and Macaques. Front. Pharmacol. 2022, 13, 923954. [Google Scholar] [CrossRef] [PubMed]
  117. Barber, T.J.; Benn, P.D. Postexposure prophylaxis for HIV following sexual exposure. Curr. Opin. HIV AIDS 2010, 5, 322–326. [Google Scholar] [CrossRef] [PubMed]
  118. Tsai, C.C.; Follis, K.E.; Sabo, A.; Beck, T.W.; Grant, R.F.; Bischofberger, N.; Benveniste, R.E.; Black, R. Prevention of SIV infection in macaques by (R)-9-(2-phosphonylmethoxypropyl)adenine. Science 1995, 270, 1197–1199. [Google Scholar] [CrossRef]
  119. Otten, R.A.; Smith, D.K.; Adams, D.R.; Pullium, J.K.; Jackson, E.; Kim, C.N.; Jaffe, H.; Janssen, R.; Butera, S.; Folks, T.M. Efficacy of postexposure prophylaxis after intravaginal exposure of pig-tailed macaques to a human-derived retrovirus (human immunodeficiency virus type 2). J. Virol. 2000, 74, 9771–9775. [Google Scholar] [CrossRef]
  120. Ayieko, J.; Petersen, M.L.; Kabami, J.; Mwangwa, F.; Opel, F.; Nyabuti, M.; Charlebois, E.D.; Peng, J.; Koss, C.A.; Balzer, L.B.; et al. Uptake and outcomes of a novel community-based HIV post-exposure prophylaxis (PEP) programme in rural Kenya and Uganda. J. Int. AIDS Soc. 2021, 24, e25670. [Google Scholar] [CrossRef]
  121. Isah, A.; Igboeli, N.U.; Dim, O.F.; Ekwuofu, A.A. HIV infections averted at PEPFAR-APIN clinics in Nigeria: A ten-year retrospective evaluation of the clinical outcomes of post-exposure prophylaxis services. Afr. J. AIDS Res. AJAR 2023, 22, 46–53. [Google Scholar] [CrossRef]
  122. Smith, D.K.; Grohskopf, L.A.; Black, R.J.; Auerbach, J.D.; Veronese, F.; Struble, K.A.; Cheever, L.; Johnson, M.; Paxton, L.A.; Onorato, I.M.; et al. Antiretroviral postexposure prophylaxis after sexual, injection-drug use, or other nonoccupational exposure to HIV in the United States: Recommendations from the U.S. Department of Health and Human Services. MMWR. Recomm. Rep. Morb. Mortal. Wkly. Rep. Recomm. Rep. 2005, 54, 1–20. [Google Scholar]
  123. Centers for Disease Control and Prevention (CDC). Update: Provisional Public Health Service recommendations for chemoprophylaxis after occupational exposure to HIV. MMWR. Morb. Mortal. Wkly. Rep. 1996, 45, 468–480. [Google Scholar]
  124. Announcement: Updated Guidelines for Antiretroviral Postexposure Prophylaxis after Sexual, Injection-Drug Use, or Other Nonoccupational Exposure to HIV—United States, 2016. MMWR. Morb. Mortal. Wkly. Rep. 2016, 65, 458. [CrossRef] [PubMed]
  125. Cresswell, F.; Asanati, K.; Bhagani, S.; Boffito, M.; Delpech, V.; Ellis, J.; Fox, J.; Furness, L.; Kingston, M.; Mansouri, M.; et al. UK guideline for the use of HIV post-exposure prophylaxis 2021. HIV Med. 2022, 23, 494–545. [Google Scholar] [CrossRef] [PubMed]
  126. WHO. Guidelines for HIV Post-Exposure Prophylaxis. Available online: https://iris.who.int/bitstream/handle/10665/378221/9789240095137-eng.pdf?sequence=1 (accessed on 10 April 2025).
  127. Eaton, L.A.; Kalichman, S. Risk compensation in HIV prevention: Implications for vaccines, microbicides, and other biomedical HIV prevention technologies. Curr. HIV/AIDS Rep. 2007, 4, 165–172. [Google Scholar] [CrossRef] [PubMed]
  128. Roland, M.E.; Neilands, T.B.; Krone, M.R.; Katz, M.H.; Franses, K.; Grant, R.M.; Busch, M.P.; Hecht, F.M.; Shacklett, B.L.; Kahn, J.O.; et al. Seroconversion following nonoccupational postexposure prophylaxis against HIV. Clin. Infect. Dis. 2005, 41, 1507–1513. [Google Scholar] [CrossRef]
  129. Kahn, J.O.; Martin, J.N.; Roland, M.E.; Bamberger, J.D.; Chesney, M.; Chambers, D.; Franses, K.; Coates, T.J.; Katz, M.H. Feasibility of postexposure prophylaxis (PEP) against human immunodeficiency virus infection after sexual or injection drug use exposure: The San Francisco PEP Study. J. Infect. Dis. 2001, 183, 707–714. [Google Scholar] [CrossRef]
  130. Winston, A.; McAllister, J.; Amin, J.; Cooper, D.A.; Carr, A. The use of a triple nucleoside-nucleotide regimen for nonoccupational HIV post-exposure prophylaxis. HIV Med. 2005, 6, 191–197. [Google Scholar] [CrossRef]
  131. Mayer, K.H.; Mimiaga, M.J.; Cohen, D.; Grasso, C.; Bill, R.; Van Derwarker, R.; Fisher, A. Tenofovir DF plus lamivudine or emtricitabine for nonoccupational postexposure prophylaxis (NPEP) in a Boston Community Health Center. J. Acquir. Immune Defic. Syndr. (1999) 2008, 47, 494–499. [Google Scholar] [CrossRef]
  132. Tosini, W.; Muller, P.; Prazuck, T.; Benabdelmoumen, G.; Peyrouse, E.; Christian, B.; Quertainmont, Y.; Bouvet, E.; Rabaud, C. Tolerability of HIV postexposure prophylaxis with tenofovir/emtricitabine and lopinavir/ritonavir tablet formulation. AIDS 2010, 24, 2375–2380. [Google Scholar] [CrossRef]
  133. Diaz-Brito, V.; León, A.; Knobel, H.; Peraire, J.; Domingo, P.; Clotet, B.; Dalmau, D.; Cruceta, A.; Arnaiz, J.A.; Gatell, J.M.; et al. Post-exposure prophylaxis for HIV infection: A clinical trial comparing lopinavir/ritonavir versus atazanavir each with zidovudine/lamivudine. Antivir. Ther. 2012, 17, 337–346. [Google Scholar] [CrossRef]
  134. McAllister, J.; Read, P.; McNulty, A.; Tong, W.W.; Ingersoll, A.; Carr, A. Raltegravir-emtricitabine-tenofovir as HIV nonoccupational post-exposure prophylaxis in men who have sex with men: Safety, tolerability and adherence. HIV Med. 2014, 15, 13–22. [Google Scholar] [CrossRef]
  135. Leal, L.; León, A.; Torres, B.; Inciarte, A.; Lucero, C.; Mallolas, J.; Laguno, M.; Martínez-Rebollar, M.; González-Cordón, A.; Manzardo, C.; et al. A randomized clinical trial comparing ritonavir-boosted lopinavir versus raltegravir each with tenofovir plus emtricitabine for post-exposure prophylaxis for HIV infection. J. Antimicrob. Chemother. 2016, 71, 1987–1993. [Google Scholar] [CrossRef] [PubMed]
  136. Leal, L.; León, A.; Torres, B.; Inciarte, A.; Lucero, C.; Mallolas, J.; Laguno, M.; Martínez-Rebollar, M.; González-Cordón, A.; Manzardo, C.; et al. A randomized clinical trial comparing ritonavir-boosted lopinavir versus maraviroc each with tenofovir plus emtricitabine for post-exposure prophylaxis for HIV infection. J. Antimicrob. Chemother. 2016, 71, 1982–1986. [Google Scholar] [CrossRef] [PubMed]
  137. Fätkenheuer, G.; Jessen, H.; Stoehr, A.; Jung, N.; Jessen, A.B.; Kümmerle, T.; Berger, M.; Bogner, J.R.; Spinner, C.D.; Stephan, C.; et al. PEPDar: A randomized prospective noninferiority study of ritonavir-boosted darunavir for HIV post-exposure prophylaxis. HIV Med. 2016, 17, 453–459. [Google Scholar] [CrossRef] [PubMed]
  138. Valin, N.; Fonquernie, L.; Daguenel, A.; Campa, P.; Anthony, T.; Guiguet, M.; Girard, P.M.; Meyohas, M.C. Evaluation of tolerability with the co-formulation elvitegravir, cobicistat, emtricitabine, and tenofovir disoproxil fumarate for post-HIV exposure prophylaxis. BMC Infect. Dis. 2016, 16, 718. [Google Scholar] [CrossRef]
  139. Milinkovic, A.; Benn, P.; Arenas-Pinto, A.; Brima, N.; Copas, A.; Clarke, A.; Fisher, M.; Schembri, G.; Hawkins, D.; Williams, A.; et al. Randomized controlled trial of the tolerability and completion of maraviroc compared with Kaletra® in combination with Truvada® for HIV post-exposure prophylaxis (MiPEP Trial). J. Antimicrob. Chemother. 2017, 72, 1760–1768. [Google Scholar] [CrossRef]
  140. Chauveau, M.; Billaud, E.; Bonnet, B.; Merrien, D.; Hitoto, H.; Bouchez, S.; Michau, C.; Hall, N.; Perez, L.; Sécher, S.; et al. Tenofovir DF/emtricitabine/rilpivirine as HIV post-exposure prophylaxis: Results from a multicentre prospective study. J. Antimicrob. Chemother. 2019, 74, 1021–1027. [Google Scholar] [CrossRef]
  141. Nie, J.; Sun, F.; He, X.; Liu, J.; Wang, M.; Li, C.; Gu, S.; Chen, Z.; Li, Y.; Chen, Y. Tolerability and Adherence of Antiretroviral Regimens Containing Long-Acting Fusion Inhibitor Albuvirtide for HIV Post-Exposure Prophylaxis: A Cohort Study in China. Infect. Dis. Ther. 2021, 10, 2611–2623. [Google Scholar] [CrossRef]
  142. Liu, A.; Xin, R.; Zhang, H.; Dai, L.; Wu, R.E.; Wang, X.; Li, A.; Hua, W.; Li, J.; Shao, Y.; et al. An open-label evaluation of safety and tolerability of coformulated bictegravir/emtricitabine/tenofovir alafenamide for post-exposure prophylaxis following potential exposure to human immunodeficiency virus-1. Chin. Med. J. 2022, 135, 2725–2729. [Google Scholar] [CrossRef]
  143. Eisinger, R.W.; Dieffenbach, C.W.; Fauci, A.S. HIV Viral Load and Transmissibility of HIV Infection: Undetectable Equals Untransmittable. JAMA 2019, 321, 451–452. [Google Scholar] [CrossRef]
  144. Cohen, M.S.; Chen, Y.Q.; McCauley, M.; Gamble, T.; Hosseinipour, M.C.; Kumarasamy, N.; Hakim, J.G.; Kumwenda, J.; Grinsztejn, B.; Pilotto, J.H.; et al. Prevention of HIV-1 infection with early antiretroviral therapy. N. Engl. J. Med. 2011, 365, 493–505. [Google Scholar] [CrossRef]
  145. Rodger, A.J.; Cambiano, V.; Bruun, T.; Vernazza, P.; Collins, S.; van Lunzen, J.; Corbelli, G.M.; Estrada, V.; Geretti, A.M.; Beloukas, A.; et al. Sexual Activity Without Condoms and Risk of HIV Transmission in Serodifferent Couples When the HIV-Positive Partner Is Using Suppressive Antiretroviral Therapy. JAMA 2016, 316, 171–181. [Google Scholar] [CrossRef] [PubMed]
  146. LeMessurier, J.; Traversy, G.; Varsaneux, O.; Weekes, M.; Avey, M.T.; Niragira, O.; Gervais, R.; Guyatt, G.; Rodin, R. Risk of sexual transmission of human immunodeficiency virus with antiretroviral therapy, suppressed viral load and condom use: A systematic review. CMAJ Can. Med. Assoc. J. = J. De L’association Medicale Can. 2018, 190, E1350–E1360. [Google Scholar] [CrossRef] [PubMed]
  147. Thomford, N.E.; Mhandire, D.; Dandara, C.; Kyei, G.B. Promoting Undetectable Equals Untransmittable in Sub-Saharan Africa: Implication for Clinical Practice and ART Adherence. Int. J. Environ. Res. Public Health 2020, 17, 6163. [Google Scholar] [CrossRef] [PubMed]
  148. WHO. Guideline on When to Start Antiretroviral Therapy and on Pre-Exposure Prophylaxis for HIV. Available online: https://iris.who.int/bitstream/handle/10665/186275/9789241509565_eng.pdf?sequence=1 (accessed on 10 April 2025).
  149. Metzger, V.T.; Lloyd-Smith, J.O.; Weinberger, L.S. Autonomous targeting of infectious superspreaders using engineered transmissible therapies. PLoS Comput. Biol. 2011, 7, e1002015. [Google Scholar] [CrossRef]
  150. Pitchai, F.N.N.; Tanner, E.J.; Khetan, N.; Vasen, G.; Levrel, C.; Kumar, A.J.; Pandey, S.; Ordonez, T.; Barnette, P.; Spencer, D.; et al. Engineered deletions of HIV replicate conditionally to reduce disease in nonhuman primates. Science 2024, 385, eadn5866. [Google Scholar] [CrossRef]
  151. Viguerie, A.; Gopalappa, C.; Lyles, C.M.; Farnham, P.G. The effects of HIV self-testing on HIV incidence and awareness of status among men who have sex with men in the United States: Insights from a novel compartmental model. Epidemics 2024, 49, 100796. [Google Scholar] [CrossRef]
  152. Booton, R.D.; Ong, J.J.; Lee, A.; Liu, A.; Huang, W.; Wei, C.; Tang, W.; Ma, W.; Vickerman, P.; Tucker, J.D.; et al. Modelling the impact of an HIV testing intervention on HIV transmission among men who have sex with men in China. HIV Med. 2021, 22, 467–477. [Google Scholar] [CrossRef]
  153. WHO. Mother-to-Child Transmission of HIV. Available online: https://www.who.int/teams/global-hiv-hepatitis-and-stis-programmes/hiv/prevention/mother-to-child-transmission-of-hiv (accessed on 10 April 2025).
  154. Kesho Bora Study, G.; de Vincenzi, I. Triple antiretroviral compared with zidovudine and single-dose nevirapine prophylaxis during pregnancy and breastfeeding for prevention of mother-to-child transmission of HIV-1 (Kesho Bora study): A randomised controlled trial. Lancet. Infect. Dis. 2011, 11, 171–180. [Google Scholar] [CrossRef]
  155. European Mode of Delivery, C. Elective caesarean-section versus vaginal delivery in prevention of vertical HIV-1 transmission: A randomised clinical trial. Lancet 1999, 353, 1035–1039. [Google Scholar] [CrossRef]
  156. Hirbod, T.; Bailey, R.C.; Agot, K.; Moses, S.; Ndinya-Achola, J.; Murugu, R.; Andersson, J.; Nilsson, J.; Broliden, K. Abundant expression of HIV target cells and C-type lectin receptors in the foreskin tissue of young Kenyan men. Am. J. Pathol. 2010, 176, 2798–2805. [Google Scholar] [CrossRef]
  157. Galiwango, R.M.; Yegorov, S.; Joag, V.; Prodger, J.; Shahabi, K.; Huibner, S.; Muyanja, E.; Kabuubi, B.R.; Namuniina, A.; Nalutaaya, A.; et al. Characterization of CD4(+) T cell subsets and HIV susceptibility in the inner and outer foreskin of Ugandan men. Am. J. Reprod. Immunol. 2019, 82, e13143. [Google Scholar] [CrossRef] [PubMed]
  158. Galiwango, R.M.; Park, D.E.; Huibner, S.; Onos, A.; Aziz, M.; Roach, K.; Anok, A.; Nnamutete, J.; Isabirye, Y.; Wasswa, J.B.; et al. Immune milieu and microbiome of the distal urethra in Ugandan men: Impact of penile circumcision and implications for HIV susceptibility. Microbiome 2022, 10, 7. [Google Scholar] [CrossRef] [PubMed]
  159. Liu, C.M.; Hungate, B.A.; Tobian, A.A.; Serwadda, D.; Ravel, J.; Lester, R.; Kigozi, G.; Aziz, M.; Galiwango, R.M.; Nalugoda, F.; et al. Male circumcision significantly reduces prevalence and load of genital anaerobic bacteria. mBio 2013, 4, e00076. [Google Scholar] [CrossRef] [PubMed]
  160. Tobian, A.A.; Serwadda, D.; Quinn, T.C.; Kigozi, G.; Gravitt, P.E.; Laeyendecker, O.; Charvat, B.; Ssempijja, V.; Riedesel, M.; Oliver, A.E.; et al. Male circumcision for the prevention of HSV-2 and HPV infections and syphilis. N. Engl. J. Med. 2009, 360, 1298–1309. [Google Scholar] [CrossRef]
  161. Tobian, A.A.; Gray, R.H.; Quinn, T.C. Male circumcision for the prevention of acquisition and transmission of sexually transmitted infections: The case for neonatal circumcision. Arch. Pediatr. Adolesc. Med. 2010, 164, 78–84. [Google Scholar] [CrossRef]
  162. UNAIDs. UNAIDS Data 2021. Available online: https://www.unaids.org/en/resources/documents/2021/2021_unaids_data (accessed on 3 May 2025).
  163. Gao, Y.; Zhan, Y.; Sun, Y.; Zheng, W.; Zhang, W.; Fu, L.; Guo, Z.; Lin, Y.F.; Li, Y.; Zheng, L.; et al. Efficacy of Voluntary Medical Male Circumcision to Prevent HIV Infection Among Men Who Have Sex With Men: A Randomized Controlled Trial. Ann. Intern. Med. 2024, 177, 719–728. [Google Scholar] [CrossRef]
  164. Yuan, T.; Fitzpatrick, T.; Ko, N.Y.; Cai, Y.; Chen, Y.; Zhao, J.; Li, L.; Xu, J.; Gu, J.; Li, J.; et al. Circumcision to prevent HIV and other sexually transmitted infections in men who have sex with men: A systematic review and meta-analysis of global data. Lancet. Glob. Health 2019, 7, e436–e447. [Google Scholar] [CrossRef]
  165. Hütter, G.; Nowak, D.; Mossner, M.; Ganepola, S.; Müssig, A.; Allers, K.; Schneider, T.; Hofmann, J.; Kücherer, C.; Blau, O.; et al. Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N. Engl. J. Med. 2009, 360, 692–698. [Google Scholar] [CrossRef]
  166. Gupta, R.K.; Abdul-Jawad, S.; McCoy, L.E.; Mok, H.P.; Peppa, D.; Salgado, M.; Martinez-Picado, J.; Nijhuis, M.; Wensing, A.M.J.; Lee, H.; et al. HIV-1 remission following CCR5Δ32/Δ32 haematopoietic stem-cell transplantation. Nature 2019, 568, 244–248. [Google Scholar] [CrossRef]
  167. Hsu, J.; Van Besien, K.; Glesby, M.J.; Pahwa, S.; Coletti, A.; Warshaw, M.G.; Petz, L.; Moore, T.B.; Chen, Y.H.; Pallikkuth, S.; et al. HIV-1 remission and possible cure in a woman after haplo-cord blood transplant. Cell 2023, 186, 1115–1126.e8. [Google Scholar] [CrossRef]
  168. Jensen, B.O.; Knops, E.; Cords, L.; Lübke, N.; Salgado, M.; Busman-Sahay, K.; Estes, J.D.; Huyveneers, L.E.P.; Perdomo-Celis, F.; Wittner, M.; et al. In-depth virological and immunological characterization of HIV-1 cure after CCR5Δ32/Δ32 allogeneic hematopoietic stem cell transplantation. Nat. Med. 2023, 29, 583–587. [Google Scholar] [CrossRef] [PubMed]
  169. Dickter, J.K.; Aribi, A.; Cardoso, A.A.; Gianella, S.; Gendzekhadze, K.; Li, S.; Feng, Y.; Chaillon, A.; Laird, G.M.; Browning, D.L.; et al. HIV-1 Remission after Allogeneic Hematopoietic-Cell Transplantation. N. Engl. J. Med. 2024, 390, 669–671. [Google Scholar] [CrossRef] [PubMed]
  170. Ni, J.; Wang, D.; Wang, S. The CCR5-Delta32 Genetic Polymorphism and HIV-1 Infection Susceptibility: A Meta-analysis. Open Med. 2018, 13, 467–474. [Google Scholar] [CrossRef] [PubMed]
  171. Xu, L.; Yang, H.; Gao, Y.; Chen, Z.; Xie, L.; Liu, Y.; Liu, Y.; Wang, X.; Li, H.; Lai, W.; et al. CRISPR/Cas9-Mediated CCR5 Ablation in Human Hematopoietic Stem/Progenitor Cells Confers HIV-1 Resistance In Vivo. Mol. Ther. J. Am. Soc. Gene Ther. 2017, 25, 1782–1789. [Google Scholar] [CrossRef]
  172. Tebas, P.; Stein, D.; Tang, W.W.; Frank, I.; Wang, S.Q.; Lee, G.; Spratt, S.K.; Surosky, R.T.; Giedlin, M.A.; Nichol, G.; et al. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N. Engl. J. Med. 2014, 370, 901–910. [Google Scholar] [CrossRef]
  173. Xu, L.; Wang, J.; Liu, Y.; Xie, L.; Su, B.; Mou, D.; Wang, L.; Liu, T.; Wang, X.; Zhang, B.; et al. CRISPR-Edited Stem Cells in a Patient with HIV and Acute Lymphocytic Leukemia. N. Engl. J. Med. 2019, 381, 1240–1247. [Google Scholar] [CrossRef]
  174. Kaminski, R.; Chen, Y.; Fischer, T.; Tedaldi, E.; Napoli, A.; Zhang, Y.; Karn, J.; Hu, W.; Khalili, K. Elimination of HIV-1 Genomes from Human T-lymphoid Cells by CRISPR/Cas9 Gene Editing. Sci. Rep. 2016, 6, 22555. [Google Scholar] [CrossRef]
  175. Mancuso, P.; Chen, C.; Kaminski, R.; Gordon, J.; Liao, S.; Robinson, J.A.; Smith, M.D.; Liu, H.; Sariyer, I.K.; Sariyer, R.; et al. CRISPR based editing of SIV proviral DNA in ART treated non-human primates. Nat. Commun. 2020, 11, 6065. [Google Scholar] [CrossRef]
  176. Johnson, N.M.; Alvarado, A.F.; Moffatt, T.N.; Edavettal, J.M.; Swaminathan, T.A.; Braun, S.E. HIV-based lentiviral vectors: Origin and sequence differences. Mol. Ther. Methods Clin. Dev. 2021, 21, 451–465. [Google Scholar] [CrossRef]
  177. Munis, A.M. Gene Therapy Applications of Non-Human Lentiviral Vectors. Viruses 2020, 12, 1106. [Google Scholar] [CrossRef]
  178. Zhang, F.; Wen, Y.; Guo, X. CRISPR/Cas9 for genome editing: Progress, implications and challenges. Hum. Mol. Genet. 2014, 23, R40–R46. [Google Scholar] [CrossRef] [PubMed]
  179. Wang, G.; Zhao, N.; Berkhout, B.; Das, A.T. CRISPR-Cas9 Can Inhibit HIV-1 Replication but NHEJ Repair Facilitates Virus Escape. Mol. Ther. J. Am. Soc. Gene Ther. 2016, 24, 522–526. [Google Scholar] [CrossRef] [PubMed]
  180. Hacein-Bey-Abina, S.; Garrigue, A.; Wang, G.P.; Soulier, J.; Lim, A.; Morillon, E.; Clappier, E.; Caccavelli, L.; Delabesse, E.; Beldjord, K.; et al. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J. Clin. Investig. 2008, 118, 3132–3142. [Google Scholar] [CrossRef] [PubMed]
  181. Hacein-Bey-Abina, S.; Von Kalle, C.; Schmidt, M.; McCormack, M.P.; Wulffraat, N.; Leboulch, P.; Lim, A.; Osborne, C.S.; Pawliuk, R.; Morillon, E.; et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 2003, 302, 415–419. [Google Scholar] [CrossRef]
  182. Leggat, D.J.; Cohen, K.W.; Willis, J.R.; Fulp, W.J.; deCamp, A.C.; Kalyuzhniy, O.; Cottrell, C.A.; Menis, S.; Finak, G.; Ballweber-Fleming, L.; et al. Vaccination induces HIV broadly neutralizing antibody precursors in humans. Science 2022, 378, eadd6502. [Google Scholar] [CrossRef]
  183. Haynes, B.F.; Wiehe, K.; Borrow, P.; Saunders, K.O.; Korber, B.; Wagh, K.; McMichael, A.J.; Kelsoe, G.; Hahn, B.H.; Alt, F.; et al. Strategies for HIV-1 vaccines that induce broadly neutralizing antibodies. Nat. Rev. Immunol. 2023, 23, 142–158. [Google Scholar] [CrossRef]
  184. Sun, C.; Zuo, T.; Wen, Z. B cell engineering in vivo: Accelerating induction of broadly neutralizing antibodies against HIV-1 infection. Signal Transduct. Target. Ther. 2023, 8, 13. [Google Scholar] [CrossRef]
  185. McFarland, E.J.; Cunningham, C.K.; Muresan, P.; Capparelli, E.V.; Perlowski, C.; Morgan, P.; Smith, B.; Hazra, R.; Purdue, L.; Harding, P.A.; et al. Safety, Tolerability, and Pharmacokinetics of a Long-Acting Broadly Neutralizing Human Immunodeficiency Virus Type 1 (HIV-1) Monoclonal Antibody VRC01LS in HIV-1-Exposed Newborn Infants. J. Infect. Dis. 2021, 224, 1916–1924. [Google Scholar] [CrossRef]
  186. Cunningham, C.K.; McFarland, E.J.; Morrison, R.L.; Capparelli, E.V.; Safrit, J.T.; Mofenson, L.M.; Mathieson, B.; Valentine, M.E.; Perlowski, C.; Smith, B.; et al. Safety, Tolerability, and Pharmacokinetics of the Broadly Neutralizing Human Immunodeficiency Virus (HIV)-1 Monoclonal Antibody VRC01 in HIV-Exposed Newborn Infants. J. Infect. Dis. 2020, 222, 628–636. [Google Scholar] [CrossRef]
  187. Corey, L.; Gilbert, P.B.; Juraska, M.; Montefiori, D.C.; Morris, L.; Karuna, S.T.; Edupuganti, S.; Mgodi, N.M.; deCamp, A.C.; Rudnicki, E.; et al. Two Randomized Trials of Neutralizing Antibodies to Prevent HIV-1 Acquisition. N. Engl. J. Med. 2021, 384, 1003–1014. [Google Scholar] [CrossRef]
  188. Bar, K.J.; Sneller, M.C.; Harrison, L.J.; Justement, J.S.; Overton, E.T.; Petrone, M.E.; Salantes, D.B.; Seamon, C.A.; Scheinfeld, B.; Kwan, R.W.; et al. Effect of HIV Antibody VRC01 on Viral Rebound after Treatment Interruption. N. Engl. J. Med. 2016, 375, 2037–2050. [Google Scholar] [CrossRef] [PubMed]
  189. Davis-Gardner, M.E.; Alfant, B.; Weber, J.A.; Gardner, M.R.; Farzan, M. A Bispecific Antibody That Simultaneously Recognizes the V2- and V3-Glycan Epitopes of the HIV-1 Envelope Glycoprotein Is Broader and More Potent than Its Parental Antibodies. mBio 2020, 11, 16. [Google Scholar] [CrossRef] [PubMed]
  190. Xu, L.; Pegu, A.; Rao, E.; Doria-Rose, N.; Beninga, J.; McKee, K.; Lord, D.M.; Wei, R.R.; Deng, G.; Louder, M.; et al. Trispecific broadly neutralizing HIV antibodies mediate potent SHIV protection in macaques. Science 2017, 358, 85–90. [Google Scholar] [CrossRef] [PubMed]
  191. Gautam, R.; Nishimura, Y.; Pegu, A.; Nason, M.C.; Klein, F.; Gazumyan, A.; Golijanin, J.; Buckler-White, A.; Sadjadpour, R.; Wang, K.; et al. A single injection of anti-HIV-1 antibodies protects against repeated SHIV challenges. Nature 2016, 533, 105–109. [Google Scholar] [CrossRef]
  192. Sok, D.; Burton, D.R. Recent progress in broadly neutralizing antibodies to HIV. Nat. Immunol. 2018, 19, 1179–1188. [Google Scholar] [CrossRef]
  193. Gaudinski, M.R.; Houser, K.V.; Doria-Rose, N.A.; Chen, G.L.; Rothwell, R.S.S.; Berkowitz, N.; Costner, P.; Holman, L.A.; Gordon, I.J.; Hendel, C.S.; et al. Safety and pharmacokinetics of broadly neutralising human monoclonal antibody VRC07-523LS in healthy adults: A phase 1 dose-escalation clinical trial. Lancet HIV 2019, 6, e667–e679. [Google Scholar] [CrossRef]
  194. Balazs, A.B.; Ouyang, Y.; Hong, C.M.; Chen, J.; Nguyen, S.M.; Rao, D.S.; An, D.S.; Baltimore, D. Vectored immunoprophylaxis protects humanized mice from mucosal HIV transmission. Nat. Med. 2014, 20, 296–300. [Google Scholar] [CrossRef]
  195. Gardner, M.R.; Fellinger, C.H.; Kattenhorn, L.M.; Davis-Gardner, M.E.; Weber, J.A.; Alfant, B.; Zhou, A.S.; Prasad, N.R.; Kondur, H.R.; Newton, W.A.; et al. AAV-delivered eCD4-Ig protects rhesus macaques from high-dose SIVmac239 challenges. Sci. Transl. Med. 2019, 11, aau5409. [Google Scholar] [CrossRef]
  196. Gardner, M.R.; Kattenhorn, L.M.; Kondur, H.R.; von Schaewen, M.; Dorfman, T.; Chiang, J.J.; Haworth, K.G.; Decker, J.M.; Alpert, M.D.; Bailey, C.C.; et al. AAV-expressed eCD4-Ig provides durable protection from multiple SHIV challenges. Nature 2015, 519, 87–91. [Google Scholar] [CrossRef]
Figure 1. Biomedical intervention strategies for HIV prevention and control. All biomedical intervention strategies are categorized based on the different stages of HIV exposure. The different colors represent distinct stages of HIV exposure, ordered from bottom to top: pre-exposure, post-exposure, and post-infection. The arrows indicate the duration of each stage. In general, with more earlier stages of virus exposure, more intervention measures are available and effective against HIV infections. TasP and U=U: treatment as prevention and undetectable = untransmittable; ART: antiretroviral therapy; HSCT: haematopoietic stem cell transplantation; PEP: post-exposure prophylaxis; PrEP: pre-exposure prophylaxis; VMMC: voluntary medical male circumcision.
Figure 1. Biomedical intervention strategies for HIV prevention and control. All biomedical intervention strategies are categorized based on the different stages of HIV exposure. The different colors represent distinct stages of HIV exposure, ordered from bottom to top: pre-exposure, post-exposure, and post-infection. The arrows indicate the duration of each stage. In general, with more earlier stages of virus exposure, more intervention measures are available and effective against HIV infections. TasP and U=U: treatment as prevention and undetectable = untransmittable; ART: antiretroviral therapy; HSCT: haematopoietic stem cell transplantation; PEP: post-exposure prophylaxis; PrEP: pre-exposure prophylaxis; VMMC: voluntary medical male circumcision.
Viruses 17 00756 g001
Figure 2. Challenges for the development of biomedical interventions against HIV infections. On the right half are the challenges posed by the characteristics of HIV itself, including high genetic variability, highly glycosylated antigens, and latent viral reservoirs. On the left half are the challenges created by the limitations of technologies, including the lack of animal infection models, and the correlates of protection, ethical, and biosafety issues. NHPs: non-human primates, DC: dendritic cell.
Figure 2. Challenges for the development of biomedical interventions against HIV infections. On the right half are the challenges posed by the characteristics of HIV itself, including high genetic variability, highly glycosylated antigens, and latent viral reservoirs. On the left half are the challenges created by the limitations of technologies, including the lack of animal infection models, and the correlates of protection, ethical, and biosafety issues. NHPs: non-human primates, DC: dendritic cell.
Viruses 17 00756 g002
Table 1. Summary of current biomedical interventions for HIV prevention and control.
Table 1. Summary of current biomedical interventions for HIV prevention and control.
CategoryApplicable PopulationProphylactic EfficacyAdvantageDisadvantage
Currently available biomedical interventionsPrEPOral PrEPUninfected people who are persistently at high risk of HIV infectionSexual transmission: 90–99% [78,79]
Drug injection transmission: 74% [80]
1. Broad range of application
2. High efficacy for HIV prevention
1. Highly affected by adherence, stigmatization issues
2. Long-term medication, drug side effects
Dapivirine ringUninfected women at high risk of HIV infectionVaginal sexual transmission: 56–63% [81,82]1. Female-led interventions
2. Long duration of protection
3. No need for oral medication, those females unable or unwilling to use oral medication can use this mode of prevention
4. Localized medication, reducing systemic side effects of medications
5. Strong concealment
1. Limited efficacy for HIV prevention
2. Limited to vaginal route of HIV transmission
3. Might cause local discomfort and inflammation
Injectable PrEPUninfected people who are persistently at high risk of HIV infectionCAB-LA: 69–88% [83,84]
Lenacapavir: 96–100% [85,86]
1. Long duration of protection
2. High efficacy for HIV prevention
1. Cost-related treatment barriers
2. Limited treatment availability
3. Drug resistance
4. LEVI syndrome
PEPUninfected people who have been exposed to HIV or accidentally exposedOccupational and nonoccupational exposure: 80–90% [87] (up to 99% for use within 2 h)1. Fast-acting, can be used for emergency prophylaxis
2. Short-term use and convenient
1. Highly affected by initiation time and adherence
2. Drug side effects
3. Not applicable to people with persistent high risk
TasP and U=UAll PLWHNearly 100% [88] (near elimination of risk of HIV transmission)1. High prevention rate
2. Applicable to all people living with HIV
3. Reduce HIV stigma
1. High compliance requirements
2. Long-term medication, drug side effects
Prevention of vertical transmissionHIV-positive women planning to become pregnant, pregnant, and breastfeeding99% [89] (risk of mother-to-child transmission reduced to <1%)1. High prevention rate
2. Improve the health of pregnant women and prevent newborns from being infected with HIV
1. Highly affected by adherence and stigmatization issues
2. Poor accessibility in underdeveloped regions where the need is greatest
VMMC #Adolescent and adult malesReduce the risk of heterosexual transmission of HIV infection by 60% [90,91,92]1. No need to use drugs
2. Long-term protection without dependence on adherence
3. Reduce the risk of other sexually transmitted infections
1. Limited preventive effect, need to be combined with other measures
2. Ineffective in MSM and female partners
3. Surgical risk
Potentially available biomedical interventions in the futureGene editing technology 1. Long-term protection without dependence on adherence
2. Removal of latent reservoirs for functional cure
1. Off-target risk
2. CCR5 receptor deficiency leads to susceptibility to other viral infections
Passive infusion of broadly neutralizing antibody 1. Highly effective and broad-spectrum
2. Long-term protection without dependence on adherence
3. Both preventive and therapeutic
1. High production and preservation costs
2. Unable to remove latent viral reservoirs
3. Repeated infusions can trigger anti-drug antibody (ADA) responses
PrEP: pre-exposure prophylaxis; PEP: post-exposure prophylaxis; TasP and U=U: treatment as prevention and undetectable = untransmittable; PLWH: people living with HIV; VMMC: voluntary medical male circumcision; MSM: men who have sex with men; CAB-LA: long-acting cabotegravir; LEVI: long-acting early viral inhibition. # The protective effect of voluntary medical male circumcision (VMMC) among men who have sex with men (MSM) remains uncertain. Biologically, VMMC would primarily benefit those engaging in exclusively insertive intercourse, given the reduced risk of HIV acquisition through the penile mucosa. While partial evidence suggests that VMMC may confer some protection for MSM—particularly those practicing insertive sex—current data are insufficient to support broad recommendations. Notably, the WHO does not presently endorse VMMC as an HIV prevention strategy for MSM.
Table 2. Summary of key Phase III PrEP randomized clinical trials.
Table 2. Summary of key Phase III PrEP randomized clinical trials.
TrialTimelineLocationPopulationSample SizeRegimenOutcome
iPrEx [78]2007–2010United States, Brazil, Ecuador, Peru, South Africa, ThailandMSM and transgender women2499Daily oral TDF/FTC36 infections in the TDF/FTC group, and 64 infections in the placebo group. Overall 44% reduction; up to 92% reduction with high adherence
TDF2 [102]2007–2010BotswanaHeterosexual men and women1219Daily oral TDF/FTC9 infections in the TDF-FTC group and 24 infections in the placebo group. 62% reduction
Bangkok Tenofovir [80]2005–2010Bangkok, ThailandPeople who use injection drugs2413Daily oral TDF17 infections in the TDF group (0.35 per 100 person-years) and 33 infections in the placebo group (0.68 per 100 person-years). 44% reduction
Partners PrEP [79]2008–2011Kenya, UgandaHeterosexual serodiscordant couples4758Daily oral TDF/FTC or TDF alone17 infections in the TDF group (0.65 per 100 person-years), 13 infections in the TDF/FTC group (0.50 per 100 person-years), and 52 in the placebo group (incidence, 1.99 per 100 person-years). 75% reduction with TDF/FTC; 67% reduction with TDF alone
PROUD [103]2012–2014United KingdomMSM544Daily oral TDF/FTC3 infections in the TDF/FTC group (1.2 per 100 person-years) and 20 infections in the placebo group (9.0 per 100 person-years). 86% reduction
IPERGAY [93]2012–2014France, CanadaMSM400On-demand TDF/FTC2 infections in the TDF-FTC group (0.91 per 100 person-years) and 14 infections in the placebo group (6.60 per 100 person-years). 86% reduction
DISCOVER [104]2016–2019United States, Canada, EuropeMSM and transgender women5387Daily oral TAF/FTC vs. TDF/FTC7 infections in the TAF/FTC group (0.16 infections per 100 person-years) and 15 infections in the TDF/FTC group (0.34 infections per 100 person-years). TAF/FTC was non-inferior to TDF/FTC
HPTN 083 [84]2016–2020United States, Latin America, Asia, AfricaMSM and transgender women4570Long-acting intramuscular cabotegravir (CAB-LA) every month vs. daily oral TDF/FTC13 infections in the CAB-LA group (0.41 per 100 person-years) and 39 in the TDF-FTC group (1.22 per 100 person-years). 66% more effective than daily TDF/FTC
HPTN 084 [83]2017–2021AfricaCisgender women3224Intramuscular CAB-LA every month vs. daily oral TDF/FTC4 infections in the CAB-LA group (0.2 per 100 person-years) and 36 infections in the TDF-FTC group (1.85 per 100 person-years). 89% reduction with CAB-LA compared to TDF/FTC
PURPOSE 1 [85]2021–2024South Africa, UgandaCisgender adolescent girls and young women5338subcutaneous lenacapavir every 6 months vs. daily oral F/TAF vs. daily oral F/TDF 0 infections in the lenacapavir group (0 per 100 person-years), 39 infections among in the F/TAF group (2.02 per 100 person-years), and 16 infections in the F/TDF group (1.69 per 100 person-years). Significantly lower than other two groups (100% efficacy in preventing HIV infections)
PURPOSE 2 [86]2021–2024United States, Argentina, Brazil, Mexico, Peru, Puerto Rico, South Africa, ThailandCisgender men, transgender women, transgender men, and gender-nonbinary persons3265subcutaneous lenacapavir every 6 months vs. daily oral F/TAF vs. daily oral F/TDF 2 infections in the lenacapavir group (0.10 per 100 person-years) and in 9 infections in the F/TDF group (0.93 per 100 person-years). Significantly lower than other two groups (reduced overall risk of infection by 96%)
TDF: tenofovir disoproxil; FTC: emtricitabine; TAF: tenofovir alafenamide; F/TAF: emtricitabine-tenofovir alafenamide; F/TDF: emtricitabine-tenofovir disoproxil.
Table 3. Summary of key PEP clinical trials.
Table 3. Summary of key PEP clinical trials.
TrialLocationExposureSample SizeRegimenProtection RatesAdverse Reaction Rate
Kahn et al. [129] (2001)United StatesNonoccupational401ZDV/3TC78%Nausea (52%), fatigue (44%), headache (24%), diarrhea (15%), and anorexia (12%)
Winston et al. [130] (2005)AustraliaNonoccupational385ZDV/3TC vs. ZDV/3TC/NFV vs. TDF/3TC/d4T75% vs. 68% vs. 85%Transaminase elevation (11% vs. 9% vs. 19%), diarrhea (6% vs. 51% vs. 25%), fatigue (39% vs. 32% vs. 30%), headache (17% vs. 12% vs. 1%), and nausea (81% vs. 42% vs. 23%)
Mayer et al. [131] (2008)United StatesNonoccupational371TDF/FTC vs. TDF/3TC vs. ZDV/3TC72% vs. 87% vs. 42%Diarrhea (47% vs. 31% vs. 10%), fatigue (30% vs. 28% vs. 39%), nausea (22% vs. 19% vs. 56%), headache (22% vs. 19% vs. 25%), and dizziness (20% vs. 16% vs. 5%)
Tosini et al. [132] (2010)FranceNonoccupational and occupational249TDF/FTC/LPV-r67%Diarrhea (80%), asthenia (66%), and abdominal pain (44%)
Diaz-Brito et al. [133] (2012)SpainNonoccupational200LPV-r vs. ATV64% vs. 64%Gastrointestinal (70% vs. 41%), neuropsychiatric (11% vs. 16%), asthenia (17% vs. 23%)
McAllister et al. [134] (2014)AustraliaNonoccupational120RAL/FTC/TDF vs. FTC/TDF92% vs. 91%Fatigue (37% vs. 26%), nausea (24% vs. 18%), abdominal cramps (21% vs. 12%), myalgias (9% vs. 0%)
Leal et al. [135] (2016)SpainNonoccupational243TDF/FTC/LPV-r vs. RAL66% vs. 80%Gastrointestinal (57% vs. 58%), neuropsychiatric (14% vs. 23%), and asthenia (18% vs. 18%)
Leal et al. [136] (2016)SpainNonoccupational237TDF/FTC/LPV-r vs. TDF/FTC/MVC56% vs. 68%Gastrointestinal (56% vs. 58%), neuropsychiatric (15% vs. 20%), and asthenia (19% vs. 18%)
Fatkenheuer et al. [137] (2016)GermanyNonoccupational and occupational 305DRV-r vs. LPV-r94% vs. 90%Diarrhea (30% vs. 52%), nausea (16% vs. 28%), fatigue (13% vs. 18%), sleep disorder(0% vs. 4%)
Valin et al. [138] (2016)FranceNonoccupational234FTC/TDF/ELV/COBI92%Fatigue (26%), nausea (25%), diarrhoea (17%), abdominal cramps (16%)
Milinkovic et al. [139] (2017)United KingdomNonoccupational213TDF/FTC/LPV-r vs. TDF/FTC/MVC65% vs. 71%Nausea or vomiting (39% vs. 30%, diarrhea (74% vs. 19%), fatigue (39% vs. 36%)
Chauveau et al. [140] (2019)FranceNonoccupational and occupational158TDF/FTC/RPV86%Fatigue (35%), nausea (22%), diarrhea (20%), abdominal cramps (16%), headache (11%)
Nie et al. [141] (2021)ChinaNonoccupational and occupational297ABT/DTG vs. ABT/TDF/3TC vs. DTG/TDF/3TC64% vs. 64% vs. 64%Dizziness (7% vs. 7% vs. 7%), diarrhea (8% vs. 6% vs. 2%), asthenia (5% vs. 4% vs. 5%), and triglycerides increase (4% vs. 2% vs. 7%)
Liu et al. [142] (2022)ChinaNonoccupational108BIC/FTC/TAF96%Creatinine elevation (4%), headache (2%), diarrhea (2%), and nausea (1%)
Lacombe et al. (2024)FranceNonoccupational226DOR
ZDV: zidovudine; 3TC: lamivudine; NFV: nelfinavir; TDF: tenofovir disoproxil; d4T: stavudine; FTC: emtricitabine; LPV-r: ritonavir-lopinavir; ATV: atazanavir; RAL: raltegravir; MVC: maraviroc; DRV-r: ritonavir-boosted darunavir; ELV: cobicistat-boosted elvitegravir; COBI: cobicistat; RPV: rilpivirine; ABT: lamivudine; DTG: dolutegravir; BIC: bictegravir; TAF: tenofovir alafenamide; DOR: doravirine.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Liao, Y.; Wen, Z.; Shi, M.; Zou, H.; Sun, C. Biomedical Interventions for HIV Prevention and Control: Beyond Vaccination. Viruses 2025, 17, 756. https://doi.org/10.3390/v17060756

AMA Style

Liao Y, Wen Z, Shi M, Zou H, Sun C. Biomedical Interventions for HIV Prevention and Control: Beyond Vaccination. Viruses. 2025; 17(6):756. https://doi.org/10.3390/v17060756

Chicago/Turabian Style

Liao, Yu, Ziyu Wen, Minjuan Shi, Huachun Zou, and Caijun Sun. 2025. "Biomedical Interventions for HIV Prevention and Control: Beyond Vaccination" Viruses 17, no. 6: 756. https://doi.org/10.3390/v17060756

APA Style

Liao, Y., Wen, Z., Shi, M., Zou, H., & Sun, C. (2025). Biomedical Interventions for HIV Prevention and Control: Beyond Vaccination. Viruses, 17(6), 756. https://doi.org/10.3390/v17060756

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop