Fact-checked by Grok 2 weeks ago

Pre-exposure prophylaxis

Pre-exposure prophylaxis () is a preventive regimen involving the administration of antiretroviral medications, such as combinations of tenofovir and emtricitabine, to -seronegative individuals at high risk of acquisition via sexual contact or injection drug use, thereby inhibiting and substantially lowering infection probability upon exposure. First demonstrated effective in the 2010 iPrEx , which enrolled over 2,400 participants and showed a 44% overall risk reduction rising to 92% among those with detectable drug levels indicating adherence, received U.S. approval for daily oral use in 2012. Systematic reviews of clinical trials confirm PrEP's efficacy exceeds 99% against acquisition in adherent users, with reductions of 54% or greater across diverse populations when taken as prescribed, though protection wanes rapidly with missed doses due to the drugs' short requiring sustained plasma concentrations. Long-acting injectable formulations, such as approved in 2021, offer alternatives to daily pills for improved convenience, demonstrating comparable or superior efficacy in trials like HPTN 083 and HPTN 084. Effective deployment necessitates quarterly testing, renal function monitoring, and counseling to ensure ongoing seronegativity and avert selection for drug-resistant strains if undetected infection occurs during use. Real-world adherence challenges undermine 's potential, with meta-analyses indicating suboptimal pill-taking in 38% of users and discontinuation in 41% within six months, correlating with higher incidence in observational cohorts compared to settings. profiles reveal mostly mild, transient side effects including gastrointestinal upset, , and , affecting up to 29% initially but rarely leading to cessation; however, rare risks like reduced or renal impairment necessitate baseline assessments, particularly in long-term users. While has contributed to incidence declines in high-prevalence groups such as men who have sex with men, broader epidemiological impact remains constrained by access inequities, cost barriers in low-resource settings, and dependence on behavioral factors like consistent use amid competing prevention modalities.

Definition and Principles

Conceptual Framework

Pre-exposure prophylaxis (PrEP) is a preventive strategy involving the administration of antimicrobial drugs or vaccines to uninfected individuals before potential exposure to a , aimed at reducing the likelihood of by interfering with pathogen entry, replication, or establishment. This approach targets high-risk populations in contexts of ongoing or predictable exposure, such as endemic regions or behavioral risk factors, by creating a prophylactic barrier that aborts early dynamics. The core principles center on pharmacokinetic and pharmacodynamic optimization to maintain agent concentrations at relevant anatomical sites—such as mucosal tissues—that exceed the pathogen's minimum inhibitory levels during the exposure window. For pharmacological implementations, this requires consistent adherence to sustain drug accumulation in target cells, capitalizing on properties like prolonged intracellular half-lives to cover intermittent exposures without daily peaks and troughs leading to vulnerability. Adherence thus forms a causal , as suboptimal levels permit infections, underscoring the framework's dependence on user alongside agent safety for prolonged administration. PrEP differs fundamentally from (PEP), which entails reactive initiation of agents within 72 hours of suspected exposure to halt nascent infection, whereas PrEP operates preemptively for anticipated risks, enabling chronic use in scenarios where exposures are unpredictable yet recurrent. This proactive timing aligns PrEP with other chemoprophylactic paradigms, such as antimalarial regimens during travel to endemic areas, prioritizing sustained protection over acute intervention. The framework's biological rationale posits that preemptive agent presence disrupts founder populations at initial replication foci, preventing dissemination and , a mechanism validated across pathogens via tissue-level modeling and analogous preventives like pre-exposure , which primes responses absent in purely pharmacological modalities. Integration with non-biomedical measures, such as barrier methods, enhances overall risk reduction without substituting for behavioral modifications.

Pharmacological and Biological Mechanisms

Pre-exposure prophylaxis relies on pharmacological agents that achieve sufficient concentrations in target tissues to inhibit pathogen replication during the initial stages of infection following exposure. For HIV, the primary regimen combines tenofovir disoproxil fumarate (TDF) and emtricitabine (FTC), both nucleoside reverse transcriptase inhibitors (NRTIs). TDF is metabolized intracellularly to tenofovir diphosphate, an adenosine nucleotide analog that competitively inhibits HIV-1 reverse transcriptase and causes premature termination of viral DNA chain elongation, preventing proviral DNA integration into the host genome. Emtricitabine undergoes phosphorylation to its triphosphate form, which similarly incorporates into nascent viral DNA, leading to chain termination due to its 3'-hydroxyl group deficiency. These mechanisms target the eclipse phase of HIV infection, where low viral inoculum requires high drug levels in mucosal tissues and lymphoid cells to block systemic dissemination. In malaria prophylaxis, drugs like atovaquone-proguanil target parasites at pre-erythrocytic and erythrocytic stages. Atovaquone collapses the parasite's mitochondrial membrane potential by inhibiting in the , halting ATP production essential for parasite survival. Proguanil metabolizes to cycloguanil, which inhibits , disrupting synthesis and production in the parasite. , used for liver-stage targeting, generates that damage parasite mitochondria and induce in hepatic schizonts, preventing exoerythrocytic replication. These actions ensure causal prophylaxis by eradicating parasites before symptomatic blood-stage infection. Biological mechanisms in vaccine-based PrEP, such as for , involve inducing adaptive immunity prior to exposure. Inactivated or recombinant vaccines stimulate B-cell activation and differentiation, producing virus-neutralizing antibodies (nAbs) against the glycoprotein, which bind the and prevent neuronal entry and retrograde axonal transport. This pre-existing humoral response, typically achieving titers above 0.5 IU/mL, facilitates rapid control of viral replication upon exposure, reducing the need for full post-exposure regimens. Cellular immunity, including + T-cell help for antibody affinity maturation and + cytotoxic responses, provides ancillary protection but is secondary to nAbs in prophylaxis efficacy.

Historical Development

Origins and Early Research

The concept of pre-exposure prophylaxis originated in the late with the developed by in 1885, initially for post-exposure use but soon adapted for prevention in high-risk individuals such as laboratory personnel. Adrien Loir, Pasteur's nephew, and Eugène Viala became among the first recipients of pre-exposure rabies vaccination following incidental contact with attenuated virus during vaccine preparation in the mid-1880s, demonstrating early recognition of prophylactic vaccination's potential to avert infection before confirmed exposure. For malaria, chemoprophylaxis traces to the , when Jesuit missionaries in used bark infusions to prevent fever among European colonizers in endemic areas, with —isolated from the bark in —becoming the cornerstone of suppressive regimens by the mid-19th century for travelers and . This approach relied on continuous dosing to inhibit parasite development in the blood, influencing later synthetic antimalarials like in the 1940s, though resistance and toxicity concerns emerged over time. Modern pharmacological PrEP for HIV emerged from 1980s explorations of antiretrovirals like (AZT) for prophylaxis, but pivotal early evidence came from nonhuman primate models in the 1990s. In a 1995 study, Tsai et al. administered (R)-9-(2-phosphonylmethoxypropyl) (PMPA, a prodrug) subcutaneously to before and after (SIV) challenge, achieving complete prevention of infection in treated animals, thus validating pre-exposure antiretroviral efficacy against lentiviral acquisition. Subsequent macaque studies in the early tested combinations like tenofovir-emtricitabine, confirming dose-dependent protection against mucosal SHIV transmission and informing human trial designs.

Pivotal Clinical Trials and Regulatory Approvals

The iPrEx trial, published in 2010, was the first randomized, double-blind, placebo-controlled study to demonstrate the efficacy of daily oral (/TDF, marketed as Truvada) for pre-exposure prophylaxis () in men who have sex with men (MSM) and women at high risk. Conducted across sites in , , , , , and the with 2,499 participants, it showed a 44% in acquisition overall, rising to 92% among participants with detectable drug levels indicating adherence. Subsequent trials confirmed and expanded these findings in heterosexual populations. The Partners PrEP study, reported in 2011, involved 4,758 heterosexual couples in and and found daily /TDF reduced HIV incidence by 75% compared to placebo, with tenofovir monotherapy showing similar efficacy. The TDF2 trial, published in 2012, tested daily /TDF in 1,219 heterosexual men and women in , yielding a 62% risk reduction, though early termination due to funding limited power. These multinational trials collectively established /TDF's prophylactic efficacy across diverse risk groups, with tied to adherence and plasma drug detection. Regulatory approvals followed swiftly. The U.S. (FDA) approved Truvada for on July 16, 2012, based primarily on iPrEx and Partners PrEP data, expanding its prior 2004 indication for treatment. The World Health Organization (WHO) issued conditional guidelines endorsing oral in 2015 for populations with substantial incidence, prioritizing MSM, couples, and others, following review of trial evidence. For non- applications, rabies pre-exposure prophylaxis relies on established inactivated vaccines like human diploid cell vaccine (approved by FDA in 1981), administered in standard regimens without recent pivotal trials altering core approvals, while malaria chemoprophylaxis uses antimalarials such as atovaquone-proguanil, approved in 2000, supported by efficacy data from field studies rather than large-scale -specific trials.

Applications by Disease

HIV Prevention

Pre-exposure prophylaxis (PrEP) for HIV consists of antiretroviral medications administered to HIV-seronegative individuals at elevated risk of acquisition through sexual contact or injection drug use. The primary oral regimen involves daily dosing of disoproxil fumarate (FTC/TDF, branded as Truvada), approved by the U.S. (FDA) in July 2012 for adults and adolescents weighing at least 35 kg. An alternative daily oral option, alafenamide (FTC/TAF, branded as Descovy), received FDA approval in October 2019 for cisgender men and transgender women who have sex with men, but not for cisgender women due to insufficient efficacy data for receptive vaginal sex. Target populations for PrEP include men who have sex with men (MSM) engaging in condomless , heterosexual individuals in partnerships, people who inject s (PWID) sharing equipment, and others with recent bacterial sexually transmitted infections or multiple partners. The Centers for Disease Control and Prevention (CDC) recommends PrEP for sexually active adults and adolescents meeting criteria such as inconsistent condom use, an HIV-positive sexual partner, or injection drug use within the six months. For MSM, event-driven dosing—known as the 2-1-1 regimen—involves two tablets 2–24 hours before sex, followed by one daily for two days—has demonstrated effectiveness in clinical settings, though it is not FDA-approved in the U.S. and requires confirmed adherence for protection. Long-acting injectable formulations address adherence challenges associated with daily pills. Cabotegravir long-acting (CAB-LA, branded as Apretude), administered as intramuscular injections every two months after initial loading doses, was FDA-approved in December 2021 for adults and adolescents at risk via sexual exposure, showing superior efficacy to daily oral in trials among MSM and transgender women. In June 2025, the FDA approved (branded as Yeztugo), a inhibitor given subcutaneously every six months, as the first biannual option, based on phase 3 trials demonstrating near-complete prevention of HIV acquisition when administered as directed. The (WHO) endorses both oral and long-acting options for at-risk populations globally, emphasizing integration with HIV testing every three months, renal function monitoring for tenofovir-based regimens, and counseling on adherence to mitigate resistance risks. Implementation requires baseline HIV testing, ongoing serostatus verification to prevent inadvertent of undiagnosed , and linkage to sexual services. Real-world application reveals disparities, with higher uptake among MSM than among heterosexual women or PWID, partly due to barriers and adherence variability; studies indicate that suboptimal pill-taking reduces protective levels, underscoring the need for tailored behavioral support.

Malaria Prevention

Pre-exposure prophylaxis for malaria, also termed chemoprophylaxis, entails the preventive use of antimalarial medications before and during travel to endemic areas to suppress infection from bites. The U.S. Centers for and Prevention (CDC) recommends four primary options—atovaquone-proguanil, , , and tafenoquine—tailored to factors such as resistance prevalence, trip length, and patient contraindications, with no regimen offering complete protection, necessitating concurrent bite prevention measures like insecticide-treated nets and repellents. Atovaquone-proguanil (Malarone) is administered daily, starting 1–2 days before entry into malarious areas, continuing through exposure, and for 7 days thereafter; adult dosing is one 250 mg atovaquone/100 mg proguanil tablet daily. Clinical trials report protective efficacies of 96% (95% CI: 72%–99%) against P. falciparum and 84% (95% CI: 44%–95%) against P. vivax, with meta-analyses confirming superior tolerability over alternatives like mefloquine or chloroquine-proguanil. Doxycycline, a tetracycline antibiotic, is taken daily at 100 mg starting 1–2 days pre-travel, during exposure, and for 4 weeks post-return due to its prolonged tissue persistence. It achieves over 95% efficacy against P. falciparum in randomized studies, comparable to mefloquine, though contraindicated in pregnant women and children under 8 years owing to risks of fetal bone/teeth effects and photosensitivity. Mefloquine is dosed weekly at 250 mg (adult) beginning 2 weeks before travel, weekly during, and for 4 weeks after, but its use has declined due to neuropsychiatric adverse events and resistance in and parts of the ; efficacy exceeds 95% against sensitive strains in non-resistant zones. Tafenoquine (Arakoda), approved by the FDA in 2018 for adults 18 years and older, requires G6PD testing to avoid in deficient individuals; it involves a 200 mg daily for 3 days pre-travel, 200 mg weekly during, and 200 mg one week post-exposure. Phase 3 trials demonstrate non-inferior efficacy to atovaquone-proguanil (98.6% vs. 100% protection against P. falciparum over 3 months), with meta-analyses affirming 200 mg weekly dosing as effective and well-tolerated for up to 6 months.
DrugAdult Dosing RegimenReported Efficacy (P. falciparum)Key Considerations
Atovaquone-proguanil1 tablet (250/100 mg) daily; start 1–2 days pre, 7 days post>95%Pregnancy category C; well-tolerated
100 mg daily; start 1–2 days pre, 4 weeks post>95%Contraindicated <8 years/pregnant; photosensitivity
Mefloquine250 mg weekly; start 2 weeks pre, 4 weeks post>95% (sensitive strains)Neuropsychiatric risks; resistance in some areas
Tafenoquine200 mg daily x3 (loading), then weekly; 200 mg post; G6PD test required98.6%Adults only; risk if G6PD deficient
Regimen choice must account for local resistance—e.g., avoiding in areas with known failures—and no prophylaxis fully eradicates liver-stage hypnozoites in P. vivax or P. ovale, potentially requiring antirelapse post-travel.

Rabies Prevention

Pre-exposure prophylaxis (PrEP) for consists of administered prior to potential exposure to the to induce immunity and simplify post-exposure management. It is recommended by the Centers for Disease Control and Prevention (CDC) for individuals at elevated risk, including veterinarians, animal handlers, laboratory workers handling , and travelers to regions with endemic where access to (PEP) may be limited. The (WHO) similarly advises PrEP for those with continual, frequent, or increased exposure risk due to occupation, residence, or travel. The current CDC regimen for immunocompetent adults aged 18 years and older involves two 1 mL intramuscular doses of rabies vaccine (human diploid cell vaccine or purified chick embryo cell vaccine) on days 0 and 7, administered in the deltoid muscle. This two-dose schedule, updated in 2022, replaced the prior three-dose series (days 0, 7, and 21–28) and provides seroprotection for up to three years in low-risk categories without boosters. For ongoing high-risk exposure, periodic serologic testing (rabies virus neutralizing antibody titer ≥0.5 IU/mL) is advised every six months to two years, with boosters as needed to maintain immunity. In children under 2 years or small children, the anterolateral thigh is preferred over the deltoid for injection. Efficacy data from clinical studies demonstrate that rabies PrEP induces robust antibody responses, with seroconversion rates exceeding 95% after the two-dose regimen, comparable to the three-dose series. A confirmed PrEP's immunogenicity across age groups, including co-administration with routine , and its role in averting rabies deaths by enabling abbreviated PEP (typically two booster doses instead of full five-dose PEP plus rabies immune globulin). In real-world scenarios, PrEP has prevented rabies in exposed individuals who delayed or incompletely received PEP, though no direct comparative trials exist due to ethical constraints on withholding in high-risk cohorts. For short-term risks (≤3 years), no routine boosters are required post-series, reducing logistical burdens.

Other Potential Applications

Tenofovir-based regimens used for have demonstrated substantial prophylactic effects against (HBV) acquisition, particularly among men who have sex with men (MSM) and other high-risk groups such as injection drug users. In a 2023 of MSM, tenofovir disoproxil fumarate/emtricitabine significantly reduced HBV incidence compared to non-users, with hazard ratios indicating near-complete protection during consistent adherence. This effect stems from tenofovir's potent inhibition of HBV , outperforming alone in populations with suboptimal immune responses or ongoing exposure risks. However, challenges include monitoring for HBV reactivation upon discontinuation and the need for long-acting formulations to improve adherence in immunodeficient individuals. For hepatitis C virus (HCV), direct-acting antivirals like sofosbuvir have been proposed as PrEP candidates for high-risk groups such as injection drug users and MSM, where reinfection rates remain elevated post-cure due to lack of sterilizing immunity. Exploratory applications include preventing HCV transmission in organ transplant recipients from viremic donors, leveraging short-course regimens to bridge high-exposure periods. Yet, economic barriers, potential for resistance emergence, and absence of large-scale trials limit widespread adoption, with current evidence confined to modeling and small observational data. Among retroviruses, strategies show promise for HIV-2 prevention in endemic West African regions, where tenofovir and integrase inhibitors like exhibit activity against its lower-transmissibility profile compared to HIV-1. For human T-lymphotropic virus type 1 (HTLV-1), demonstrates inhibition, but no clinical trials have evaluated efficacy, underscoring the need for further research amid absent vaccines or approved antivirals. HTLV-1 screening in blood products and high-risk populations remains the primary control measure. Respiratory viruses represent another frontier, with reducing influenza infection risk by 86% in a phase 3 trial of household contacts, positioning it as a viable option during outbreaks. For , has been investigated primarily for , with limited data hampered by rapid viral evolution and resistance concerns; no regulatory approvals exist as of 2022. Broader application to viruses like () lacks dedicated trials, though monoclonal antibodies serve prophylactic roles in vulnerable infants. Overall, these applications remain investigational, constrained by adherence issues, cost, and the imperative for virus-specific adaptations beyond paradigms.

Efficacy Evidence

Clinical Trial Data

The iPrEx , a randomized, double-blind, placebo-controlled study published in 2010, enrolled 2,499 men who have sex with men and transgender women at high risk for acquisition across , , , the , , and . Participants received daily oral emtricitabine-tenofovir disoproxil fumarate (FTC-TDF) or placebo; overall, FTC-TDF reduced incidence by 44% (95% CI, 15 to 63; 36 infections in placebo arm vs. 20 in FTC-TDF arm), with efficacy rising to 92% (95% CI, 40 to 99) among those with detectable plasma tenofovir levels indicating adherence. In the iPrEx open-label extension, overall effectiveness was 50%, but reached 92% for participants taking four or more doses per week. The PROUD trial, an open-label randomized controlled study reported in 2015, involved 544 high-risk men who have sex with men in , comparing immediate versus deferred initiation of daily oral FTC-TDF. Immediate reduced incidence by 86% (95% CI, 43 to 96; one infection in immediate arm vs. nine in deferred arm over ), demonstrating high real-world when integrated into sexual health clinics with supportive counseling. Pivotal trials for heterosexual populations included Partners PrEP (2011), a randomized placebo-controlled study of 4,758 serodiscordant heterosexual couples in and , where daily FTC-TDF yielded a 75% reduction in acquisition among HIV-negative partners (95% , 55 to 87), contingent on adherence. A 2019 meta-analysis of 11 randomized trials (n=18,172) across diverse populations confirmed oral 's relative risk reduction of 0.46 (95% , 0.33 to 0.64) versus or no PrEP over 4 months to 4 years, with efficacy strongly correlated to drug detection in or . Long-acting cabotegravir trials, such as HPTN 083 (2020) and HPTN 084 (2021), evaluated injectable every two months versus daily oral FTC-TDF in men who have sex with men/ women and women, respectively; both showed cabotegravir superiority, with incidence reductions of 66-89% relative to oral , highlighting adherence advantages for long-acting formulations. For , clinical trials of antimalarial chemoprophylaxis (sometimes termed ) focus on regimens like daily atovaquone-proguanil or , but efficacy data emphasize intermittent preventive treatment; a 2022 trial of dihydroartemisinin-piperaquine in pregnant women with showed 80-90% against placental , though broader PrEP applications remain limited by resistance and adherence challenges. Rabies pre-exposure prophylaxis trials primarily assess immunogenicity rather than endpoints, given rarity; a 2023 study of single-visit intradermal regimens achieved rates comparable to standard three-dose intramuscular schedules (≥0.5 IU/mL in >99% of participants), supporting simplified protocols for travelers and high-risk workers without compromising boostability.
TrialYearPopulationInterventionEfficacy (HIV Incidence Reduction)Key Notes
iPrEx2010MSM/TG (n=2,499)Daily FTC-TDF vs. 44% overall; 92% with adherenceAdherence-dependent; global sites
PROUD2015MSM (n=544)Immediate vs. deferred FTC-TDF86%Open-label; clinics
Partners PrEP2011Heterosexual (n=4,758)Daily FTC-TDF vs. 75%Adherence key;
HPTN 083/0842020/2021MSM/TG and women LA vs. FTC-TDF66-89% relative to oralLong-acting; superior adherence

Real-World Implementation Outcomes

Real-world studies of oral HIV PrEP, primarily emtricitabine/tenofovir disoproxil fumarate (F/TDF), have demonstrated effectiveness reductions of HIV incidence ranging from 60% overall among high-risk men who have sex with men (MSM) to 93% among those with high consumption levels, compared to near-100% efficacy in randomized controlled trials where adherence was rigorously monitored.00106-2/fulltext) This disparity arises primarily from suboptimal adherence in routine care, with many users exhibiting infrequent or inconsistent dosing that diminishes protective drug levels.00106-2/fulltext) Population-based cohort analyses, such as one from , , covering 2017–2022, reported zero HIV seroconversions among adherent PrEP users, affirming preventive efficacy under real-world conditions despite variable adherence patterns. In diverse U.S. settings, implementation data link higher coverage to measurable declines in new diagnoses; states with the top quartile of PrEP users per capita from 2018–2022 saw a reduction in incidence relative to lower-coverage states, correlating with post-regulatory approvals. Australian government-subsidized programs yielded a 78.5% incidence (0.56 per 1000 person-years) among users achieving 60% or greater days covered by PrEP from 2018–2022, using non-PrEP high-risk cohorts as comparators, though overall program impact was tempered by discontinuation rates exceeding 50% within one year. For women, real-world F/TDF PrEP mirrored , with adjusted ratios indicating substantial in cohorts followed through , contingent on consistent use. Long-acting injectable PrEP options, such as (CAB-LA), have shown superior real-world outcomes in early ; U.S. cohort studies from 2021–2024 reported over 99% in preventing acquisition among MSM and women, attributed to biannual dosing that mitigates daily adherence challenges. However, global uptake remains limited, with only 1.3 million people accessing in 2022—far below the 10 million high-risk individuals targeted—resulting in minimal acceleration of decline in low- and middle-income countries. Disparities persist, with lower in subgroups facing barriers, such as rural populations or those without routine , underscoring gaps beyond pharmacological efficacy.

Safety and Adverse Effects

Common Side Effects

Common side effects of disoproxil fumarate (Truvada), the original formulation approved for , primarily involve gastrointestinal disturbances such as , , , and , occurring in approximately 5-19% of users in clinical trials, though rates were often comparable to groups (e.g., 3.8% for / versus 2.6% ). and decreased weight or appetite are also frequently reported, affecting 3-7% of participants, with these symptoms typically mild, transient, and resolving within the first month of use. For emtricitabine/tenofovir alafenamide (Descovy), a later with reduced renal and toxicity, common side effects mirror those of Truvada but with potentially lower incidence due to the lower tenofovir dose; these include (up to 5%), , , , and or discomfort, reported in ≥2% of trial participants and generally self-limiting. Real-world data from open-label extensions of trials like iPrEx confirm that gastrointestinal issues and prompt discontinuation in less than 1% of adherent users, underscoring their rarity as barriers to continued use. These effects are dose-dependent and more pronounced during , often mitigated by taking the medication with ; reveals no excess serious adverse events beyond in large-scale studies involving over 5,000 participants. While sources like CDC guidelines emphasize their tolerability, some peer-reviewed analyses note underreporting in trials due to short follow-up, though long-term data affirm low persistence of mild symptoms.

Long-Term Risks

Long-term use of tenofovir disoproxil fumarate/emtricitabine (TDF/FTC), the primary regimen for pre-exposure prophylaxis (), has been associated with modest declines in renal function and bone mineral density (BMD), though serious adverse events remain rare across large-scale trials. A of 13 randomized controlled trials involving 15,678 participants found no significant increase in grade 3 or higher elevations ( 0%, 95% 0%–0%) or fractures (risk difference 0%, 95% CI 0%–1%), with only a borderline increase in all-grade elevations ( 0.02, 95% 0.00–0.03). These effects are typically reversible upon discontinuation, with BMD losses of approximately 1.2% at the and 0.5% at the in highly adherent users, often recovering partially or fully after stopping . Renal risks are higher among individuals with predisposing factors such as age over 40–50 years, preexisting , or concurrent use of nephrotoxic drugs, with real-world data indicating few cases of clinically significant impairment during extended use.00004-2/fulltext) A 2022 analysis of over 5 years of daily and event-driven reported stable kidney function in most users, supporting routine monitoring of clearance every 3–6 months to mitigate potential . Bone density reductions are similarly influenced by adherence levels and baseline risk factors like low body weight or , but meta-analyses have not identified clinically meaningful fracture increases or progression in cohorts. No robust evidence links long-term TDF/FTC PrEP to elevated risks of , cardiovascular events, or hepatic dysfunction beyond acute gastrointestinal effects, with serious rates comparable to in extended follow-up. Newer formulations, such as /emtricitabine (TAF/FTC), demonstrate reduced renal and BMD impacts due to lower plasma tenofovir exposure, offering alternatives for those with risk factors while maintaining efficacy.00071-0/abstract) Overall, empirical data affirm that these risks are outweighed by prevention benefits in high-risk populations, provided monitoring protocols are followed. For non- PrEP applications, such as for bacterial sexually transmitted infections, long-term antimicrobial use raises concerns for disruption and , though data remain limited to shorter durations.

Adherence and Behavioral Dynamics

Factors Influencing Adherence

Adherence to pre-exposure prophylaxis () for prevention varies widely, with systematic reviews indicating that protective drug levels require at least 80% adherence for near-complete efficacy, as demonstrated in trials like Partners PrEP where high adherence correlated with 100% prevention (95% CI 83.7-100%). Factors influencing adherence span individual, interpersonal, and structural domains, often interacting to undermine consistent daily dosing of oral tenofovir disoproxil fumarate-emtricitabine. Individual factors include low perceived HIV risk, reported by over 70% of women in the FEM-PrEP trial, which diminishes motivation for regimen maintenance despite objective risk. Early side effects such as , , and , occurring in the first month of use, prompt discontinuation in multiple studies, while forgetting doses emerges as the most frequent self-reported cause of suboptimal adherence across diverse populations including men who have sex with men (MSM) and heterosexuals. Younger age, particularly among adolescents and young adults, correlates with lower adherence proportions in meta-analyses, compounded by substance use and inconsistent daily routines. Interpersonal and social factors prominently feature , which drives self-stigmatization and nondisclosure of use, as evidenced in reviews spanning MSM, individuals, and women. Limited decision-making autonomy, especially in relationships or communities with conservative norms, reduces adherence among young women and sex workers, where partner dynamics or family can either reinforce (e.g., supportive living arrangements) or hinder compliance. elements like medical mistrust and further impede uptake and persistence in key populations such as . Structural and systemic factors involve costs, which deter sustained use absent subsidies, alongside barriers to healthcare including provider and proximity. Logistical challenges of daily adherence, such as pill burden and integration into routines, exacerbate non-compliance, particularly in low-resource settings with weak policy support. Behavioral risks like condomless sex or multiple partners can motivate initial uptake but falter without enabling factors such as or pill packaging aids, which studies link to improved retention among female sex workers. Overall, interventions targeting these multilevel determinants, including counseling and mobile reminders, have boosted adherence in 67% of evaluated studies.

Risk Compensation and Behavioral Disinhibition

Risk compensation, also known as behavioral disinhibition, refers to the phenomenon where individuals perceive reduced personal risk due to protective interventions like and subsequently increase high-risk behaviors, such as condomless sex or multiple partners, potentially undermining overall preventive benefits or elevating other health risks. In the context of , this has been hypothesized to occur among men who have sex with men (MSM), the primary population studied, where high efficacy against might lead to decreased condom use, thereby raising (STI) incidence despite protection. Randomized controlled trials (RCTs) of daily oral generally reported limited evidence of . A narrative synthesis across multiple RCTs found no between-group differences in use, with some studies showing stability or even increases in safer practices, and no rises in sexual partners or rates attributable to . For instance, early trials like iPrEx observed no significant uptick in condomless sex during blinded phases. However, open-label extensions and real-world demonstration projects revealed more pronounced behavioral shifts. A of 13 open-label studies involving 5008 MSM users documented increases in condomless sex in most cohorts, including a decline in use scores from 2.0 to 1.5 for regular partners and 3.1 to 2.4 for casual ones in the VicPrEP over 12 months. Similarly, the PRELUDE reported condomless sex with HIV-positive or unknown-status partners rising from 80.0% to 91.1% at 12 months. STI data further substantiate disinhibition in post-trial settings. The same of eight studies (n=4388) found elevated STI odds, with rectal incidence at OR 1.59 (95% CI 1.19–2.13) and any STI at OR 1.24 (95% CI 0.99–1.54) among users; VicPrEP specifically noted STI rates climbing from 43.2 to 119.8 per 100 person-years. A U.S. (n=112) confirmed significant rises in condomless receptive acts (mean increase 0.49–0.87, p<0.05 across visits), though STI rates did not differ significantly from non-PrEP comparators at 24 weeks (17.22% vs. 12.5%, p=0.375). These patterns were more evident in later studies (post-2016 OR 1.47, 95% CI 1.05–2.05), suggesting growing as PrEP awareness and access expanded. Critics argue that observed behavioral changes do not equate to net harm, as PrEP's high HIV efficacy (e.g., >99% with adherence) prevents despite disinhibition, and qualitative data indicate shifts toward rather than deliberate risk-seeking. One posits no empirical link between PrEP-related condomless sex and elevated HIV incidence, framing disinhibition concerns as overstated from theoretical models ill-suited to HIV dynamics. Nonetheless, causal evidence from real-world implementations underscores increased STI burdens, particularly bacterial infections treatable but recurrent, necessitating integrated screening and counseling to mitigate broader epidemiological impacts. appears most pronounced among already high-risk MSM subgroups, supporting targeted rather than universal PrEP deployment with behavioral monitoring.

Controversies and Criticisms

Development of Drug Resistance

Drug resistance in the context of HIV pre-exposure prophylaxis (PrEP) refers to the emergence of HIV variants with mutations that confer reduced susceptibility to the inhibitors (NRTIs) typically used in PrEP regimens, such as tenofovir disoproxil fumarate (TDF) and emtricitabine (FTC). These mutations arise primarily during breakthrough infections, where HIV acquisition occurs despite PrEP use, often due to suboptimal drug levels from poor adherence or initiation of PrEP during undiagnosed acute HIV infection. In such cases, incomplete viral suppression allows selective pressure favoring resistant strains, potentially compromising future antiretroviral therapy (ART) options, though alternative drug classes like integrase inhibitors remain effective. Key resistance mutations associated with TDF include K65R in the gene, which reduces tenofovir incorporation efficiency, while FTC resistance is commonly linked to M184V/I that alter binding. In clinical trials of daily oral F/TDF , such as those pooled in a 2024 meta-analysis of 72 studies, breakthrough infections were rare (incidence <1 per 1000 person-years with high adherence), and (DRMs) were detected in only a subset of seroconverters, with no significant increase in DRM risk compared to arms. However, among users who seroconverted, approximately 6% developed TDF and/or resistance if not acutely infected at initiation, escalating to 61% when was started during acute HIV infection due to high viral loads overwhelming partial protection. Real-world data indicate higher DRM prevalence in certain cohorts, such as a 2025 cross-sectional study in Kenya finding 22% of newly diagnosed HIV-positive individuals with prior PrEP exposure harboring TDF/FTC-associated mutations, attributed to delayed HIV testing and adherence lapses. Breakthrough infections remain infrequent with optimal adherence (e.g., <0.3% in cabotegravir PrEP trials), but poor adherence creates windows for infection with wild-type or pre-existing resistant strains, amplifying transmission risks. Mathematical models project that widespread PrEP rollout could elevate population-level resistance to 8.2% under combined ART/PrEP scenarios, though net HIV incidence reductions (up to one-third) may offset this by limiting overall viral spread. Mitigation strategies emphasize routine HIV testing before and during PrEP initiation to exclude acute , alongside adherence monitoring via or self-report, as resistant breakthrough cases can delay viral suppression in subsequent . Population for transmitted DRMs is recommended, particularly in high-PrEP uptake settings, to track any shifts, though shows limited clinical impact on outcomes to date due to PrEP's overall in averting .

Increases in Sexually Transmitted Infections

Observational studies have documented elevated rates of bacterial sexually transmitted infections (STIs) among individuals using pre-exposure prophylaxis (), particularly men who have sex with men (MSM). A of real-world data found that PrEP use was associated with a 24% increase in bacterial STI incidence, attributed in part to behavioral factors such as reduced use. In , over 50% of PrEP initiators were diagnosed with an STI within 30 days of starting the regimen, highlighting rapid post-initiation vulnerabilities. Evidence of —wherein perceived protection leads to increased sexual risk-taking—contributes to these trends. Prospective cohort analyses indicate that users often report more anal sex partners and lower condom adherence compared to pre- periods, correlating with higher , , and diagnoses. For instance, in a Danish of MSM, STI rates rose after PrEP initiation despite stable overall prevention efficacy, with self-reported increases in partner numbers and condomless encounters. While some randomized trials report stable or reduced STI rates under controlled conditions, real-world implementation reveals divergent outcomes, with PrEP cohorts experiencing STI incidences up to 90 per 100 person-years. Selection of higher-risk individuals partially explains elevated baselines, but longitudinal comparisons pre- and post-PrEP initiation demonstrate incidence increases exceeding secular trends, underscoring causal influences from disinhibited behaviors. Critics note that confounding by increased screening among PrEP users may inflate reported rates, yet adjusted models confirm net elevations. Overall U.S. STI surveillance post-2012 PrEP approval shows population-level rises in syphilis and gonorrhea among MSM, temporally aligned with expanded PrEP access.

Economic Burdens and Access Barriers

The high cost of pre-exposure prophylaxis () regimens represents a significant economic burden, particularly in high-income settings without comprehensive coverage. , the for brand-name tenofovir disoproxil fumarate-emtricitabine (Truvada) exceeds $2,000 per month without , though equivalents can lower this to around $60 per month. Even modest out-of-pocket () expenses, such as copays rising from $0 to $10, can double abandonment rates from 5.5% to over 11%, with rates escalating to 42.6% at OOP costs above $500, disproportionately affecting low-income and uninsured individuals. In 2024, approximately 13% of private U.S. plans did not designate as no-cost sharing under preventive service mandates, exacerbating financial barriers and contributing to unmet need among vulnerable populations. These costs impose broader systemic burdens, as reduced PrEP access correlates with increased incidence and lifetime treatment expenses estimated at $400,000 per person in the U.S. A projected 3% annual decline in PrEP coverage could yield 8,618 additional infections over a decade, incurring $3.6 billion in undiscounted medical costs. Public programs, including expansions and manufacturer patient assistance, mitigate some burdens for eligible groups—such as near-free access for those below 150% of the federal poverty level—but gaps persist for the uninsured and those in states with limited coverage policies. Globally, while federal and philanthropic funding supports domestic prevention (e.g., $1.01 billion allocated by the CDC in 2025), scaling remains constrained by reimbursement shifts and manufacturer pricing dynamics that strain provider finances. Access barriers extend beyond direct costs to structural and geographic factors, particularly in low- and middle-income countries (LMICs) where uptake lags despite high burden. Regulatory hurdles, supply chain limitations, and uneven rollout—prioritizing urban over rural areas—hinder equitable distribution, with only modest progress in PEPFAR-supported regions as of 2024. Recent agreements aim to address affordability, such as (a long-acting injectable) priced at $40 annually for 120 LMICs starting in 2027 through voluntary licensing and generic production, potentially averting millions of infections if scaled. However, implementation requires substantial resource investments for delivery , and persistent challenges like , provider shortages, and low continue to limit initiation among key populations. In high-burden LMICs, these barriers widen disparities, as evidenced by suboptimal coverage despite global targets, underscoring the need for integrated financing to offset both acquisition and service delivery costs.

Recent Advances and Future Directions

Long-Acting Injectable Formulations

Long-acting injectable formulations of pre-exposure prophylaxis (PrEP) represent an advancement aimed at improving adherence by reducing dosing frequency compared to daily oral regimens. Cabotegravir long-acting (CAB-LA), an integrase strand transfer inhibitor, was the first such formulation approved for HIV prevention. Administered as intramuscular gluteal injections, it provides sustained plasma concentrations for protection against HIV acquisition. The U.S. (FDA) approved CAB-LA (branded as Apretude) on December 20, 2021, for individuals at risk of acquisition, including adults and adolescents weighing at least 35 kg. Efficacy was established in two phase 3 trials: HPTN 083, involving men and women who have sex with men, and HPTN 084, involving women. In HPTN 083, CAB-LA reduced incidence to 0.41 events per 100 person-years versus 1.22 for daily oral tenofovir disoproxil fumarate/emtricitabine (), yielding a of 0.34 (95% 0.18-0.62), indicating superiority. HPTN 084 similarly demonstrated CAB-LA's superiority, with incidence of 0.21 per 100 person-years versus 1.69 for TDF/FTC ( 0.12, 95% 0.05-0.27). Real-world observational data from 2024 reported over 99% effectiveness in preventing acquisition among users. Dosing for CAB-LA begins with an initial injection (600 mg), followed by a second at one month, then maintenance injections every two months. Protection is achieved within seven days after the first dose in 95% of individuals, based on pharmacokinetic modeling. Common adverse events include injection-site reactions (e.g., pain, swelling) in up to 82% of participants, though most are mild and transient; serious hypersensitivity or is rare but requires monitoring. CAB-LA addresses adherence challenges of oral , where suboptimal pill-taking leads to breakthrough infections, by enabling clinic-based that confirms engagement. In June 2025, the FDA approved (branded as Yeztugo), a inhibitor administered subcutaneously every six months, marking the first ultra-long-acting injectable . The PURPOSE 1 trial in women showed zero HIV seroconversions among 2,134 lenacapavir recipients versus 39 in the TDF/FTC group, achieving 100% efficacy (95% CI 83.0-100). PURPOSE 2, including diverse populations, reported a 96% compared to background incidence. Initial dosing requires two injections one day apart (two months post-first dose if delayed), with protection onset within days. Injection-site reactions occur in about 63% of users, primarily mild. This biannual regimen further minimizes adherence barriers, particularly in resource-limited settings. Pipeline developments include investigational ultra-long-acting formulations dosed every four to six months, showing sustained in early trials, though not yet approved as of October 2025. These injectables prioritize pharmacokinetic profiles ensuring protective drug levels, contrasting with oral PrEP's reliance on daily user compliance, which meta-analyses indicate fails in 20-50% of users due to forgetfulness or .

Emerging Research and Innovations

In 2024, the PURPOSE 1 and PURPOSE 2 phase 3 trials demonstrated that subcutaneous , a long-acting inhibitor administered every six months, achieved 100% efficacy in preventing acquisition among women in , with zero infections observed among over 2,000 participants. In PURPOSE 2, involving men who have sex with men and gender-diverse individuals, reduced incidence by 89% compared to daily oral tenofovir disoproxil fumarate-emtricitabine (Truvada), with an incidence rate ratio of 0.11 (95% , 0.02-0.51). These results positioned as superior to existing daily options in high-risk populations, prompting its FDA approval as Yeztugo for in June 2025, marking the first six-month protection regimen. Phase 1 studies in 2025 further advanced formulations, showing sustained plasma concentrations and antiviral activity comparable to twice-yearly dosing when administered once yearly, potentially reducing injection frequency and improving adherence in resource-limited settings. The endorsed injectable for prevention in July 2025, citing its efficacy data and potential to address gaps in daily uptake, though implementation challenges include high costs estimated at $28,000 annually per person in low-income countries. Merck's investigational MK-8527, an oral next-generation reverse transcriptase translocation inhibitor, progressed to phase 3 trials in July 2025 following phase 2 data presented at the International AIDS Society Conference, which confirmed monthly dosing tolerability, favorable , and no significant bone or renal toxicity signals in diverse populations. This candidate aims to offer a less frequent oral alternative to daily regimens, with global trials supported by the Bill & Melinda Gates Foundation targeting high-burden regions. Ongoing research explores multimodal innovations, such as integrating with post-exposure prophylaxis to curb bacterial sexually transmitted infections, though randomized trials like Doxy-PrEP have shown mixed adherence impacts without altering core prevention efficacy. Nonhuman models continue to validate novel delivery systems, including modifications for extended-release implants, informing human trials for ultra-long-acting options beyond current injectables. These developments prioritize regimens minimizing user burden while maintaining high barrier to resistance, informed by real-world data indicating that infrequent dosing correlates with better retention rates exceeding 90% in select cohorts.