Next Article in Journal
Relationship Between Human Papilloma Virus and Upper Gastrointestinal Cancers
Previous Article in Journal
Infection and Genomic Properties of Single- and Double-Stranded DNA Cellulophaga Phages
Previous Article in Special Issue
Double-Negative T-Cells during Acute Human Immunodeficiency Virus and Simian Immunodeficiency Virus Infections and Following Early Antiretroviral Therapy Initiation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Acute HIV-1 Infection: Paradigm and Singularity

by
Antoine Chéret
1,2,3
1
Inserm U1016, CNRS UMR 8104, Institut Cochin, Université Paris Descartes, 75014 Paris, France
2
Service Plateforme de Diagnostic et Thérapeutique Pluridisciplinaire, Centre Hospitalier Universitaire, 97159 Pointe à Pitre, Guadeloupe, France
3
INSERM-CIC-1424, Centre Hospitalier Universitaire, 97159 Pointe à Pitre, Guadeloupe, France
Viruses 2025, 17(3), 366; https://doi.org/10.3390/v17030366
Submission received: 2 February 2025 / Revised: 26 February 2025 / Accepted: 1 March 2025 / Published: 3 March 2025
(This article belongs to the Special Issue Acute HIV Infections)

Abstract

:
Acute HIV-1 infection (AHI) is a transient period where the virus causes evident damage to the immune system, including an extensive apoptosis of CD4+ T cells associated with a high level of activation and a major cytokine storm to fight the invading virus. HIV infection establishes persistence by integrating the viral genome into host cell DNA in both replicating and non-replicating forms, effectively hiding from immune surveillance within infected lymphocytes as cellular reservoirs. The measurement of total HIV-1 DNA in peripheral blood mononuclear cells (PBMCs) is a reliable reflection of this reservoir. Initiating treatments during AHI with nucleoside reverse transcriptase inhibitors (NRTIs) and/or integrase strand transfer inhibitors (INSTIs) is essential to alter the dynamics of the global reservoir expansion, and to reduce the establishment of long-lived cellular and tissue reservoirs, while preserving and enhancing specific and non-specific immune responses. Furthermore, some of the patients treated at the AHI stage may become post-treatment controllers and should be informative regarding the mechanism of viral control, so patients treated during AHI are undoubtedly the best candidates to test innovative remission strategies toward a functional cure that could play a pivotal role in long-term HIV control. AHI is characterized by high levels of viral replication, with a significant increase in the risk of HIV transmission. Detecting AHI and initiating early treatment following diagnosis provides a window of opportunity to control the epidemic, particularly in high-risk populations.

1. Introduction

Acute HIV-1 infection (AHI) is a phase marked by a high risk of transmission and intense multiplication of the virus within the host during the 12 weeks following infection. In 2023, the United Nations estimated that there were 39.9 million people living with HIV (PLWH) and around 1.3 million who had been newly infected [1]. AHI has long been recognized as a period of high transmission risk due to the elevated viral loads observed during this phase and the lack of awareness of HIV status, with the risk estimated to be up to 26 times higher than during the chronic phase [2,3,4,5]. The epidemic poses a major risk for men who have sex with men (MSM) in both developed and developing countries [1]. In 2016, the CDC estimated that approximately 4% of the new HIV infections in the United States involved transmission from a person with acute HIV [3]. AHI can present without symptoms or with non-specific, variable, and self-limiting signs, often leading to misdiagnosis as another viral infection [6]. Consequently, a high level of clinical suspicion and thorough assessment of risk factors are essential for its identification. Since 2015, the World Health Organization (WHO) guidelines have recommended initiating combination antiretroviral therapy (ART) in PLWH, regardless of CD4+ T-cell count [7]. The use of second-generation INSTIs has been increasingly recommended by experts and incorporated into current guidelines for the management of AHI [8,9,10]. Such treatments should include two nucleoside inhibitors (tenofovir disoproxil or tenofovir alafenamide and lamivudine or emtricitabine) in combination with dolutegravir [11] or bictegravir or alternatively, a boosted protease inhibitor. The combination therapy is also adjusted according to the results of genotypic antiretroviral resistance tests, tolerance, and comorbidities, with the aim of maintaining long-term virological success. Early treatment initiation during AHI promotes individual and public health benefits, including preventing AIDS (Acquired Immunodeficiency Syndrome) progression and reducing secondary HIV transmission [12]. This early intervention also positions these patients as optimal candidates for future HIV treatment strategies, as their immune system is less damaged, and the viral replication and evolution are limited. Therefore, there is a strong rationale for establishing policies to promote early HIV diagnosis, especially for patients with symptoms consistent with AHI or recent exposure to HIV, to ensure timely entry into care and prompt treatment.
This article reviews the key aspects of AHI and its treatment implications, focusing on strategies aimed at ending the HIV epidemic through remission and the pursuit of a cure, which remain significant challenges.

2. HIV-1 Infection and Viral Reservoir Establishment

Following mucosal HIV infection, the virus spreads via myeloid cells such as dendritic cells (DCs) and macrophages. Soon after HIV-1 exposure, these cells migrate to draining lymph nodes and transfer it to CD4+ T cells through virological synapses in the absence of a productive infection [13,14]. They allow a significant spread of viral infection to tissues, and HIV can persist for years in myeloid cells [15], highlighting their specific role in long-term viral persistence [16]. In SIV models, infected DCs reach lymph nodes within 24 h, marking the ‘eclipse stage’, a period lasting 7–21 days during which the virus remains undetectable [17,18]. During the acute phase, the virus predominantly targets CCR5+ cells, including CD4+ T cells and myeloid cells [19,20]. Although R5- and X4-tropic variants may coexist in the source, X4 variants contribute minimally to early infection [21]. However, X4 or mixed X4/dual-tropic viruses are linked to faster disease progression due to their impact on lymphocyte homeostasis [22,23]. X4 or mixed R5/X4 variants typically emerge in advanced stages, especially in patients with CD4+ T-cell counts below 200 cells/µL [24].
Phylogenetic studies of the circulating viruses in acute HIV infection (AHI) cohorts have shown that 30–50% of transmissions occur within shared clusters [25,26,27]. However, some studies indicate multiple viral founders in cases involving more permissive transmission routes, such as men who have sex with men (MSM) and intravenous drug users (IVUs) [20,28]. Interestingly, another study found a similar frequency of single-virus transmission across MSM, heterosexuals, and IVUs [29], with variations likely due to the AHI stage and immune pressure over time [12]. Typically, transmission involves a single or limited number of replication-competent virions that lead to productive infection [20,30,31]. The virus rapidly spreads to secondary lymphoid organs, including the lymph nodes, spleen, and gut-associated lymphoid tissues, which contain 98% of all CD4+ T cells and are sites of high levels of replication. This leads to multi-organ invasion, including the genital tract [32] and central nervous system [33]. The peak of HIV-RNA levels occurs within two to four weeks post-infection, establishing a ‘viral set point’ with prognostic significance approximately 31 days (interquartile range 18–42 days) after initial plasma viremia detection [34,35,36,37]. Notably, viral replication capacity in AHI influenced by the gag gene correlates with the viral set point and significantly impacts disease progression [38].
A latent HIV cellular reservoir is rapidly established after transmission through proviral integration into host cell DNA [39,40]. In AHI, virus production primarily occurs in activated CD4+ T cells, forming an ‘active reservoir’ that amplifies the amount of virus and leads to a stable, quiescent reservoir of transcriptionally silent cells that maintain long-term viral persistence [41,42]. Total HIV-1 DNA in PBMCs (HIV-DNA) is a relevant clinical marker of the reservoir size, affecting disease progression despite its inability to differentiate between replication-competent from defective virus [43]. Key reservoirs include the gut, rich in CCR5+CD4+ and TH17 cells [44,45]; the central nervous system (CNS), involving infected microglia and astrocytes [46]; and the seminal compartment, which accumulates HIV progressively during AHI [47,48]. Early AHI stages show lower HIV levels in blood and tissues [40], but reservoir levels in PBMCs during AHI are higher compared to the chronic phase, predicting disease progression [43,49]. Patients with high levels of integrated HIV-DNA shortly after infection are at a higher risk of rapid progression towards AIDS, indicating that the rapidity of progression is well determined soon after infection [50]. Integrated HIV-DNA likely accounts for most of the predictive value of total HIV-DNA observed in previous studies, as this stable form of HIV persistence plays a key role in driving the course of infection [50].
The quiescent CD4+ T-memory cells are the form of HIV reservoir that has been most studied. With an increasing expression of CCR5, we distinguish naive (TNs), central memory (TCMs), transitional memory (TTMs), effector memory (TEMs), and effector T cells [51,52]. These various lymphocyte subpopulations exhibit different half-lives, as well as distinct activation and proliferation capacity. Quiescent CD4+ T cells exhibit high infection levels in AHI [27,42,53]. Long-lived TCMs and transitional memory cells, although low in quantity, are critical to reservoir maintenance due to their long half-lives, and TCMs ensure the long-term memory of the immune response in humans [54,55]. In AHI, the TTM reservoir is dominant [27], whereas in chronic infection, TCM cells with extended half-lives play a major role in maintaining viral persistence, sustaining residual replication, and contributing to progressive CD4+ T-cell depletion [56,57].

3. Immune Response

During acute HIV infection (AHI), myeloid cells such as macrophages and dendritic cells represent the first line of defense [16]. Myeloid cells detect pathogens through pattern recognition receptors (PRRs) located on their membranes or within the cytosol, recognizing viral pathogen-associated molecular patterns (PAMPs). This recognition triggers the secretion of interferon (IFN)-α/β, the primary antiviral molecule, initiating the immune response against the virus [58,59]. These cells initiate the adaptive immune response in lymphoid tissues by presenting antigens and releasing cytokines and chemokines, which guide the polarization and migration of naïve T and B cells [60]. Both cell-mediated and soluble factors are generated, leading to intense immune activation with elevated plasma levels of several cytokines and chemokines, including rapid and transient increases in type I and III interferons that predict disease progression [61] and interleukin-15; a significant rise in inducible protein 10 levels; more sustained increases in tumor necrosis factor alpha and monocyte chemoattractant protein-1; slower increases in additional pro-inflammatory factors such as interleukin-6, interleukin-8, interleukin-18, and interferon-gamma; and a late rise in the immunoregulatory cytokine IL-10 levels [62]. Cytokine and chemokine levels in plasma rise with viral replication [62], which recruits and activates immune cells, including macrophages, natural killer (NK) cells, and dendritic cells. Among them, interleukin-12, interleukin-15, and interleukin-2 are powerful activators of NK cells [16]. Innate immune cells play a crucial role in the early stages of HIV infection to control viral transmission and dissemination [63,64] and modulate the adaptive immune response [16,65]. While NK cell and DC functions are impaired during chronic HIV-1 infection, they may instead be enhanced during AHI, leading to increased immune activation and viral replication [65].
Cytokine storms induce apoptosis in HIV-specific and non-specific B and T cells [65,66,67]. This response leads to the exhaustion and senescence of T cells, as well as the depletion of T and B cells [68] in the blood and gastrointestinal lymphoid tissue (GALT) [69], resulting in follicular hyperplasia, immune ‘burn out’ in lymphoid tissues [70], lymph node fibrosis, regulatory T-cell (Treg) proliferation [71], and microbial translocation, especially during chronic infection [72,73,74]. This exaggerated cytokine response likely contributes to the early immunopathology of the infection and associated long-term consequences [62]. Additionally, host restriction factors (HRFs) play a vital role in the early antiviral response. These cellular proteins suppress viral replication and dissemination by interfering with key stages of the viral life cycle, including entry, genome transcription and replication, protein translation, viral particle assembly, and release [75].
CD4+ and CD8+ T-cell responses play key roles in controlling viral replication during AHI [76,77]. CD8+ T cells initially control viremia by recognizing early-stage viral epitopes (envelope glycoprotein (Env), negative regulatory factor (Nef), group-specific antigen (Gag), protein 24 (p24), proteins coded by the Polymerase gene (Pol)) [78]; the strength and timing of the cytotoxic response influence the viral set point [37]. This response is highly specific to autologous epitopes with limited cross-reactivity and involves cytolytic activity through perforin, a key mechanism for inhibiting viral replication, as observed in HIV controllers who present strong CD8+ T-cell activity and were able to suppress HIV infection [79]. Additionally, these cells secrete β-chemokines (Regulated upon Activation, Normal T-cell-Expressed and -Secreted (RANTES), Macrophage Inflammatory Protein-1 alpha (MIP-1α), Macrophage Inflammatory Protein-1 beta (MIP-1β)), which further contribute to viral control [80]. Responses to conserved epitopes are linked to long-term control, as ‘protective’ Human Leukocyte Antigen (HLA)-I alleles (e.g., HLA-B27, HLA-B5701) correlate with lower viral loads and slower disease progression [81] due to the recognition of less variable viral regions (e.g., Gag) [82,83]. However, this response diminishes over time due to mutant viral escape, chronic activation, and reduced TH1 response [82]. CD8+ T cells also lose the ability to upregulate perforin after peak viremia [84,85]. Innate immune cells, including dendritic cells, monocytes, and NK cells, influence disease progression rates, and certain receptor–HLA combinations are linked with better NK function and viremic control [86,87].
The CD4+ T-cell response, particularly through its TH1 helper function, is crucial in controlling HIV infection by recruiting immune cells, aiding lymphocyte expansion, and enabling direct cytotoxicity [88]. This response, however, is significantly compromised due to early CD4+ T-cell depletion post-Fiebig I [89,90]. The beneficial impact of CD4+ T-cell function on lowering the viral set point and attenuating disease progression during AHI has been established [91,92,93]. Although both CD4+ and CD8+ T-cell responses are weak in AHI, a small percentage of HIV controllers (0.5%) maintain strong CD4 and CD8 responses, contributing to natural viral control [79,94,95]. High cytokine levels during AHI cause uncontrolled CD4+ T-cell proliferation, depleting memory cells and impeding immune compensation [96]. CD4+ T-cell count falls quickly in most patients, often dropping below normal levels (<500/mm3) [97]. CD4+ T-cell function declines due to rapid anergy, which results in the overexpression of the negative regulatory factor PD-1 [98], blocking of which promotes better control of viral replication [99]. The significant loss of CD4+ T helpers and continuous activation are responsible for the defective functionality of CD8+ T cells [100].
The depletion of T lymphocytes and the disruption of T-cell homeostasis presumably drive and precede the progressive loss of B cells [101], notably due to the loss of T-cell types important for the development of germinal centers, with up to 50% of these centers being lost within the first 80 days of infection. Moreover, persistent B-cell activation at the time of AHI creates large populations requiring T-cell help, which is unavailable due to the loss of antigen-specific T cells. Without this support, B cells undergo apoptosis within days [102]. High cytokine production also drives excessive B-cell activation, contributing to a reduction in B cells [101], with functional disruption leading to hypergammaglobulinemia, polyclonal anti-HIV activation [103], weaker responses to non-HIV antigens, inadequate antibody production, and diminished long-term B-cell memory [104,105]. Initial gp41 antibodies appear around day 13, gp120 antibodies on day 14, and specific IgA in mucosal tissues three weeks post-infection, although these are non-neutralizing. Neutralizing antibodies emerge at 12 weeks but are limited in targeting the transmitted virus, failing to impact evolving variants [106].
These immune responses ultimately yield partial viral control, establishing a virologic set point in untreated individuals [19]. Interdependence between immune activation and the viral reservoir creates a self-sustaining cycle that is central to HIV persistence and poses a significant challenge to eradication efforts [107] (Figure 1).

4. Diagnosing Acute HIV Infection: A Critical Issue

Clinical symptoms, resembling a flu-like syndrome, are present in 23–92% [6,108] of patients with AHI; the proportion of asymptomatic or mildly symptomatic acute HIV infections (AHIs) remains difficult to determine, as individuals without symptoms often do not seek medical care. Most of the studies recruited symptomatic study populations and thus give a biased view of acute infection. AHI is symptomatic in over 50% of cases [109]; however, more than 90% of AHI cases remain undiagnosed [110] primarily due to the highly variable clinical manifestations [111]. Most symptoms are non-specific, and atypical clinical presentations are common. The lack of specific symptoms or a combination of symptoms with adequate diagnostic sensitivity and specificity makes the prompt diagnosis of acute HIV infection particularly challenging [6]. Symptom severity, quantity, and, in some studies, duration of AHI are associated with the rate of subsequent disease progression, especially when neurological symptoms are present [112]. The symptoms are the consequences of high levels of cellular activation and inflammation [108,113]. They usually appear within 10 to 15 days, and persist up to 10 weeks after acute infection [108,114]. HIV infection is a multi-organ disease; the main symptoms may include fever, headache, myalgia, pharyngitis, lymphadenopathy, skin rash, gastrointestinal problems, and neurocognitive disorders [114]. AHI presents a mononucleosis-like syndrome that is often accompanied by thrombocytopenia and hepatic cytolysis [11,114]. However, clinical and biological features are subject to individual variations: the intensity of symptoms is correlated with HIV-RNA [115] and HIV-DNA levels in PBMCs [116] and inversely correlated with the number of CD4+ T-cell count [113,116]. The severity of symptoms is also associated with the risk of disease progression [117,118]. Any acute viral syndrome in a patient at risk of HIV exposure should prompt consideration of an HIV diagnosis. Therefore, increased awareness among primary care physicians should be encouraged, particularly when encountering individuals from high-risk populations.
Following viral transmission, an eclipse phase of approximately ten days occurs before HIV-RNA can be detected in blood plasma. The kinetics of antibody appearance (assessed by ELISA), combined with the quantification of HIV-RNA and/or HIV-DNA levels (in newborns) [119] and the p24 antigen, have enabled the establishment of diagnostic criteria for AHI. The most widely used classification system, known as ‘Fiebig stages’ [120], is based on the kinetics of these markers. Nevertheless, a new classification may emerge with the advent of more sensitive fourth-generation ELISA tests that incorporate P24 antigen detection [121]. The number of HIV antibodies detected by Western blot (WB) correlates with the time elapsed since infection: ≤1 antibody after a median of 28 days post-infection (range: 18–49 days), 2–4 antibodies after 39 days (range: 16–118 days), ≥5 antibodies after 63 days (range: 18–241 days) [116]. The distinction between acute and early HIV infection has varied definitions, but it is commonly associated with the full development of antibody responses and the establishment of a stable viral load set point, which typically occurs 4 to 5 weeks after the onset of viremia [35]. Thus, during the very early stages of AHI, the diagnosis can be established by the presence of a negative or weakly positive HIV-1 ELISA (third or fourth generation), a negative or incomplete (≤1 antibody) Western blot for HIV-1, and a positive HIV-RNA load and/or positive p24 antigen (Fiebig stages I, II, and III). In later stages, the diagnosis of AHI may be established by a positive ELISA assay with an incomplete HIV-1 Western blot (≥2 and <5 antibodies, specifically the presence of anti-p24 antibodies along with anti-gp160, anti-gp120, or anti-gp41 antibodies) and a positive HIV-RNA load (Fiebig stages IV and V) [122]. However, immunoblots may not be effective in confirming the recency of HIV-1 infections [123]. Alternatively, rapid diagnostic tests (RDTs), which do not require laboratory infrastructure, are gaining public health interest. These tests promote access to HIV screening, providing results within 30 min, and enabling use in non-clinical ‘outside the walls’ settings for populations at risk of HIV acquisition. There are various RDTs (using immunochromatographic agglutinations) that can be achieved on different biological fluids (plasma, whole blood, or saliva). While they are less sensitive than ELISA tests, particularly when samples are taken during the seroconversion phase, and their performance is weaker with saliva samples [124], RDTs remain valuable for mass screening in high-risk populations [123]. For the earliest stages of HIV, the use of nucleic acid testing (NAT) allows viral RNA detection even before antibodies or p24 antigens are present, thereby identifying acute infections more reliably. Although studies have reported that fourth-generation ELISA tests for HIV, which combine the p24 antigen and antibody detection, are effective in identifying infection starting at Fiebig stage II, stage I infections remain omitted [125]. In clinical settings, combining fourth-generation assays with NAT could improve early detection in high-risk patients, particularly those presenting with symptoms consistent with acute HIV infection (Figure 2). This approach can help bridge the detection gap at Fiebig stage I, enabling timely diagnosis and reducing potential transmission risks [125]. According to Powers et al., more targeted and frequent testing in high-risk populations is essential for identifying acute HIV infections [126]. The need for targeted screening within high-risk groups undoubtedly requires better integration into public health responses. The revelation of serological status allows and frequently leads to a modification of sexual behavior which in turn helps reduce the risk of transmission [127]. Without these strategies, efforts to steer the epidemic toward elimination could be undermined. Early detection during the acute phase is crucial for initiating treatment to control the spread of HIV and prevent the establishment of large HIV reservoirs, which are central to long-term persistence, transmission risk, and disease progression [126]. Early and effective screening in symptomatic patients or those with recent high-risk exposure should facilitate prompt treatment, yielding proven benefits both individually and collectively.

5. Key Virological Considerations in Acute HIV Treatment

Early treatment during acute HIV infection (AHI) can reduce plasma HIV-RNA to undetectable levels (<50 copies/mL) within months [11,128,129,130]. Additionally, more pronounced benefits are observed when treatment is initiated earlier in AHI, including a lower viral set point and better preservation of CD4+ T-cell counts [129,131]. Despite the benefits of initiating ART during AHI, low-level viremia persists, originating from at least two cellular reservoir compartments: one where viral production gradually declines over time and another where it remains stable for at least seven years [132]. The clearance of HIV-1 infected cells is linked to the extent of viral replication, as measured by cell-associated RNA levels in both blood and lymph nodes [132]. The gut-associated lymphoid tissue (GALT), which harbors 60% of the body’s lymphocytes, plays a major role in HIV persistence and is therefore a critical target for strategies aimed at reducing this viral reservoir. [43]. HIV DNA levels in GALT decrease following the initiation of cART in AHI [133] but persist nonetheless, with variations observed across different gut sites [45,134,135]. Studies have shown that starting ART in AHI significantly impacts the HIV blood reservoir, with faster and more profound reductions compared to treatment during the chronic phase [43]. Total HIV-DNA decreases particularly within the first eight months of AHI treatment [136] and interestingly, correlates well with integrated HIV-DNA levels [137]. The OPTIPRIM-ANRS 147 and 148 clinical trials demonstrated that two years of ART significantly reduced HIV-DNA in PBMCs and rectal tissue, particularly levels in short-lived memory T cells (TTMs, TEMs). These cells, which constitute the primary reservoir, have limited proliferative capacity and shorter lifespans due to their higher susceptibility to activation and viral production [30]. Early treatment during AHI stabilizes the reservoir, preserving long-lived memory T cells (TNs, TCMs) and limiting the reservoir to short-lived cells [27,30,138,139]. This effect likely results from suppressed viral replication and reduced memory T-cell activation. Over time, ART allows HIV-DNA decline, likely reflecting the death of short-lived infected cells [136].
Conversely, in patients treated during the chronic phase, total HIV-DNA levels showed the most significant decline during the first year of ART, primarily due to the clearance of infected activated cells. However, this decline slowed thereafter, as integrated HIV-DNA in latent cells persists at high levels [140]. Early ART administration reduces the initial, labile, unintegrated forms of HIV and limits integrated forms, largely eliminating the majority of short-lived cells infected during AHI. In contrast, chronic-phase reservoirs consist primarily of stable proviruses in long-lived cells unaffected by ART [50,141]. Thus, early ART during AHI prevents the establishment of stable, hard-to-eliminate reservoirs. Studying integrated and total cell-associated HIV-DNA could help identify optimal candidates for curative strategies, potentially supporting a functional cure when combined with other therapeutic interventions. In various cohort studies and randomized trials on AHI treatment, partial or even long-term viral control was observed in some patients who discontinued treatment [142,143,144,145]. Early treatment initiation delays the onset of virologic rebound, with some studies suggesting an optimal start time of around three weeks post-infection [146,147]. Additionally, the total cell-associated HIV-DNA level has been shown to predict the timing of viral rebound following treatment discontinuation [148]. In a very limited number of individuals designated post-treatment HIV controllers (PTCs), viremia remains suppressed for prolonged periods after ART withdrawal [149]. The protection of central memory and naive CD4+ T cells is a key factor in post-treatment controllers (PTCs) who were treated during AHI [30]. The estimated probability of maintaining viral control 24 months after early treatment interruption is approximately 15% among patients who received treatment during AHI versus less than 1% of those not treated [145]. This immune protection resembles that observed in individuals with protective HLA alleles, such as HLA-B5701, HLA-B5703, and HLA-B*27, which are commonly associated with elite controllers but less frequent in human PTCs [150,151]. Similar patterns have also been noted in animal models, such as sooty mangabeys and early-treated monkeys [152]. A low HIV reservoir level, especially when predominantly found in naive and central memory CD4+ T cells, is strongly associated with favorable outcomes in patients treated during AHI [30].
Historically, HIV-1 clade B strains predominated in Europe and North America, shaping the design of diagnostic tests and treatment guidelines. However, globalization and migrations have increased viral genetic diversity in these regions, with expansion of HIV-1 non-B clades (A, C, D, and recombinants) [153]. This diversification directly impacts acute HIV infection because some non-B clades may lead to faster progression to AIDS and exhibit specific resistance patterns to some antiretrovirals, potentially complicating initial clinical management and the accurate measurement of viral load [154]. Due to variations in virulence and differing susceptibility or resistance patterns to antiviral therapies, the spread of non-B clades should be monitored and considered in future studies, particularly in European countries [155,156]. Viral mutations can progressively occur after the peak of viremia in acute HIV infection [20].
Early treatment limits the virus’s genetic diversity in the blood [27,157] and gut-associated lymphoid tissue (GALT), thereby potentially enhancing the immune response by reducing epitope variation, which otherwise lowers the effectiveness of specific anti-HIV responses [158]. This low genetic diversity, in contrast to chronically treated patients, is maintained under treatment [30]. Treating patients during AHI prevents viral evolution and the accumulation of defective proviruses, as observed in post-treatment controllers (PTCs) [157]. While there are ongoing discussions among scientists regarding the significance of integrated defective proviruses, it cannot be denied that they may contribute to HIV pathogenesis by producing viral RNAs and proteins that drive persistent immune stimulation, leading to lymphocyte activation and inflammation [159,160]. Viral variabilities, both quantitatively and qualitatively, during the course of an HIV infection are important pathogenetic considerations when aiming to reduce the reservoir. The investigation of additional predictive markers, especially through virological and immunological evaluations, remains a pressing challenge during acute HIV infection [161].

6. Key Immunological Considerations in Acute HIV Treatment

By inhibiting viral replication, early treatment during AHI effectively reduces immune activation [30,133,162], can abrogate the HIV-induced cytokine storm [163], and may help limit the systemic spread of viral products [164,165]. This normalization may lower the risk of disease progression compared to patients who are treated later [166,167]. However, in monkey models, lymphoid tissue activation and germinal center hyperplasia persist, indicating ongoing viral replication in HIV reservoir cells [168]. Additionally, low-grade inflammation continues in two-thirds of people living with HIV (PLWH) on antiretroviral therapy (ART), indicating a persistent inflammatory signature [169], even when ART is initiated during AHI. This sustained inflammation may predispose PLWH to cardiovascular and other comorbidities, particularly if treated during chronic infection [170]. Chronic immune activation in HIV likely stems from various factors, including HIV’s direct cytopathic effects, the release of pro-inflammatory cytokines, loss of regulatory T cells, viral co-infections (e.g., cytomegalovirus), and microbial translocation from compromised gut barriers [18]. This widespread inflammation may also impact the central nervous system (CNS), where elevated neuroinflammatory markers like neopterin have been observed [171]. While early ART initiation can reduce cerebrospinal fluid inflammation within six months, inflammatory markers in the blood often remain elevated, highlighting the need for further research into early ART’s potential role in limiting HIV-associated effects on the brain [33].
ART during AHI also leads to a steady gain in CD4+ T-cell count and rapid viral suppression (<50 copies/mL) [128,129,172,173]. CD4+ and CD8+ T-cell functions are preserved [174,175,176] with the maintenance of HIV-specific CD4+ T-cell help, even when treatment is initiated at later stages of AHI [72]. However, mucosal CD4+ T cells are better conserved when treatment begins in early AHI [165,177]. This is similar to what is observed in HIV controllers [178], whereas in patients with chronic HIV, ART improves mucosal CD4+ T-cell differentiation but cannot prevent the persistent depletion of total CD4+ T cells [164]. Early treatment can partially restore gut CD4+ T cells, though some degree of functional impairment may persist. Starting ART before Fiebig stage I/II can also prevent both the quantitative and qualitative loss of Th17 cells, which are crucial for maintaining mucosal barriers [89]. Furthermore, the deactivation of CD8+ T cells with favorable functional profiles allows for memory T-cell proliferation and function [179], especially when ART is initiated before peak viremia [180]. These memory T cells, which are absent in untreated or later-treated individuals, are crucial for virologic control and are associated with viral clearance in SIV infection [181]. In macaque-modeled studies, early ART promotes post-treatment controller (PTC) status by expanding central memory CD8+ T cells with enhanced survival and proliferation [182]. CD4/CD8 recovery is faster and more robust in those treated during AHI [183,184], particularly at the very early stages (Fiebig stage I). However, persistent immune dysfunction is reflected in a reduced CD4/CD8 ratio [40] which is associated with ongoing chronic inflammation [185].
Additionally, AHI treatment restores natural killer and dendritic cell numbers [186,187] and preserves B-cell responses to HIV and non-HIV antigens by maintaining functional follicular T helpers and B cells [101,188]. Surprisingly, extremely early ART initiation may prevent the development of an antiviral response altogether, as observed in rare non-reactive cases detected with fourth-generation assays [189]. However, such early treatment does not prevent viral rebound after ART discontinuation [190]. In clinical practice, very early diagnosis is not so feasible, and patients are generally treated around four weeks post-infection [11,128]. Optimal immune priming, balancing early ART benefits, and proper immune response development are crucial for maximizing the advantages of early treatment, as observed in PTC studies [182].
The benefits of early treatment may not always persist after treatment interruption, supporting the rationale for discontinuing treatment only under strictly controlled conditions or within therapeutic trials. Lastly, qualitative and quantitative differences in the reservoir between patients treated during AHI versus chronic HIV infection highlight the multiple advantages of initiating treatment during AHI. The main benefits of initiating treatment during acute HIV infection compared to late treatment are shown in Table 1.

7. Clinical Implications and Challenges in Acute HIV Treatment

The immunovirological impact of treatment during symptomatic AHI helps reduce the intensity and duration of symptoms, decreases the frequency of opportunistic infections, and lowers mortality and morbidity in these patients [167,175,191]. Interestingly, women who initiate ART during AHI exhibit a stronger immunovirological response compared to men, which may provide them with additional protection against adverse clinical events and premature aging [192]. In addition, treating AHI has a neuroprotective effect by limiting immunological processes that are deleterious to the central nervous system (CNS). It likely arrests the effects of HIV on the CNS, offering a better neuroprotection when compared to initiation during chronic infection [33]. Furthermore, immune recovery with reduced inflammation [185] and relative preservation of immune cells [176] allow a significant improvement in patient quality of life [172]. Nevertheless, inflammation persists even after starting ART and achieving undetectable plasma HIV RNA levels [169]. Factors that may contribute to chronic immune activation include the direct cytopathic effect of HIV, the production of pro-inflammatory cytokines by innate immune cells, the depletion of regulatory T cells, viral co-infections like cytomegalovirus (CMV), and the translocation of microbial products through damaged intestinal mucosal barriers [18]. Moreover, defective HIV proviruses can trigger immune responses through their RNA transcripts and viral proteins, which form virus-like particles. This continuous production of HIV-1 proteins from defective proviruses, even without active replication, contributes to persistent immune activation, despite undetectable HIV-1 levels [193]. Interestingly, studies in PTCs have shown that the combination of low viral diversity and defective provirus levels, along with specific host and immune factors, likely contributes to the control of HIV infection in PTCs who were treated during AHI, highlighting another benefit of treatment at the time of AHI [157]. Also, despite persistent immune activation, long-term clinical benefits, such as reductions in cardiovascular events, cancer, and HIV-related complications, are expected in patients treated during AHI and should be further explored in the coming years [33].
Several studies conducted in European or North American countries suggest that newly infected patients are a major source of transmission, maintaining the epidemic within risk groups [194]. These studies have detected transmission of new viral clusters from patients with acute or early infection [25,195]. The question of treatment as prevention (TASP) is particularly significant in AHI with high levels of genital and plasma viral load [196]. The transmission risk during AHI is 26 times greater than that of the chronic stage [2] and estimated to be responsible for up to 50% of all new infections [195]. The risk of sexual HIV-1 transmission is also closely correlated with the level of HIV-1 RNA in genital secretions [197], which follows the dynamic of HIV-1 RNA in blood [198,199]. Early treatment during AHI facilitates the purging of viral particles and infected cells in the genital compartment [47,48]. Consequently, drug pharmacokinetics may be a factor to consider for treatment regimens, including drugs that concentrate in the genital tract of men and women. Integrase inhibitors are among the most effective drugs for rapidly decreasing viral load, including within the genital tract [48]. In addition, the ‘chemsex’ practice of using drugs to enhance and extend sexual experiences has become increasingly prevalent in urban communities, particularly among men who have sex with men (MSM) [200]. This behavior significantly increases the risk of HIV transmission especially for patients with AHI [200,201]. Moreover, chemsex is associated with increased frequency of sexual partner changes and decreased condom use, due to drug-induced loss of inhibition and reduced engagement in HIV testing [202]. A higher prevalence of chemsex of 29% has been reported in HIV+ MSM [203,204]. Additionally, individuals involved in chemsex tend to have less consistent adherence to HIV treatment, further exacerbating the risk of untreated acute infection or therapeutic non-adherence and complicating efforts to prevent HIV transmission [202,203]. Patients who engage in chemsex should be incorporated into preventive and educational programs aimed at reducing the occurrence of high-risk sexual behaviors and STIs [203].
More recently, pre-exposure prophylaxis (PrEP) with emtricitabine/tenofovir is a new high-efficacy tool to reduce HIV incidence among MSM when adherence is optimal [205,206,207]. Despite its high efficacy, non-adherence to daily PrEP remains a significant issue. A study of 210 gay men engaged in PrEP in California and New York revealed that over two-thirds missed doses, with an average of four to five missed doses in the past month. Socio-behavioral factors, such as attitudes, perceived control, and social barriers, contributed to non-adherence [208]. Enhancing adherence could help reduce new HIV infections. Healthcare providers should play a major role in promoting adherence through strategies that encourage habit formation, ultimately helping to reduce transmission risks particularly in high-risk groups [208]. In addition, there is a context of declining condom use in the general MSM population, including those living with HIV, evidenced by the concomitant increase in the incidence of bacterial STIs since the early 2000s, notably in France [209,210,211]. Although rare, HIV infections can occur despite high adherence to PrEP. With the anticipated broader use of long-acting PrEP, new challenges are likely to arise. Prompt identification of these infections should be integrated into PrEP follow-up to minimize the risk of overlooked HIV infections and exposure to suboptimal antiretroviral concentrations [211].
To date, some studies have evaluated the immunovirological efficacy of treatments and observed that virological rebounds following treatment interruption increase the risk of transmission, as reported in the Swiss cohort study [212] and the OPTIPRIM ANRS-147 clinical trial, in which a case of viral transmission occurred one month after treatment interruption [128]. Finally, the significantly small proportion of patients treated during AHI who achieve PTC [145] strongly argues against treatment cessation outside of clinical trial settings. In this context, a functional cure strategy aims to reduce the viral reservoir and enhance immune control, allowing HIV suppression without ART. Over the years, several approaches have emerged, including ‘shock-and-kill’, which reactivates latent HIV with latency reversal agents before eliminating infected cells, and ‘block-and-lock’, which prevents viral reactivation. Therapeutic vaccination, the use of PD-1 inhibitors and Toll-Like Receptor agonists to enhance HIV-specific CD8+ T-cell responses to target and eliminate infected cells, and adoptive chimeric antigen receptor (CAR) T-cell therapy are also being investigated as potential strategies [213]. Another strategy for eliminating the virus involves the use of HIV-1-specific broadly neutralizing antibodies (bNAbs), which target a conserved region of the HIV envelope, allowing them to neutralize a broad range of HIV strains, and they are being actively studied to control HIV replication in patients treated during AHI [214,215,216]. The simplification of treatment during the chronic phase, aimed at reducing long-term side effects while preserving immunovirological efficacy, remains a subject of ongoing debate. Nevertheless, another challenge lies in defining the optimal timing for simplification, such as the use of dual therapy, particularly for patients who initiated and maintained early treatment after AHI.

8. Conclusions

Despite decades of HIV prevention efforts, the HIV epidemic persists, underscoring the critical challenge of detecting infections at the earliest stage. Improving awareness, strengthening screening programs, and ensuring quick access to care—particularly for high-risk populations—are essential. Initiating antiretroviral therapy during acute HIV infection not only helps limit transmission within at-risk populations but also provides substantial immunological and virological benefits at the individual level.
Innovative strategies, including clinical studies exploring ART interruption and immune-based interventions such as broadly neutralizing antibodies (bNAbs), present promising avenues toward a functional cure. Prioritizing patients treated during the acute phase of HIV infection for these studies could further progress toward the development of more effective treatments. Taking these factors into account will accelerate progress in combating the HIV epidemic and advancing toward the goal of a functional cure.

Funding

This research received no external funding.

Acknowledgments

We thank Christine Rouzioux for reviewing the paper and Jean Pierre Routy for English editing.

Conflicts of Interest

Antoine Chéret declares no conflicts of interest.

References

  1. Joint United Nations Programme on HIV/AIDS (UNAIDS). Global HIV Statistics 2023 Fact Sheet. July 2024. Available online: https://www.unaids.org/en/resources/fact-sheet (accessed on 2 March 2025).
  2. Hollingsworth, T.D.; Anderson, R.M.; Fraser, C. HIV-1 transmission, by stage of infection. J. Infect. Dis. 2008, 198, 687–693. [Google Scholar] [CrossRef] [PubMed]
  3. Li, Z.; Purcell, D.W.; Sansom, S.L.; Hayes, D.; Hall, H.I. Vital Signs: HIV Transmission Along the Continuum of Care United States, 2016. MMWR Morb. Mortal. Wkly. Rep. 2019, 68, 267–272. [Google Scholar] [CrossRef] [PubMed]
  4. Wawer, M.J.; Gray, R.H.; Sewankambo, N.K.; Serwadda, D.; Li, X.; Laeyendecker, O.; Kiwanuka, N.; Kigozi, G.; Kiddugavu, M.; Lutalo, T.; et al. Rates of HIV-1 transmission per coital act, by stage of HIV-1 infection, in Rakai, Uganda. J. Infect. Dis. 2005, 191, 1403–1409. [Google Scholar] [CrossRef] [PubMed]
  5. Pilcher, C.D.; Joaki, G.; Hoffman, I.F.; Martinson, F.E.; Mapanje, C.; Stewart, P.W.; Powers, K.A.; Galvin, S.; Chilongozi, D.; Gama, S.; et al. Amplified transmission of HIV-1: Comparison of HIV-1 concentrations in semen and blood during acute and chronic infection. AIDS 2007, 21, 1723–1730. [Google Scholar] [CrossRef]
  6. Daar, E.S.; Pilcher, C.D.; Hecht, F.M. Clinical presentation and diagnosis of primary HIV-1 infection. Curr. Opin. HIV AIDS 2008, 3, 10–15. [Google Scholar] [CrossRef]
  7. Organisation, W.H. Consolidated Guidelines on HIV, Viral Hepatitis and STI Prevention, Diagnosis, Treatment and Care for Key Populations. 2022. Available online: https://www.who.int/publications/i/item/9789240052390 (accessed on 2 March 2025).
  8. Delobel, P. Initiation d’un Premier Traitement Antirétroviral Chez l’adulte Vivant avec le VIH. 2024. Available online: https://www.has-sante.fr/jcms/p_3545694/fr/initiation-d-un-premier-traitement-antiretroviral-chez-l-adulte-vivant-avec-le-vih (accessed on 2 March 2025).
  9. Panel on Antiretroviral Guidelines for Adults and Adolescents. US guidelines for the Use of Antiretroviral Agents in Adults and Adolescents with HIV. Dep. Health Hum. Serv. 2024, 6. [Google Scholar]
  10. EACS Guidelines for the Management of People Living with HIV in Europe. Available online: https://www.eacsociety.org/media/guidelines-12.0.pdf (accessed on 2 March 2025).
  11. Cheret, A.; Bauer, R.; Meiffredy, V.; Lopez, P.; Ajana, F.; Lacombe, K.; Morlat, P.; Lascoux, C.; Reynes, J.; Calin, R.; et al. Once-daily dolutegravir versus darunavir plus cobicistat in adults at the time of primary HIV-1 infection: The OPTIPRIM2-ANRS 169 randomized, open-label, Phase 3 trial. J. Antimicrob. Chemother. 2022, 77, 2506–2515. [Google Scholar] [CrossRef]
  12. Robb, M.L.; Ananworanich, J. Lessons from acute HIV infection. Curr. Opin. HIV AIDS 2016, 11, 555–560. [Google Scholar] [CrossRef]
  13. Lore, K.; Smed-Sorensen, A.; Vasudevan, J.; Mascola, J.R.; Koup, R.A. Myeloid and plasmacytoid dendritic cells transfer HIV-1 preferentially to antigen-specific CD4+ T cells. J. Exp. Med. 2005, 201, 2023–2033. [Google Scholar] [CrossRef]
  14. Russell, R.A.; Martin, N.; Mitar, I.; Jones, E.; Sattentau, Q.J. Multiple proviral integration events after virological synapse-mediated HIV-1 spread. Virology 2013, 443, 143–149. [Google Scholar] [CrossRef]
  15. Veenhuis, R.T.; Abreu, C.M.; Costa, P.A.G.; Ferreira, E.A.; Ratliff, J.; Pohlenz, L.; Shirk, E.N.; Rubin, L.H.; Blankson, J.N.; Gama, L.; et al. Monocyte-derived macrophages contain persistent latent HIV reservoirs. Nat. Microbiol. 2023, 8, 833–844. [Google Scholar] [CrossRef] [PubMed]
  16. Altfeld, M.; Gale, M., Jr. Innate immunity against HIV-1 infection. Nat. Immunol. 2015, 16, 554–562. [Google Scholar] [CrossRef] [PubMed]
  17. Hu, J.; Gardner, M.B.; Miller, C.J. Simian immunodeficiency virus rapidly penetrates the cervicovaginal mucosa after intravaginal inoculation and infects intraepithelial dendritic cells. J. Virol. 2000, 74, 6087–6095. [Google Scholar] [CrossRef] [PubMed]
  18. McMichael, A.J.; Borrow, P.; Tomaras, G.D.; Goonetilleke, N.; Haynes, B.F. The immune response during acute HIV-1 infection: Clues for vaccine development. Nat. Rev. Immunol. 2010, 10, 11–23. [Google Scholar] [CrossRef]
  19. Cohen, M.S.; Shaw, G.M.; McMichael, A.J.; Haynes, B.F. Acute HIV-1 Infection. N. Engl. J. Med. 2011, 364, 1943–1954. [Google Scholar] [CrossRef]
  20. Keele, B.F.; Giorgi, E.E.; Salazar-Gonzalez, J.F.; Decker, J.M.; Pham, K.T.; Salazar, M.G.; Sun, C.; Grayson, T.; Wang, S.; Li, H.; et al. Identification and characterization of transmitted and early founder virus envelopes in primary HIV-1 infection. Proc. Natl. Acad. Sci. USA 2008, 105, 7552–7557. [Google Scholar] [CrossRef]
  21. Long, E.M.; Rainwater, S.M.; Lavreys, L.; Mandaliya, K.; Overbaugh, J. HIV type 1 variants transmitted to women in Kenya require the CCR5 coreceptor for entry, regardless of the genetic complexity of the infecting virus. AIDS Res. Hum. Retroviruses 2002, 18, 567–576. [Google Scholar] [CrossRef]
  22. Shepherd, J.C.; Jacobson, L.P.; Qiao, W.; Jamieson, B.D.; Phair, J.P.; Piazza, P.; Quinn, T.C.; Margolick, J.B. Emergence and persistence of CXCR4-tropic HIV-1 in a population of men from the multicenter AIDS cohort study. J. Infect. Dis. 2008, 198, 1104–1112. [Google Scholar] [CrossRef]
  23. Sucupira, M.C.; Sanabani, S.; Cortes, R.M.; Giret, M.T.; Tomiyama, H.; Sauer, M.M.; Sabino, E.C.; Janini, L.M.; Kallas, E.G.; Diaz, R.S. Faster HIV-1 disease progression among Brazilian individuals recently infected with CXCR4-utilizing strains. PLoS ONE 2012, 7, e30292. [Google Scholar] [CrossRef]
  24. Moyle, G.J.; Wildfire, A.; Mandalia, S.; Mayer, H.; Goodrich, J.; Whitcomb, J.; Gazzard, B.G. Epidemiology and predictive factors for chemokine receptor use in HIV-1 infection. J. Infect. Dis. 2005, 191, 866–872. [Google Scholar] [CrossRef]
  25. Frange, P.; Meyer, L.; Deveau, C.; Tran, L.; Goujard, C.; Ghosn, J.; Girard, P.M.; Morlat, P.; Rouzioux, C.; Chaix, M.L. Recent HIV-1 infection contributes to the viral diffusion over the French territory with a recent increasing frequency. PLoS ONE 2012, 7, e31695. [Google Scholar] [CrossRef] [PubMed]
  26. Castor, D.; Low, A.; Evering, T.; Karmon, S.; Davis, B.; Figueroa, A.; LaMar, M.; Garmon, D.; Mehandru, S.; Markowitz, M. Transmitted drug resistance and phylogenetic relationships among acute and early HIV-1-infected individuals in New York City. J. Acquir. Immune Defic. Syndr. 2012, 61, 1–8. [Google Scholar] [CrossRef] [PubMed]
  27. Cheret, A.; Bacchus, C.; Avettand-Fenoel, V.; Nembot, G.; Melard, A.; Blanc, C.; Lascoux-Combe, C.; Slama, L.; Allegre, T.; Allavena, C.; et al. A single HIV-1 cluster and a skewed immune homeostasis drive the early spread of HIV among resting CD4+ cell subsets within one month post-infection. PLoS ONE 2013, 8, e64219. [Google Scholar]
  28. Li, H.; Bar, K.J.; Wang, S.; Decker, J.M.; Chen, Y.; Sun, C.; Salazar-Gonzalez, J.F.; Salazar, M.G.; Learn, G.H.; Morgan, C.J.; et al. High Multiplicity Infection by HIV-1 in Men Who Have Sex with Men. PLoS Pathog. 2010, 6, e1000890. [Google Scholar] [CrossRef]
  29. Tully, D.C.; Ogilvie, C.B.; Batorsky, R.E.; Bean, D.J.; Power, K.A.; Ghebremichael, M.; Bedard, H.E.; Gladden, A.D.; Seese, A.M.; Amero, M.A.; et al. Differences in the Selection Bottleneck between Modes of Sexual Transmission Influence the Genetic Composition of the HIV-1 Founder Virus. PLoS Pathog. 2016, 12, e1005619. [Google Scholar] [CrossRef]
  30. Cheret, A.; Bacchus-Souffan, C.; Avettand-Fenoel, V.; Melard, A.; Nembot, G.; Blanc, C.; Samri, A.; Saez-Cirion, A.; Hocqueloux, L.; Lascoux-Combe, C.; et al. Combined ART started during acute HIV infection protects central memory CD4+ T cells and can induce remission. J. Antimicrob. Chemother. 2015, 70, 2108–2120. [Google Scholar] [CrossRef]
  31. Kariuki, S.M.; Selhorst, P.; Arien, K.K.; Dorfman, J.R. The HIV-1 transmission bottleneck. Retrovirology 2017, 14, 22. [Google Scholar] [CrossRef]
  32. Miller, W.C.; Rosenberg, N.E.; Rutstein, S.E.; Powers, K.A. Role of acute and early HIV infection in the sexual transmission of HIV. Curr. Opin. HIV AIDS 2010, 5, 277–282. [Google Scholar] [CrossRef]
  33. Chan, P.; Spudich, S. Central Nervous System Effects of Early HIV Infection and Consequences of Antiretroviral Therapy Initiation during Acute HIV. Viruses 2024, 16, 1082. [Google Scholar] [CrossRef]
  34. Haase, A.T. Population biology of HIV-1 infection: Viral and CD4+ T cell demographics and dynamics in lymphatic tissues. Annu. Rev. Immunol. 1999, 17, 625–656. [Google Scholar] [CrossRef]
  35. Robb, M.L.; Eller, L.A.; Kibuuka, H.; Rono, K.; Maganga, L.; Nitayaphan, S.; Kroon, E.; Sawe, F.K.; Sinei, S.; Sriplienchan, S.; et al. Prospective Study of Acute HIV-1 Infection in Adults in East Africa and Thailand. N. Engl. J. Med. 2016, 374, 2120–2130. [Google Scholar] [CrossRef] [PubMed]
  36. Kelley, C.F.; Hernandez-Ramos, I.; Franco-Paredes, C.; del Rio, C. Clinical, epidemiologic characteristics of foreign-born Latinos with HIV/AIDS at an urban HIV clinic. AIDS Read. 2007, 17, 73–74, 78–80, 85–88. [Google Scholar] [PubMed]
  37. Ndhlovu, Z.M.; Kamya, P.; Mewalal, N.; Kloverpris, H.N.; Nkosi, T.; Pretorius, K.; Laher, F.; Ogunshola, F.; Chopera, D.; Shekhar, K.; et al. Magnitude and Kinetics of CD8+ T Cell Activation during Hyperacute HIV Infection Impact Viral Set Point. Immunity 2015, 43, 591–604. [Google Scholar] [CrossRef] [PubMed]
  38. Selhorst, P.; Combrinck, C.; Ndabambi, N.; Ismail, S.D.; Abrahams, M.R.; Lacerda, M.; Samsunder, N.; Garrett, N.; Abdool Karim, Q.; Abdool Karim, S.S.; et al. Replication Capacity of Viruses from Acute Infection Drives Hiv-1 Disease Progression. J. Virol. 2017, 91, 10–1128. [Google Scholar] [CrossRef]
  39. Abreu, C.M.; Veenhuis, R.T.; Avalos, C.R.; Graham, S.; Parrilla, D.R.; Ferreira, E.A.; Queen, S.E.; Shirk, E.N.; Bullock, B.T.; Li, M.; et al. Myeloid and CD4 T Cells Comprise the Latent Reservoir in Antiretroviral Therapy-Suppressed SIVmac251-Infected Macaques. mBio 2019, 10. [Google Scholar] [CrossRef]
  40. Ananworanich, J.; Sacdalan, C.P.; Pinyakorn, S.; Chomont, N.; de Souza, M.; Luekasemsuk, T.; Schuetz, A.; Krebs, S.J.; Dewar, R.; Jagodzinski, L.; et al. Virological and immunological characteristics of HIV-infected individuals at the earliest stage of infection. J. Virus Erad. 2016, 2, 43–48. [Google Scholar] [CrossRef]
  41. 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]
  42. Chun, T.W.; Engel, D.; Berrey, M.M.; Shea, T.; Corey, L.; Fauci, A.S. Early establishment of a pool of latently infected, resting CD4(+) T cells during primary HIV-1 infection. Proc. Natl. Acad. Sci. USA 1998, 95, 8869–8873. [Google Scholar] [CrossRef]
  43. Avettand-Fenoel, V.; Hocqueloux, L.; Ghosn, J.; Cheret, A.; Frange, P.; Melard, A.; Viard, J.P.; Rouzioux, C. Total HIV-1 DNA, a Marker of Viral Reservoir Dynamics with Clinical Implications. Clin. Microbiol. Rev. 2016, 29, 859–880. [Google Scholar] [CrossRef]
  44. Kim, C.J.; McKinnon, L.R.; Kovacs, C.; Kandel, G.; Huibner, S.; Chege, D.; Shahabi, K.; Benko, E.; Loutfy, M.; Ostrowski, M.; et al. Mucosal Th17 cell function is altered during HIV infection and is an independent predictor of systemic immune activation. J. Immunol. 2013, 191, 2164–2173. [Google Scholar] [CrossRef]
  45. Yukl, S.A.; Shergill, A.K.; Ho, T.; Killian, M.; Girling, V.; Epling, L.; Li, P.; Wong, L.K.; Crouch, P.; Deeks, S.G.; et al. The distribution of HIV DNA and RNA in cell subsets differs in gut and blood of HIV-positive patients on ART: Implications for viral persistence. J. Infect. Dis. 2013, 208, 1212–1220. [Google Scholar] [CrossRef] [PubMed]
  46. Valcour, V.; Chalermchai, T.; Sailasuta, N.; Marovich, M.; Lerdlum, S.; Suttichom, D.; Suwanwela, N.C.; Jagodzinski, L.; Michael, N.; Spudich, S.; et al. Central nervous system viral invasion and inflammation during acute HIV infection. J. Infect. Dis. 2012, 206, 275–282. [Google Scholar] [CrossRef] [PubMed]
  47. Cheret, A.; Durier, C.; Melard, A.; Ploquin, M.; Heitzmann, J.; Lecuroux, C.; Avettand-Fenoel, V.; David, L.; Pialoux, G.; Chennebault, J.M.; et al. Impact of early cART on HIV blood and semen compartments at the time of primary infection. PLoS ONE 2017, 12, e0180191. [Google Scholar] [CrossRef] [PubMed]
  48. Mariaggi, A.A.; Bauer, R.; Charre, C.; Gardiennet, E.; Meiffredy, V.; Ajana, F.; Lacombe, K.; Pialoux, G.; Cua, E.; Rouzioux, C.; et al. HIV-1-RNA and total HIV-1-DNA loads in the genital compartment in men receiving dolutegravir- versus darunavir-based combined ART (cART) regimens during primary HIV infection. J. Antimicrob. Chemother. 2022, 77, 735–739. [Google Scholar] [CrossRef]
  49. Goujard, C.; Bonarek, M.; Meyer, L.; Bonnet, F.; Chaix, M.L.; Deveau, C.; Sinet, M.; Galimand, J.; Delfraissy, J.F.; Venet, A.; et al. CD4 cell count and HIV DNA level are independent predictors of disease progression after primary HIV type 1 infection in untreated patients. Clin. Infect. Dis. 2006, 42, 709–715. [Google Scholar] [CrossRef]
  50. Tremeaux, P.; Lenfant, T.; Boufassa, F.; Essat, A.; Melard, A.; Gousset, M.; Delelis, O.; Viard, J.P.; Bary, M.; Goujard, C.; et al. Increasing contribution of integrated forms to total HIV DNA in blood during HIV disease progression from primary infection. EBioMedicine 2019, 41, 455–464. [Google Scholar] [CrossRef]
  51. Sallusto, F.; Geginat, J.; Lanzavecchia, A. Central memory and effector memory T cell subsets: Function, generation, and maintenance. Annu. Rev. Immunol. 2004, 22, 745–763. [Google Scholar] [CrossRef]
  52. Appay, V.; Douek, D.C.; Price, D.A. CD8+ T cell efficacy in vaccination and disease. Nat. Med. 2008, 14, 623–628. [Google Scholar] [CrossRef]
  53. Ganesan, A.; Chattopadhyay, P.K.; Brodie, T.M.; Qin, J.; Gu, W.; Mascola, J.R.; Michael, N.L.; Follmann, D.A.; Roederer, M. Immunologic and virologic events in early HIV infection predict subsequent rate of progression. J. Infect. Dis. 2010, 201, 272–284. [Google Scholar] [CrossRef]
  54. Buzon, M.J.; Sun, H.; Li, C.; Shaw, A.; Seiss, K.; Ouyang, Z.; Martin-Gayo, E.; Leng, J.; Henrich, T.J.; Li, J.Z.; et al. HIV-1 persistence in CD4+ T cells with stem cell-like properties. Nat. Med. 2014, 20, 139–142. [Google Scholar] [CrossRef]
  55. Gattinoni, L.; Speiser, D.E.; Lichterfeld, M.; Bonini, C. T memory stem cells in health and disease. Nat. Med. 2017, 23, 18–27. [Google Scholar] [CrossRef] [PubMed]
  56. Chomont, N.; El-Far, M.; Ancuta, P.; Trautmann, L.; Procopio, F.A.; Yassine-Diab, B.; Boucher, G.; Boulassel, M.R.; Ghattas, G.; Brenchley, J.M.; et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat. Med. 2009, 15, 893–900. [Google Scholar] [CrossRef] [PubMed]
  57. Palmer, S.; Maldarelli, F.; Wiegand, A.; Bernstein, B.; Hanna, G.J.; Brun, S.C.; Kempf, D.J.; Mellors, J.W.; Coffin, J.M.; King, M.S. Low-level viremia persists for at least 7 years in patients on suppressive antiretroviral therapy. Proc. Natl. Acad. Sci. USA 2008, 105, 3879–3884. [Google Scholar] [CrossRef] [PubMed]
  58. McNab, F.; Mayer-Barber, K.; Sher, A.; Wack, A.; O’Garra, A. Type I interferons in infectious disease. Nat. Rev. Immunol. 2015, 15, 87–103. [Google Scholar] [CrossRef]
  59. Rustagi, A.; Gale, M., Jr. Innate antiviral immune signaling, viral evasion and modulation by HIV-1. J. Mol. Biol. 2014, 426, 1161–1177. [Google Scholar] [CrossRef]
  60. Arango Duque, G.; Descoteaux, A. Macrophage cytokines: Involvement in immunity and infectious diseases. Front. Immunol. 2014, 5, 491. [Google Scholar] [CrossRef]
  61. Roberts, L.; Passmore, J.A.; Williamson, C.; Little, F.; Bebell, L.M.; Mlisana, K.; Burgers, W.A.; van Loggerenberg, F.; Walzl, G.; Djoba Siawaya, J.F.; et al. Plasma cytokine levels during acute HIV-1 infection predict HIV disease progression. AIDS 2010, 24, 819–831. [Google Scholar] [CrossRef]
  62. Stacey, A.R.; Norris, P.J.; Qin, L.; Haygreen, E.A.; Taylor, E.; Heitman, J.; Lebedeva, M.; DeCamp, A.; Li, D.; Grove, D.; et al. Induction of a striking systemic cytokine cascade prior to peak viremia in acute human immunodeficiency virus type 1 infection, in contrast to more modest and delayed responses in acute hepatitis B and C virus infections. J. Virol. 2009, 83, 3719–3733. [Google Scholar] [CrossRef]
  63. Naranbhai, V.; Abdool Karim, S.S.; Altfeld, M.; Samsunder, N.; Durgiah, R.; Sibeko, S.; Abdool Karim, Q.; Carr, W.H. Innate immune activation enhances hiv acquisition in women, diminishing the effectiveness of tenofovir microbicide gel. J. Infect. Dis. 2012, 206, 993–1001. [Google Scholar] [CrossRef]
  64. Scott-Algara, D.; Truong, L.X.; Versmisse, P.; David, A.; Luong, T.T.; Nguyen, N.V.; Theodorou, I.; Barre-Sinoussi, F.; Pancino, G. Cutting edge: Increased NK cell activity in HIV-1-exposed but uninfected Vietnamese intravascular drug users. J. Immunol. 2003, 171, 5663–5667. [Google Scholar] [CrossRef]
  65. Borrow, P. Innate immunity in acute HIV-1 infection. Curr. Opin. HIV AIDS 2011, 6, 353–363. [Google Scholar] [CrossRef]
  66. Doisne, J.M.; Urrutia, A.; Lacabaratz-Porret, C.; Goujard, C.; Meyer, L.; Chaix, M.L.; Sinet, M.; Venet, A. CD8+ T cells specific for EBV, cytomegalovirus, and influenza virus are activated during primary HIV infection. J. Immunol. 2004, 173, 2410–2418. [Google Scholar] [CrossRef] [PubMed]
  67. Finkel, T.H.; Tudor-Williams, G.; Banda, N.K.; Cotton, M.F.; Curiel, T.; Monks, C.; Baba, T.W.; Ruprecht, R.M.; Kupfer, A. Apoptosis occurs predominantly in bystander cells and not in productively infected cells of HIV- and SIV-infected lymph nodes. Nat. Med. 1995, 1, 129–134. [Google Scholar] [CrossRef] [PubMed]
  68. Peng, X.; Ouyang, J.; Isnard, S.; Lin, J.; Fombuena, B.; Zhu, B.; Routy, J.P. Sharing CD4+ T Cell Loss: When COVID-19 and HIV Collide on Immune System. Front. Immunol. 2020, 11, 596631. [Google Scholar] [CrossRef] [PubMed]
  69. Brenchley, J.M.; Price, D.A.; Douek, D.C. HIV disease: Fallout from a mucosal catastrophe? Nat. Immunol. 2006, 7, 235–239. [Google Scholar] [CrossRef]
  70. Lafeuillade, A.; Poggi, C.; Tamalet, C.; Profizi, N. Human immunodeficiency virus type 1 dynamics in different lymphoid tissue compartments. J. Infect. Dis. 1997, 176, 804–806. [Google Scholar] [CrossRef]
  71. Estes, J.D. Pathobiology of HIV/SIV-associated changes in secondary lymphoid tissues. Immunol. Rev. 2013, 254, 65–77. [Google Scholar] [CrossRef]
  72. Chevalier, M.F.; Didier, C.; Girard, P.M.; Manea, M.E.; Campa, P.; Barre-Sinoussi, F.; Scott-Algara, D.; Weiss, L. CD4 T-Cell Responses in Primary HIV Infection: Interrelationship with Immune Activation and Virus Burden. Front. Immunol. 2016, 7, 395. [Google Scholar] [CrossRef]
  73. Klatt, N.R.; Funderburg, N.T.; Brenchley, J.M. Microbial translocation, immune activation, and HIV disease. Trends Microbiol. 2013, 21, 6–13. [Google Scholar] [CrossRef]
  74. Hoenigl, M.; Perez-Santiago, J.; Nakazawa, M.; de Oliveira, M.F.; Zhang, Y.; Finkelman, M.A.; Letendre, S.; Smith, D.; Gianella, S. (1-->3)-beta-d-Glucan: A Biomarker for Microbial Translocation in Individuals with Acute or Early HIV Infection? Front. Immunol. 2016, 7, 404. [Google Scholar] [CrossRef]
  75. Gong, Y.; Zhang, H.; Feng, J.; Ying, L.; Ji, M.; Wei, S.; Ma, Q. Time-restricted feeding improves metabolic syndrome by activating thermogenesis in brown adipose tissue and reducing inflammatory markers. Front. Immunol. 2025, 16, 1501850. [Google Scholar] [CrossRef] [PubMed]
  76. Koup, R.A.; Safrit, J.T.; Cao, Y.; Andrews, C.A.; McLeod, G.; Borkowsky, W.; Farthing, C.; Ho, D.D. Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J. Virol. 1994, 68, 4650–4655. [Google Scholar] [CrossRef] [PubMed]
  77. Pantaleo, G.; Demarest, J.F.; Soudeyns, H.; Graziosi, C.; Denis, F.; Adelsberger, J.W.; Borrow, P.; Saag, M.S.; Shaw, G.M.; Sekaly, R.P.; et al. Major expansion of CD8+ T cells with a predominant V beta usage during the primary immune response to HIV. Nature 1994, 370, 463–467. [Google Scholar] [CrossRef] [PubMed]
  78. Goonetilleke, N.; Liu, M.K.; Salazar-Gonzalez, J.F.; Ferrari, G.; Giorgi, E.; Ganusov, V.V.; Keele, B.F.; Learn, G.H.; Turnbull, E.L.; Salazar, M.G.; et al. The first T cell response to transmitted/founder virus contributes to the control of acute viremia in HIV-1 infection. J. Exp. Med. 2009, 206, 1253–1272. [Google Scholar] [CrossRef]
  79. Saez-Cirion, A.; Lacabaratz, C.; Lambotte, O.; Versmisse, P.; Urrutia, A.; Boufassa, F.; Barre-Sinoussi, F.; Delfraissy, J.F.; Sinet, M.; Pancino, G.; et al. HIV controllers exhibit potent CD8 T cell capacity to suppress HIV infection ex vivo and peculiar cytotoxic T lymphocyte activation phenotype. Proc. Natl. Acad. Sci. USA 2007, 104, 6776–6781. [Google Scholar] [CrossRef]
  80. Cocchi, F.; DeVico, A.L.; Garzino-Demo, A.; Arya, S.K.; Gallo, R.C.; Lusso, P. Identification of RANTES, MIP-1 alpha, and MIP-1 beta as the major HIV-suppressive factors produced by CD8+ T cells. Science 1995, 270, 1811–1815. [Google Scholar] [CrossRef]
  81. Walker, B.D.; Yu, X.G. Unravelling the mechanisms of durable control of HIV-1. Nat. Rev. Immunol. 2013, 13, 487–498. [Google Scholar] [CrossRef]
  82. Emu, B.; Sinclair, E.; Hatano, H.; Ferre, A.; Shacklett, B.; Martin, J.N.; McCune, J.M.; Deeks, S.G. HLA class I-restricted T-cell responses may contribute to the control of human immunodeficiency virus infection, but such responses are not always necessary for long-term virus control. J. Virol. 2008, 82, 5398–5407. [Google Scholar] [CrossRef]
  83. Altfeld, M.; Kalife, E.T.; Qi, Y.; Streeck, H.; Lichterfeld, M.; Johnston, M.N.; Burgett, N.; Swartz, M.E.; Yang, A.; Alter, G.; et al. HLA Alleles Associated with Delayed Progression to AIDS Contribute Strongly to the Initial CD8(+) T Cell Response against HIV-1. PLoS Med. 2006, 3, e403. [Google Scholar] [CrossRef]
  84. Demers, K.R.; Makedonas, G.; Buggert, M.; Eller, M.A.; Ratcliffe, S.J.; Goonetilleke, N.; Li, C.K.; Eller, L.A.; Rono, K.; Maganga, L.; et al. Temporal Dynamics of CD8+ T Cell Effector Responses during Primary HIV Infection. PLoS Pathog. 2016, 12, e1005805. [Google Scholar] [CrossRef]
  85. Roberts, E.R.; Carnathan, D.G.; Li, H.; Shaw, G.M.; Silvestri, G.; Betts, M.R. Collapse of Cytolytic Potential in SIV-Specific CD8+ T Cells Following Acute SIV Infection in Rhesus Macaques. PLoS Pathog. 2016, 12, e1006135. [Google Scholar] [CrossRef] [PubMed]
  86. Florez-Alvarez, L.; Hernandez, J.C.; Zapata, W. NK Cells in HIV-1 Infection: From Basic Science to Vaccine Strategies. Front. Immunol. 2018, 9, 2290. [Google Scholar] [CrossRef] [PubMed]
  87. Pohlmeyer, C.W.; Gonzalez, V.D.; Irrinki, A.; Ramirez, R.N.; Li, L.; Mulato, A.; Murry, J.P.; Arvey, A.; Hoh, R.; Deeks, S.G.; et al. Identification of NK Cell Subpopulations That Differentiate HIV-Infected Subject Cohorts with Diverse Levels of Virus Control. J. Virol. 2019, 93. [Google Scholar] [CrossRef] [PubMed]
  88. Sant, A.J.; McMichael, A. Revealing the role of CD4(+) T cells in viral immunity. J. Exp. Med. 2012, 209, 1391–1395. [Google Scholar] [CrossRef]
  89. Schuetz, A.; Deleage, C.; Sereti, I.; Rerknimitr, R.; Phanuphak, N.; Phuang-Ngern, Y.; Estes, J.D.; Sandler, N.G.; Sukhumvittaya, S.; Marovich, M.; et al. Initiation of ART during early acute HIV infection preserves mucosal Th17 function and reverses HIV-related immune activation. Pathogens 2014, 10, e1004543. [Google Scholar] [CrossRef]
  90. Mattapallil, J.J.; Douek, D.C.; Hill, B.; Nishimura, Y.; Martin, M.; Roederer, M. Massive infection and loss of memory CD4+ T cells in multiple tissues during acute SIV infection. Nature 2005, 434, 1093–1097. [Google Scholar] [CrossRef]
  91. Schieffer, M.; Jessen, H.K.; Oster, A.F.; Pissani, F.; Soghoian, D.Z.; Lu, R.; Jessen, A.B.; Zedlack, C.; Schultz, B.T.; Davis, I.; et al. Induction of Gag-specific CD4 T cell responses during acute HIV infection is associated with improved viral control. J. Virol. 2014, 88, 7357–7366. [Google Scholar] [CrossRef]
  92. Soghoian, D.Z.; Jessen, H.; Flanders, M.; Sierra-Davidson, K.; Cutler, S.; Pertel, T.; Ranasinghe, S.; Lindqvist, M.; Davis, I.; Lane, K.; et al. HIV-specific cytolytic CD4 T cell responses during acute HIV infection predict disease outcome. Sci. Transl. Med. 2012, 4, 123ra25. [Google Scholar] [CrossRef]
  93. Frater, J.; Ewings, F.; Hurst, J.; Brown, H.; Robinson, N.; Fidler, S.; Babiker, A.; Weber, J.; Porter, K.; Phillips, R.E. HIV-1-specific CD4(+) responses in primary HIV-1 infection predict disease progression. AIDS 2014, 28, 699–708. [Google Scholar] [CrossRef]
  94. Potter, S.J.; Lacabaratz, C.; Lambotte, O.; Perez-Patrigeon, S.; Vingert, B.; Sinet, M.; Colle, J.H.; Urrutia, A.; Scott-Algara, D.; Boufassa, F.; et al. Preserved central memory and activated effector memory CD4+ T-cell subsets in human immunodeficiency virus controllers: An ANRS EP36 study. J. Virol. 2007, 81, 13904–13915. [Google Scholar] [CrossRef]
  95. 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] [PubMed]
  96. Hellerstein, M.K.; Hoh, R.A.; Hanley, M.B.; Cesar, D.; Lee, D.; Neese, R.A.; McCune, J.M. Subpopulations of long-lived and short-lived T cells in advanced HIV-1 infection. J. Clin. Investig. 2003, 112, 956–966. [Google Scholar] [CrossRef] [PubMed]
  97. Margolick, J.B.; Gange, S.J.; Detels, R.; O’Gorman, M.R.; Rinaldo, C.R., Jr.; Lai, S. Impact of inversion of the CD4/CD8 ratio on the natural history of HIV-1 infection. J. Acquir. Immune Defic. Syndr. 2006, 42, 620–626. [Google Scholar] [CrossRef] [PubMed]
  98. Chapon, M.; Randriamampita, C.; Maubec, E.; Badoual, C.; Fouquet, S.; Wang, S.F.; Marinho, E.; Farhi, D.; Garcette, M.; Jacobelli, S.; et al. Progressive upregulation of PD-1 in primary and metastatic melanomas associated with blunted TCR signaling in infiltrating T lymphocytes. J. Investig. Dermatol. 2011, 131, 1300–1307. [Google Scholar] [CrossRef]
  99. Palmer, B.E.; Neff, C.P.; Lecureux, J.; Ehler, A.; Dsouza, M.; Remling-Mulder, L.; Korman, A.J.; Fontenot, A.P.; Akkina, R. In vivo blockade of the PD-1 receptor suppresses HIV-1 viral loads and improves CD4+ T cell levels in humanized mice. J. Immunol. 2013, 190, 211–219. [Google Scholar] [CrossRef]
  100. Autran, B.; Hadida, F.; Haas, G. Evolution and plasticity of CTL responses against HIV. Curr. Opin. Immunol. 1996, 8, 546–553. [Google Scholar] [CrossRef]
  101. Moir, S.; Buckner, C.M.; Ho, J.; Wang, W.; Chen, J.; Waldner, A.J.; Posada, J.G.; Kardava, L.; O’Shea, M.A.; Kottilil, S.; et al. B cells in early and chronic HIV infection: Evidence for preservation of immune function associated with early initiation of antiretroviral therapy. Blood 2010, 116, 5571–5579. [Google Scholar] [CrossRef]
  102. Levesque, M.C.; Moody, M.A.; Hwang, K.K.; Marshall, D.J.; Whitesides, J.F.; Amos, J.D.; Gurley, T.C.; Allgood, S.; Haynes, B.B.; Vandergrift, N.A.; et al. Polyclonal B cell differentiation and loss of gastrointestinal tract germinal centers in the earliest stages of HIV-1 infection. PLoS Med. 2009, 6, e1000107. [Google Scholar] [CrossRef]
  103. Lane, H.C.; Masur, H.; Edgar, L.C.; Whalen, G.; Rook, A.H.; Fauci, A.S. Abnormalities of B-cell activation and immunoregulation in patients with the acquired immunodeficiency syndrome. N. Engl. J. Med. 1983, 309, 453–458. [Google Scholar] [CrossRef]
  104. Hart, M.; Steel, A.; Clark, S.A.; Moyle, G.; Nelson, M.; Henderson, D.C.; Wilson, R.; Gotch, F.; Gazzard, B.; Kelleher, P. Loss of discrete memory B cell subsets is associated with impaired immunization responses in HIV-1 infection and may be a risk factor for invasive pneumococcal disease. J. Immunol. 2007, 178, 8212–8220. [Google Scholar] [CrossRef]
  105. Moir, S.; Fauci, A.S. B cells in HIV infection and disease. Nat. Rev. Immunol. 2009, 9, 235–245. [Google Scholar] [CrossRef] [PubMed]
  106. Wei, X.; Decker, J.M.; Wang, S.; Hui, H.; Kappes, J.C.; Wu, X.; Salazar-Gonzalez, J.F.; Salazar, M.G.; Kilby, J.M.; Saag, M.S.; et al. Antibody neutralization and escape by HIV-1. Nature 2003, 422, 307–312. [Google Scholar] [CrossRef] [PubMed]
  107. Klatt, N.R.; Chomont, N.; Douek, D.C.; Deeks, S.G. Immune activation and HIV persistence: Implications for curative approaches to HIV infection. Immunol. Rev. 2013, 254, 326–342. [Google Scholar] [CrossRef] [PubMed]
  108. Braun, D.L.; Kouyos, R.D.; Balmer, B.; Grube, C.; Weber, R.; Gunthard, H.F. Frequency and Spectrum of Unexpected Clinical Manifestations of Primary HIV-1 Infection. Clin. Infect. Dis. 2015, 61, 1013–1021. [Google Scholar] [CrossRef]
  109. Schacker, T.; Collier, A.C.; Hughes, J.; Shea, T.; Corey, L. Clinical and epidemiologic features of primary HIV infection. Ann. Intern. Med. 1996, 125, 257–264. [Google Scholar] [CrossRef]
  110. Lievre, L.; Deveau, C.; Gerbe, J.; Enel, P.; Tran, L.; De Castro, N.; Costagliola, D.; Meyer, L.; Primo Study, G.; Clinical Epidemieology, G. Yearly number of patients diagnosed with primary HIV-1 infection in France estimated by a capture-recapture approach. AIDS 2006, 20, 2392–2395. [Google Scholar] [CrossRef]
  111. Kahn, J.O.; Walker, B.D. Acute human immunodeficiency virus type 1 infection. N. Engl. J. Med. 1998, 339, 33–39. [Google Scholar] [CrossRef]
  112. Boufassa, F.; Bachmeyer, C.; Carre, N.; Deveau, C.; Persoz, A.; Jadand, C.; Sereni, D.; Bucquet, D. Influence of neurologic manifestations of primary human immunodeficiency virus infection on disease progression. SEROCO Study Group. J. Infect. Dis. 1995, 171, 1190–1195. [Google Scholar] [CrossRef]
  113. Crowell, T.A.; Colby, D.J.; Pinyakorn, S.; Fletcher, J.L.K.; Kroon, E.; Schuetz, A.; Krebs, S.J.; Slike, B.M.; Leyre, L.; Chomont, N.; et al. Acute Retroviral Syndrome Is Associated With High Viral Burden, CD4 Depletion, and Immune Activation in Systemic and Tissue Compartments. Clin. Infect. Dis. 2018, 66, 1540–1549. [Google Scholar] [CrossRef]
  114. Hoenigl, M.; Green, N.; Camacho, M.; Gianella, S.; Mehta, S.R.; Smith, D.M.; Little, S.J. Signs or Symptoms of Acute HIV Infection in a Cohort Undergoing Community-Based Screening. Emerg. Infect. Dis. 2016, 22, 532–534. [Google Scholar] [CrossRef]
  115. Kelley, C.F.; Barbour, J.D.; Hecht, F.M. The relation between symptoms, viral load, and viral load set point in primary HIV infection. J. Acquir. Immune Defic. Syndr. 2007, 45, 445–448. [Google Scholar] [CrossRef] [PubMed]
  116. Ghosn, J.; Deveau, C.; Chaix, M.L.; Goujard, C.; Galimand, J.; Zitoun, Y.; Allegre, T.; Delfraissy, J.F.; Meyer, L.; Rouzioux, C. Despite being highly diverse, immunovirological status strongly correlates with clinical symptoms during primary HIV-1 infection: A cross-sectional study based on 674 patients enrolled in the ANRS CO 06 PRIMO cohort. J. Antimicrob. Chemother. 2010, 65, 741–748. [Google Scholar] [CrossRef] [PubMed]
  117. Abdel Babiker, S.D.; Daniela De Angelis, D.E. Kholoud Porter, Time from HIV-1 seroconversion to AIDS and death before widespread use of highly-active antiretroviral therapy: A collaborative re-analysis. Collaborative Group on AIDS Incubation and HIV Survival including the CASCADE EU Concerted Action. Concerted Action on SeroConversion to AIDS and Death in Europe. Lancet 2000, 355, 1131–1137. [Google Scholar]
  118. Braun, D.L.; Kouyos, R.; Oberle, C.; Grube, C.; Joos, B.; Fellay, J.; McLaren, P.J.; Kuster, H.; Gunthard, H.F. A novel Acute Retroviral Syndrome Severity Score predicts the key surrogate markers for HIV-1 disease progression. PLoS ONE 2014, 9, e114111. [Google Scholar] [CrossRef]
  119. Avettand-Fenoel, V.; Chaix, M.L.; Blanche, S.; Burgard, M.; Floch, C.; Toure, K.; Allemon, M.C.; Warszawski, J.; Rouzioux, C. LTR real-time PCR for HIV-1 DNA quantitation in blood cells for early diagnosis in infants born to seropositive mothers treated in HAART area (ANRS CO 01). J. Med. Virol. 2009, 81, 217–223. [Google Scholar]
  120. Fiebig, E.W.; Wright, D.J.; Rawal, B.D.; Garrett, P.E.; Schumacher, R.T.; Peddada, L.; Heldebrant, C.; Smith, R.; Conrad, A.; Kleinman, S.H.; et al. Dynamics of HIV viremia and antibody seroconversion in plasma donors: Implications for diagnosis and staging of primary HIV infection. AIDS 2003, 17, 1871–1879. [Google Scholar] [CrossRef]
  121. Ananworanich, J.; Fletcher, J.L.; Pinyakorn, S.; van Griensven, F.; Vandergeeten, C.; Schuetz, A.; Pankam, T.; Trichavaroj, R.; Akapirat, S.; Chomchey, N.; et al. A novel acute HIV infection staging system based on 4th generation immunoassay. Retrovirology 2013, 10, 56. [Google Scholar] [CrossRef]
  122. Morlat, P. Prise en Charge Médicale des Personnes Vivant avec le VIH, Recommandations du Groupe d’Experts sous la Direction du Professeur P. Morlat; Conseil National du Sida et des Hépatites Virales: Paris, France, 2013; 476p. [Google Scholar]
  123. Mariaggi, A.A.; Gardiennet, E.; Stefic, K.; Essat, A.; Cheret, A.; Goujard, C.; Meyer, L.; Barin, F.; Avettand-Fenoel, V.; Cohort, A.P. Immunoblots may not be effective in confirming the recency of HIV-1 infection. J. Virol. Methods 2021, 290, 114074. [Google Scholar] [CrossRef]
  124. Pavie, J.; Rachline, A.; Loze, B.; Niedbalski, L.; Delaugerre, C.; Laforgerie, E.; Plantier, J.C.; Rozenbaum, W.; Chevret, S.; Molina, J.M.; et al. Sensitivity of five rapid HIV tests on oral fluid or finger-stick whole blood: A real-time comparison in a healthcare setting. PLoS ONE 2010, 5, e11581. [Google Scholar] [CrossRef]
  125. Stekler, J.D.; Tapia, K.; Maenza, J.; Stevens, C.E.; Ure, G.A.; O’Neal, J.D.; Lane, A.; Mullins, J.I.; Coombs, R.W.; Holte, S.; et al. No Time to Delay! Fiebig Stages and Referral in Acute HIV infection: Seattle Primary Infection Program Experience. AIDS Res. Hum. Retroviruses 2018, 34, 657–666. [Google Scholar] [CrossRef]
  126. Powers, K.A.; Ghani, A.C.; Miller, W.C.; Hoffman, I.F.; Pettifor, A.E.; Kamanga, G.; Martinson, F.E.; Cohen, M.S. The role of acute and early HIV infection in the spread of HIV and implications for transmission prevention strategies in Lilongwe, Malawi: A modelling study. Lancet 2011, 378, 256–268. [Google Scholar] [CrossRef] [PubMed]
  127. Seng, R.; Rolland, M.; Beck-Wirth, G.; Souala, F.; Deveau, C.; Delfraissy, J.F.; Goujard, C.; Meyer, L. Trends in unsafe sex and influence of viral load among patients followed since primary HIV infection, 2000–2009. AIDS 2011, 25, 977–988. [Google Scholar] [CrossRef] [PubMed]
  128. Cheret, A.; Nembot, G.; Melard, A.; Lascoux, C.; Slama, L.; Miailhes, P.; Yeni, P.; Abel, S.; Avettand-Fenoel, V.; Venet, A.; et al. Intensive five-drug antiretroviral therapy regimen versus standard triple-drug therapy during primary HIV-1 infection (OPTIPRIM-ANRS 147): A randomised, open-label, phase 3 trial. Lancet Infect. Dis. 2015, 15, 387–396. [Google Scholar] [CrossRef] [PubMed]
  129. Fidler, S.; Porter, K.; Ewings, F.; Frater, J.; Ramjee, G.; Cooper, D.; Rees, H.; Fisher, M.; Schechter, M.; Kaleebu, P.; et al. Short-course antiretroviral therapy in primary HIV infection. N. Engl. J. Med. 2013, 368, 207–217. [Google Scholar]
  130. Yerly, S.; Kaiser, L.; Perneger, T.V.; Cone, R.W.; Opravil, M.; Chave, J.P.; Furrer, H.; Hirschel, B.; Perrin, L. Time of initiation of antiretroviral therapy: Impact on HIV-1 viraemia. The Swiss HIV Cohort Study. AIDS 2000, 14, 243–249. [Google Scholar] [CrossRef]
  131. Grijsen, M.L.; Steingrover, R.; Wit, F.W.; Jurriaans, S.; Verbon, A.; Brinkman, K.; van der Ende, M.E.; Soetekouw, R.; de Wolf, F.; Lange, J.M.; et al. No treatment versus 24 or 60 weeks of antiretroviral treatment during primary HIV infection: The randomized Primo-SHM trial. PLoS Med. 2012, 9, e1001196. [Google Scholar] [CrossRef]
  132. Yerly, S.; Perneger, T.V.; Vora, S.; Hirschel, B.; Perrin, L. Decay of cell-associated HIV-1 DNA correlates with residual replication in patients treated during acute HIV-1 infection. AIDS 2000, 14, 2805–2812. [Google Scholar] [CrossRef]
  133. Ananworanich, J.; Schuetz, A.; Vandergeeten, C.; Sereti, I.; de Souza, M.; Rerknimitr, R.; Dewar, R.; Marovich, M.; van Griensven, F.; Sekaly, R.; et al. Impact of multi-targeted antiretroviral treatment on gut T cell depletion and HIV reservoir seeding during acute HIV infection. PLoS ONE 2012, 7, e33948. [Google Scholar] [CrossRef]
  134. Lampinen, T.M.; Critchlow, C.W.; Kuypers, J.M.; Hurt, C.S.; Nelson, P.J.; Hawes, S.E.; Coombs, R.W.; Holmes, K.K.; Kiviat, N.B. Association of antiretroviral therapy with detection of HIV-1 RNA and DNA in the anorectal mucosa of homosexual men. AIDS 2000, 14, F69–F75. [Google Scholar] [CrossRef]
  135. Belmonte, L.; Olmos, M.; Fanin, A.; Parodi, C.; Bare, P.; Concetti, H.; Perez, H.; de Bracco, M.M.; Cahn, P. The intestinal mucosa as a reservoir of HIV-1 infection after successful HAART. AIDS 2007, 21, 2106–2108. [Google Scholar] [CrossRef]
  136. Laanani, M.; Ghosn, J.; Essat, A.; Melard, A.; Seng, R.; Gousset, M.; Panjo, H.; Mortier, E.; Girard, P.M.; Goujard, C.; et al. Impact of the Timing of Initiation of Antiretroviral Therapy During Primary HIV-1 Infection on the Decay of Cell-Associated HIV-DNA. Clin Infect Dis 2015, 60, 1715–1721. [Google Scholar] [CrossRef] [PubMed]
  137. Mexas, A.M.; Graf, E.H.; Pace, M.J.; Yu, J.J.; Papasavvas, E.; Azzoni, L.; Busch, M.P.; Di Mascio, M.; Foulkes, A.S.; Migueles, S.A.; et al. Concurrent measures of total and integrated HIV DNA monitor reservoirs and ongoing replication in eradication trials. AIDS 2012, 26, 2295–2306. [Google Scholar] [CrossRef] [PubMed]
  138. Li, J.Z.; Gandhi, R.T. The sooner, the better: More evidence that early antiretroviral therapy lowers viral reservoirs in HIV-infected infants. J. Infect. Dis. 2014, 210, 1519–1522. [Google Scholar] [CrossRef] [PubMed]
  139. Gantner, P.; Barnig, C.; Partisani, M.; Lee, G.Q.; Beck-Wirth, G.; Faller, J.P.; Martinot, M.; Mosheni-Zadeh, M.; Cheneau, C.; Batard, M.L.; et al. Distribution and reduction magnitude of HIV-DNA burden in CD4+ T cell subsets depend on art initiation timing. AIDS 2018, 32, 921–926. [Google Scholar] [CrossRef]
  140. Murray, J.M.; Zaunders, J.; Emery, S.; Cooper, D.A.; Hey-Nguyen, W.J.; Koelsch, K.K.; Kelleher, A.D. HIV dynamics linked to memory CD4+ T cell homeostasis. PLoS ONE 2017, 12, e0186101. [Google Scholar] [CrossRef]
  141. McHantaf, G.; Cheret, A.; Melard, A.; Essat, A.; Gardiennet, E.; Bauer, R.; Charre, C.; Meiffredy, V.; Piroth, L.; Goujard, C.; et al. The build-up of stock of stable integrated proviruses overtime explains the difficulty in reducing HIV-1 DNA levels when treatment is initiated at the chronic stage of the infection. J. Virus Erad. 2023, 9, 100357. [Google Scholar] [CrossRef]
  142. Lisziewicz, J.; Rosenberg, E.; Lieberman, J.; Jessen, H.; Lopalco, L.; Siliciano, R.; Walker, B.; Lori, F. Control of HIV despite the discontinuation of antiretroviral therapy. N. Engl. J. Med. 1999, 340, 1683–1684. [Google Scholar] [CrossRef]
  143. Hocqueloux, L.; Prazuck, T.; Avettand-Fenoel, V.; Lafeuillade, A.; Cardon, B.; Viard, J.P.; Rouzioux, C. Long-term immunovirologic control following antiretroviral therapy interruption in patients treated at the time of primary HIV-1 infection. Aids 2010, 24, 1598–1601. [Google Scholar] [CrossRef]
  144. Goujard, C.; Girault, I.; Rouzioux, C.; Lecuroux, C.; Deveau, C.; Chaix, M.L.; Jacomet, C.; Talamali, A.; Delfraissy, J.F.; Venet, A.; et al. HIV-1 control after transient antiretroviral treatment initiated in primary infection: Role of patient characteristics and effect of therapy. Antivir. Ther. 2012, 17, 1001–1009. [Google Scholar] [CrossRef]
  145. Saez-Cirion, A.; Bacchus, C.; Hocqueloux, L.; Avettand-Fenoel, V.; Girault, I.; Lecuroux, C.; Potard, V.; Versmisse, P.; Melard, A.; Prazuck, T.; et al. Post-treatment HIV-1 controllers with a long-term virological remission after the interruption of early initiated antiretroviral therapy ANRS VISCONTI Study. PLoS Pathog. 2013, 9, e1003211. [Google Scholar] [CrossRef]
  146. Steingrover, R.; Pogany, K.; Fernandez Garcia, E.; Jurriaans, S.; Brinkman, K.; Schuitemaker, H.; Miedema, F.; Lange, J.M.; Prins, J.M. HIV-1 viral rebound dynamics after a single treatment interruption depends on time of initiation of highly active antiretroviral therapy. AIDS 2008, 22, 1583–1588. [Google Scholar] [CrossRef] [PubMed]
  147. Steingrover, R.; Garcia, E.F.; van Valkengoed, I.G.; Bekker, V.; Bezemer, D.; Kroon, F.P.; Dekker, L.; Prins, M.; de Wolf, F.; Lange, J.M.; et al. Transient lowering of the viral set point after temporary antiretroviral therapy of primary HIV type 1 infection. AIDS Res. Hum. Retroviruses 2010, 26, 379–387. [Google Scholar] [CrossRef] [PubMed]
  148. Williams, J.P.; Hurst, J.; Stohr, W.; Robinson, N.; Brown, H.; Fisher, M.; Kinloch, S.; Cooper, D.; Schechter, M.; Tambussi, G.; et al. HIV-1 DNA predicts disease progression and post-treatment virological control. Elife 2014, e03821. [Google Scholar] [CrossRef] [PubMed]
  149. 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]
  150. Lambotte, O.; Boufassa, F.; Madec, Y.; Nguyen, A.; Goujard, C.; Meyer, L.; Rouzioux, C.; Venet, A.; Delfraissy, J.F. HIV controllers: A homogeneous group of HIV-1-infected patients with spontaneous control of viral replication. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2005, 41, 1053–1056. [Google Scholar] [CrossRef]
  151. Dalmasso, C.; Carpentier, W.; Meyer, L.; Rouzioux, C.; Goujard, C.; Chaix, M.L.; Lambotte, O.; Avettand-Fenoel, V.; Le Clerc, S.; de Senneville, L.D.; et al. Distinct genetic loci control plasma HIV-RNA and cellular HIV-DNA levels in HIV-1 infection: The ANRS Genome Wide Association 01 study. PLoS ONE 2008, 3, e3907. [Google Scholar] [CrossRef]
  152. Sodora, D.L.; Ross, T.M. Simian immunodeficiency virus pathogenesis. Curr. HIV Res. 2009, 7, 1. [Google Scholar] [CrossRef]
  153. Nchinda, N.; Elangovan, R.; Yun, J.; Dickson-Tetteh, L.; Kirtley, S.; Hemelaar, J.; WHO-UNAIDS Network for HIV Isolation and Characterisation. Characterisation, Global associations of key populations with HIV-1 recombinants: A systematic review, global survey, and individual participant data meta-analysis. Front. Public Health 2023, 11, 1153638. [Google Scholar] [CrossRef]
  154. Bimela, J.S.; Nanfack, A.J.; Yang, P.; Dai, S.; Kong, X.P.; Torimiro, J.N.; Duerr, R. Antiretroviral Imprints and Genomic Plasticity of HIV-1 pol in Non-clade B: Implications for Treatment. Front. Microbiol. 2021, 12, 812391. [Google Scholar] [CrossRef]
  155. Visseaux, B.; Assoumou, L.; Mahjoub, N.; Grude, M.; Trabaud, M.A.; Raymond, S.; Wirden, M.; Morand-Joubert, L.; Roussel, C.; Montes, B.; et al. Surveillance of HIV-1 primary infections in France from 2014 to 2016: Toward stable resistance, but higher diversity, clustering and virulence? J. Antimicrob. Chemother. 2020, 75, 183–193. [Google Scholar] [CrossRef]
  156. Haggblom, A.; Svedhem, V.; Singh, K.; Sonnerborg, A.; Neogi, U. Virological failure in patients with HIV-1 subtype C receiving antiretroviral therapy: An analysis of a prospective national cohort in Sweden. Lancet HIV 2016, 3, e166–e174. [Google Scholar] [CrossRef] [PubMed]
  157. Tremeaux, P.; Lemoine, F.; Melard, A.; Gousset, M.; Boufassa, F.; Orr, S.; Monceaux, V.; Gascuel, O.; Lambotte, O.; Hocqueloux, L.; et al. In-Depth Characterization of Full-Length Archived Viral Genomes after Nine Years of Posttreatment HIV Control. Microbiol. Spectr. 2023, 11, e0326722. [Google Scholar] [CrossRef] [PubMed]
  158. Evering, T.H.; Mehandru, S.; Racz, P.; Tenner-Racz, K.; Poles, M.A.; Figueroa, A.; Mohri, H.; Markowitz, M. Absence of HIV-1 evolution in the gut-associated lymphoid tissue from patients on combination antiviral therapy initiated during primary infection. PLoS Pathog. 2012, 8, e1002506. [Google Scholar] [CrossRef] [PubMed]
  159. Imamichi, H.; Dewar, R.L.; Adelsberger, J.W.; Rehm, C.A.; O’Doherty, U.; Paxinos, E.E.; Fauci, A.S.; Lane, H.C. Defective HIV-1 proviruses produce novel protein-coding RNA species in HIV-infected patients on combination antiretroviral therapy. Proc. Natl. Acad. Sci. USA 2016, 113, 8783–8788. [Google Scholar] [CrossRef]
  160. Baxter, A.E.; O’Doherty, U.; Kaufmann, D.E. Beyond the replication-competent HIV reservoir: Transcription and translation-competent reservoirs. Retrovirology 2018, 15, 18. [Google Scholar] [CrossRef]
  161. Hurst, J.; Hoffmann, M.; Pace, M.; Williams, J.P.; Thornhill, J.; Hamlyn, E.; Meyerowitz, J.; Willberg, C.; Koelsch, K.K.; Robinson, N.; et al. Immunological biomarkers predict HIV-1 viral rebound after treatment interruption. Nat. Commun. 2015, 6, 8495. [Google Scholar] [CrossRef]
  162. Haissman, J.M.; Vestergaard, L.S.; Sembuche, S.; Erikstrup, C.; Mmbando, B.; Mtullu, S.; Lemnge, M.M.; Gerstoft, J.; Ullum, H. Plasma cytokine levels in Tanzanian HIV-1-infected adults and the effect of antiretroviral treatment. J. Acquir. Immune Defic. Syndr. 2009, 52, 493–497. [Google Scholar] [CrossRef]
  163. Muema, D.M.; Akilimali, N.A.; Ndumnego, O.C.; Rasehlo, S.S.; Durgiah, R.; Ojwach, D.B.A.; Ismail, N.; Dong, M.; Moodley, A.; Dong, K.L.; et al. Association between the cytokine storm, immune cell dynamics, and viral replicative capacity in hyperacute HIV infection. BMC Med. 2020, 18, 81. [Google Scholar] [CrossRef]
  164. Allers, K.; Puyskens, A.; Epple, H.J.; Schurmann, D.; Hofmann, J.; Moos, V.; Schneider, T. The effect of timing of antiretroviral therapy on CD4 T-cell reconstitution in the intestine of HIV-infected patients. Mucosal. Immunol. 2015, 9, 265–274. [Google Scholar] [CrossRef]
  165. Krebs, S.J.; Ananworanich, J. Immune activation during acute HIV infection and the impact of early antiretroviral therapy. Curr. Opin. HIV AIDS 2016, 11, 163–172. [Google Scholar] [CrossRef]
  166. Ipp, H.; Zemlin, A.E.; Erasmus, R.T.; Glashoff, R.H. Role of inflammation in HIV-1 disease progression and prognosis. Crit. Rev. Clin. Lab. Sci. 2014, 51, 98–111. [Google Scholar] [CrossRef] [PubMed]
  167. Herout, S.; Mandorfer, M.; Breitenecker, F.; Reiberger, T.; Grabmeier-Pfistershammer, K.; Rieger, A.; Aichelburg, M.C. Impact of Early Initiation of Antiretroviral Therapy in Patients with Acute HIV Infection in Vienna, Austria. PLoS ONE 2016, 11, e0152910. [Google Scholar] [CrossRef] [PubMed]
  168. Hong, J.J.; Silveira, E.; Amancha, P.K.; Byrareddy, S.N.; Gumber, S.; Chang, K.T.; Ansari, A.A.; Villinger, F. Early initiation of antiretroviral treatment postSIV infection does not resolve lymphoid tissue activation. AIDS 2017, 31, 1819–1824. [Google Scholar] [CrossRef] [PubMed]
  169. Lederman, M.M.; Funderburg, N.T.; Sekaly, R.P.; Klatt, N.R.; Hunt, P.W. Residual Immune Dysregulation Syndrome in Treated HIV infection. Adv. Immunol. 2013, 119, 51–83. [Google Scholar]
  170. Novelli, S.; Lecuroux, C.; Goujard, C.; Reynes, J.; Villemant, A.; Blum, L.; Essat, A.; Avettand-Fenoel, V.; Launay, O.; Molina, J.M.; et al. Persistence of monocyte activation under treatment in people followed since acute HIV-1 infection relative to participants at high or low risk of HIV infection. EBioMedicine 2020, 62, 103129. [Google Scholar] [CrossRef]
  171. Wang, S.X.; Ho, E.L.; Grill, M.; Lee, E.; Peterson, J.; Robertson, K.; Fuchs, D.; Sinclair, E.; Price, R.W.; Spudich, S. Peripheral neuropathy in primary HIV infection associates with systemic and central nervous system immune activation. J. Acquir. Immune Defic. Syndr. 2014, 66, 303–310. [Google Scholar] [CrossRef]
  172. Grijsen, M.; Koster, G.; van Vonderen, M.; van Kasteren, M.; Kootstra, G.; Steingrover, R.; de Wolf, F.; Prins, J.; Nieuwkerk, P. Temporary antiretroviral treatment during primary HIV-1 infection has a positive impact on health-related quality of life: Data from the Primo-SHM cohort study. HIV Med. 2012, 13, 630–635. [Google Scholar] [CrossRef]
  173. Autran, B.; Carcelain, G.; Li, T.S.; Blanc, C.; Mathez, D.; Tubiana, R.; Katlama, C.; Debre, P.; Leibowitch, J. Positive effects of combined antiretroviral therapy on CD4+ T cell homeostasis and function in advanced HIV disease. Science 1997, 277, 112–116. [Google Scholar] [CrossRef]
  174. Rosenberg, E.S.; Billingsley, J.M.; Caliendo, A.M.; Boswell, S.L.; Sax, P.E.; Kalams, S.A.; Walker, B.D. Vigorous HIV-1-specific CD4+ T cell responses associated with control of viremia. Science 1997, 278, 1447–1450. [Google Scholar] [CrossRef]
  175. Hecht, F.M.; Wang, L.; Collier, A.; Little, S.; Markowitz, M.; Margolick, J.; Kilby, J.M.; Daar, E.; Conway, B.; Holte, S. A multicenter observational study of the potential benefits of initiating combination antiretroviral therapy during acute HIV infection. J. Infect. Dis. 2006, 194, 725–733. [Google Scholar] [CrossRef]
  176. Oxenius, A.; Price, D.A.; Easterbrook, P.J.; O’Callaghan, C.A.; Kelleher, A.D.; Whelan, J.A.; Sontag, G.; Sewell, A.K.; Phillips, R.E. Early highly active antiretroviral therapy for acute HIV-1 infection preserves immune function of CD8+ and CD4+ T lymphocytes. Proc. Natl. Acad. Sci. USA 2000, 97, 3382–3387. [Google Scholar] [CrossRef] [PubMed]
  177. Deleage, C.; Schuetz, A.; Alvord, W.G.; Johnston, L.; Hao, X.P.; Morcock, D.R.; Rerknimitr, R.; Fletcher, J.L.; Puttamaswin, S.; Phanuphak, N.; et al. Impact of early cART in the gut during acute HIV infection. JCI Insight 2016, 1, e87065. [Google Scholar] [CrossRef] [PubMed]
  178. Cellerai, C.; Harari, A.; Stauss, H.; Yerly, S.; Geretti, A.M.; Carroll, A.; Yee, T.; Ainsworth, J.; Williams, I.; Sweeney, J.; et al. Early and prolonged antiretroviral therapy is associated with an HIV-1-specific T-cell profile comparable to that of long-term non-progressors. PLoS ONE 2011, 6, e18164. [Google Scholar] [CrossRef]
  179. Lecuroux, C.; Girault, I.; Boutboul, F.; Urrutia, A.; Goujard, C.; Meyer, L.; Lambotte, O.; Chaix, M.L.; Martinez, V.; Autran, B.; et al. Antiretroviral therapy initiation during primary HIV infection enhances both CD127 expression and the proliferative capacity of HIV-specific CD8+ T cells. AIDS 2009, 23, 1649–1658. [Google Scholar] [CrossRef] [PubMed]
  180. Ndhlovu, Z.M.; Kazer, S.W.; Nkosi, T.; Ogunshola, F.; Muema, D.M.; Anmole, G.; Swann, S.A.; Moodley, A.; Dong, K.; Reddy, T.; et al. Augmentation of HIV-specific T cell function by immediate treatment of hyperacute HIV-1 infection. Sci. Transl. Med. 2019, 11, eaau0528. [Google Scholar] [CrossRef]
  181. Hansen, S.G.; Piatak, M., Jr.; Ventura, A.B.; Hughes, C.M.; Gilbride, R.M.; Ford, J.C.; Oswald, K.; Shoemaker, R.; Li, Y.; Lewis, M.S.; et al. Immune clearance of highly pathogenic SIV infection. Nature 2013, 502, 100–104. [Google Scholar] [CrossRef]
  182. Passaes, C.; Desjardins, D.; Chapel, A.; Monceaux, V.; Lemaitre, J.; Melard, A.; Perdomo-Celis, F.; Planchais, C.; Gourves, M.; Dimant, N.; et al. Early antiretroviral therapy favors post-treatment SIV control associated with the expansion of enhanced memory CD8(+) T-cells. Nat. Commun. 2024, 15, 178. [Google Scholar] [CrossRef]
  183. Thornhill, J.; Inshaw, J.; Oomeer, S.; Kaleebu, P.; Cooper, D.; Ramjee, G.; Schechter, M.; Tambussi, G.; Fox, J.; Miro, J.M.; et al. Enhanced normalisation of CD4/CD8 ratio with early antiretroviral therapy in primary HIV infection. J. Int. AIDS Soc. 2014, 17 (Suppl. 3), 19480. [Google Scholar] [CrossRef]
  184. Cao, W.; Mehraj, V.; Trottier, B.; Baril, J.G.; Leblanc, R.; Lebouche, B.; Cox, J.; Tremblay, C.; Lu, W.; Singer, J.; et al. Early Initiation Rather Than Prolonged Duration of Antiretroviral Therapy in HIV Infection Contributes to the Normalization of CD8 T-Cell Counts. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2016, 62, 250–257. [Google Scholar] [CrossRef]
  185. Sereti, I.; Krebs, S.J.; Phanuphak, N.; Fletcher, J.L.; Slike, B.; Pinyakorn, S.; O’Connell, R.J.; Rupert, A.; Chomont, N.; Valcour, V.; et al. Persistent, Albeit Reduced, Chronic Inflammation in Persons Starting Antiretroviral Therapy in Acute HIV Infection. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2017, 64, 124–131. [Google Scholar] [CrossRef]
  186. Vasan, S.; Poles, M.A.; Horowitz, A.; Siladji, E.E.; Markowitz, M.; Tsuji, M. Function of NKT cells, potential anti-HIV effector cells, are improved by beginning HAART during acute HIV-1 infection. Int. Immunol. 2007, 19, 943–951. [Google Scholar] [CrossRef] [PubMed]
  187. Killian, M.S.; Fujimura, S.H.; Hecht, F.M.; Levy, J.A. Similar changes in plasmacytoid dendritic cell and CD4 T-cell counts during primary HIV-1 infection and treatment. AIDS 2006, 20, 1247–1252. [Google Scholar] [CrossRef] [PubMed]
  188. Planchais, C.; Hocqueloux, L.; Ibanez, C.; Gallien, S.; Copie, C.; Surenaud, M.; Kok, A.; Lorin, V.; Fusaro, M.; Delfau-Larue, M.H.; et al. Early Antiretroviral Therapy Preserves Functional Follicular Helper T and HIV-Specific B Cells in the Gut Mucosa of HIV-1-Infected Individuals. J. Immunol. 2018, 200, 3519–3529. [Google Scholar] [CrossRef] [PubMed]
  189. de Souza, M.S.; Pinyakorn, S.; Akapirat, S.; Pattanachaiwit, S.; Fletcher, J.L.; Chomchey, N.; Kroon, E.D.; Ubolyam, S.; Michael, N.L.; Robb, M.L.; et al. Initiation of Antiretroviral Therapy During Acute HIV-1 Infection Leads to a High Rate of Nonreactive HIV Serology. Clin. Infect. Dis. 2016, 63, 555–561. [Google Scholar] [CrossRef]
  190. 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]
  191. Pantazis, N.; Touloumi, G.; Vanhems, P.; Gill, J.; Bucher, H.C.; Porter, K. The effect of antiretroviral treatment of different durations in primary HIV infection. AIDS 2008, 22, 2441–2450. [Google Scholar] [CrossRef]
  192. Novelli, S.; Delobel, P.; Bouchaud, O.; Avettand-Fenoel, V.; Fialaire, P.; Cabie, A.; Souala, F.; Raffi, F.; Catalan, P.; Weiss, L.; et al. Enhanced immunovirological response in women compared to men after antiretroviral therapy initiation during acute and early HIV-1 infection: Results from a longitudinal study in the French ANRS Primo cohort. J. Int. AIDS Soc. 2020, 23, e25485. [Google Scholar] [CrossRef]
  193. Imamichi, H.; Smith, M.; Adelsberger, J.W.; Izumi, T.; Scrimieri, F.; Sherman, B.T.; Rehm, C.A.; Imamichi, T.; Pau, A.; Catalfamo, M.; et al. Defective HIV-1 proviruses produce viral proteins. Proc. Natl. Acad. Sci. USA 2020, 117, 3704–3710. [Google Scholar] [CrossRef]
  194. Ambrosioni, J.; Junier, T.; Delhumeau, C.; Calmy, A.; Hirschel, B.; Zdobnov, E.; Kaiser, L.; Yerly, S. Impact of highly active antiretroviral therapy on the molecular epidemiology of newly diagnosed HIV infections. AIDS 2012, 26, 2079–2086. [Google Scholar] [CrossRef]
  195. Brenner, B.G.; Roger, M.; Routy, J.P.; Moisi, D.; Ntemgwa, M.; Matte, C.; Baril, J.G.; Thomas, R.; Rouleau, D.; Bruneau, J.; et al. High rates of forward transmission events after acute/early HIV-1 infection. J. Infect. Dis. 2007, 195, 951–959. [Google Scholar] [CrossRef]
  196. O’Brien, M.; Markowitz, M. Should we treat acute HIV infection? Curr. HIV/AIDS Rep. 2012, 9, 101–110. [Google Scholar] [CrossRef]
  197. Baeten, J.M.; Kahle, E.; Lingappa, J.R.; Coombs, R.W.; Delany-Moretlwe, S.; Nakku-Joloba, E.; Mugo, N.R.; Wald, A.; Corey, L.; Donnell, D.; et al. Genital HIV-1 RNA predicts risk of heterosexual HIV-1 transmission. Sci. Transl. Med. 2011, 3, 77ra29. [Google Scholar] [CrossRef] [PubMed]
  198. Pilcher, C.D.; Tien, H.C.; Eron, J.J., Jr.; Vernazza, P.L.; Leu, S.Y.; Stewart, P.W.; Goh, L.E.; Cohen, M.S. Brief but efficient: Acute HIV infection and the sexual transmission of HIV. J. Infect. Dis. 2004, 189, 1785–1792. [Google Scholar] [CrossRef] [PubMed]
  199. Pullium, J.K.; Adams, D.R.; Jackson, E.; Kim, C.N.; Smith, D.K.; Janssen, R.; Gould, K.; Folks, T.M.; Butera, S.; Otten, R.A. Pig-tailed macaques infected with human immunodeficiency virus (HIV) type 2GB122 or simian/HIV89.6p express virus in semen during primary infection: New model for genital tract shedding and transmission. J. Infect. Dis. 2001, 183, 1023–1030. [Google Scholar] [CrossRef] [PubMed]
  200. Curtis, T.J.; Rodger, A.J.; Burns, F.; Nardone, A.; Copas, A.; Wayal, S. Patterns of sexualised recreational drug use and its association with risk behaviours and sexual health outcomes in men who have sex with men in London, UK: A comparison of cross-sectional studies conducted in 2013 and 2016. Sex Transm. Infect. 2020, 96, 197–203. [Google Scholar] [CrossRef]
  201. Hegazi, A.; Lee, M.J.; Whittaker, W.; Green, S.; Simms, R.; Cutts, R.; Nagington, M.; Nathan, B.; Pakianathan, M.R. Chemsex and the city: Sexualised substance use in gay bisexual and other men who have sex with men attending sexual health clinics. Int. J. STD AIDS 2017, 28, 362–366. [Google Scholar] [CrossRef]
  202. Maxwell, S.; Shahmanesh, M.; Gafos, M. Chemsex behaviours among men who have sex with men: A systematic review of the literature. Int. J. Drug Policy 2019, 63, 74–89. [Google Scholar] [CrossRef]
  203. Gonzalez-Baeza, A.; Dolengevich-Segal, H.; Perez-Valero, I.; Cabello, A.; Tellez, M.J.; Sanz, J.; Perez-Latorre, L.; Bernardino, J.I.; Troya, J.; De La Fuente, S.; et al. Sexualized Drug Use (Chemsex) Is Associated with High-Risk Sexual Behaviors and Sexually Transmitted Infections in HIV-Positive Men Who Have Sex with Men: Data from the U-SEX GESIDA 9416 Study. AIDS Patient Care STDS 2018, 32, 112–118. [Google Scholar] [CrossRef]
  204. Pufall, E.L.; Kall, M.; Shahmanesh, M.; Nardone, A.; Gilson, R.; Delpech, V.; Ward, H.; Positive Voices Study Group; Hart, G.; Anderson, J.; et al. Sexualized drug use (‘chemsex’) and high-risk sexual behaviours in HIV-positive men who have sex with men. HIV Med. 2018, 19, 261–270. [Google Scholar] [CrossRef]
  205. 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]
  206. Liu, A.Y.; Cohen, S.E.; Vittinghoff, E.; Anderson, P.L.; Doblecki-Lewis, S.; Bacon, O.; Chege, W.; Postle, B.S.; Matheson, T.; Amico, K.R.; et al. Preexposure Prophylaxis for HIV Infection Integrated With Municipal- and Community-Based Sexual Health Services. JAMA Intern. Med. 2016, 176, 75–84. [Google Scholar] [CrossRef] [PubMed]
  207. Baltes, V.; de Boissieu, P.; Champenois, K.; Luan, L.; Seng, R.; Essat, A.; Novelli, S.; Spire, B.; Molina, J.M.; Goujard, C.; et al. Sexual behaviour and STIs among MSM living with HIV in the PrEP era: The French ANRS PRIMO cohort study. J. Int. AIDS Soc. 2024, 27, e26226. [Google Scholar] [CrossRef] [PubMed]
  208. Dai, M.; Calabrese, C. Socio-behavioral factors related to PrEP non-adherence among gay male PrEP users living in California and New York: A behavioral theory informed approach. J. Behav. Med. 2022, 45, 240–251. [Google Scholar] [CrossRef] [PubMed]
  209. Kenyon, C.; Van Dijck, C.; Florence, E. Facing increased sexually transmitted infection incidence in HIV preexposure prophylaxis cohorts: What are the underlying determinants and what can be done? Curr. Opin. Infect. Dis. 2020, 33, 51–58. [Google Scholar] [CrossRef]
  210. Methy, N.; Meyer, L.; Bajos, N.; Velter, A. Generational analysis of trends in unprotected sex in France among men who have sex with men: The major role of context-driven evolving patterns. PLoS ONE 2017, 12, e0171493. [Google Scholar]
  211. Moschese, D.; Lazzarin, S.; Colombo, M.L.; Caruso, F.; Giacomelli, A.; Antinori, S.; Gori, A. Breakthrough Acute HIV Infections among Pre-Exposure Prophylaxis Users with High Adherence: A Narrative Review. Viruses 2024, 16, 951. [Google Scholar] [CrossRef]
  212. Wyl, V.; Gianella, S.; Fischer, M.; Niederoest, B.; Kuster, H.; Battegay, M.; Bernasconi, E.; Cavassini, M.; Rauch, A.; Hirschel, B.; et al. Early antiretroviral therapy during primary HIV-1 infection results in a transient reduction of the viral setpoint upon treatment interruption. PLoS ONE 2011, 6, e27463. [Google Scholar]
  213. Sugiyama, F.H.C.; Dietz, L.L.; Sogaard, O.S. Utilizing immunotherapy towards achieving a functional cure for HIV-1. Curr. Opin. HIV AIDS 2024, 19, 187–193. [Google Scholar] [CrossRef]
  214. Program, U.M.H.R. Safety and Efficacy of Neutralizing Antibodies and Vaccination for Induction of HIV Remission (RV582). Available online: https://clinicaltrials.gov/study/NCT05769569?cond=acute%20hiv&intr=bNAbs&rank=3 (accessed on 2 March 2025).
  215. NIAID. Evaluation of Safety, Immunogenicity and Efficacy of a Triple Immune Regimen in Adults Initiated on ART During Acute HIV-1. Available online: https://clinicaltrials.gov/study/NCT06071767?cond=acute%20hiv&intr=bNAbs&rank=2 (accessed on 2 March 2025).
  216. ANRS. Phase II Trial of ART + Dual bNAbs vs. ART + Placebo During Primary HIV-1 Infection-impact on Post-ART Control (RHIEVIERA-02). Available online: https://clinicaltrials.gov/study/NCT05300035?cond=hiv&intr=bNAbs&rank=3 (accessed on 2 March 2025).
Figure 1. Patho-physiological model of acute and recent HIV infection. Acute HIV infection marks the onset of a virological and immune storm at its peak. The initial viral replication in infected productive cells leads to a high production of virions and viral proteins, which triggers first intense innate immunity response (including myeloid cells) and contribute to the immune storm by inducing activation and inflammation, with the production of inflammatory cytokines and chemokines, and mobilizing cellular and immune responses. Acute infection elicits intense immune responses, characterized by massive T cell activation and proliferation, which facilitate cell infection and the establishment of a comprehensive HIV reservoir. This reservoir includes both latently infected and productive cells within lymph nodes and tissues. A more or less attenuated level of inflammation and various immune stimulations then persist, maintaining a continuous model with constant viral replication in the absence of treatment.
Figure 1. Patho-physiological model of acute and recent HIV infection. Acute HIV infection marks the onset of a virological and immune storm at its peak. The initial viral replication in infected productive cells leads to a high production of virions and viral proteins, which triggers first intense innate immunity response (including myeloid cells) and contribute to the immune storm by inducing activation and inflammation, with the production of inflammatory cytokines and chemokines, and mobilizing cellular and immune responses. Acute infection elicits intense immune responses, characterized by massive T cell activation and proliferation, which facilitate cell infection and the establishment of a comprehensive HIV reservoir. This reservoir includes both latently infected and productive cells within lymph nodes and tissues. A more or less attenuated level of inflammation and various immune stimulations then persist, maintaining a continuous model with constant viral replication in the absence of treatment.
Viruses 17 00366 g001
Figure 2. Development of Biological Markers at the time of Acute and recent HIV Infection: The progression of HIV-1 infection can be divided into six distinct stages named Fiebig stages based on results of standard laboratory tests. They are characterized by the sequential detection starting with HIV-1 RNA, p24 antigen and specific antibodies against HIV-1 proteins, detected by Elisa (the 4th generation, the more sensitive), including anti-p34, detected by Western blot analysis. In the very first step of acute HIV infection, the pattern of the Western blot may be indeterminate, then weak or with an isolated anti-p24 band, followed by reactivity against gp41 and progressing to a complete pattern. HIV viral reservoir can be estimated using the quantification of total HIV-DNA in PBMC, which is detectable in the first step. For newborns, the diagnosis of acute HIV infection can be established by quantifying plasma HIV-RNA in the first days of life, as well as by measuring total blood HIV-DNA. A plus sign indicates a positive test result, a minus sign denotes a negative result, and a plus-minus sign represents a borderline-positive result. Adapted from Cohen et al, NEJM, 2011 [19].
Figure 2. Development of Biological Markers at the time of Acute and recent HIV Infection: The progression of HIV-1 infection can be divided into six distinct stages named Fiebig stages based on results of standard laboratory tests. They are characterized by the sequential detection starting with HIV-1 RNA, p24 antigen and specific antibodies against HIV-1 proteins, detected by Elisa (the 4th generation, the more sensitive), including anti-p34, detected by Western blot analysis. In the very first step of acute HIV infection, the pattern of the Western blot may be indeterminate, then weak or with an isolated anti-p24 band, followed by reactivity against gp41 and progressing to a complete pattern. HIV viral reservoir can be estimated using the quantification of total HIV-DNA in PBMC, which is detectable in the first step. For newborns, the diagnosis of acute HIV infection can be established by quantifying plasma HIV-RNA in the first days of life, as well as by measuring total blood HIV-DNA. A plus sign indicates a positive test result, a minus sign denotes a negative result, and a plus-minus sign represents a borderline-positive result. Adapted from Cohen et al, NEJM, 2011 [19].
Viruses 17 00366 g002
Table 1. Main benefits of initiating treatment during Acute HIV Infection compared to late treatment.
Table 1. Main benefits of initiating treatment during Acute HIV Infection compared to late treatment.
Patient treated at the time of acute infectionPatient treated at the chronic phase
Transmission is typically homogeneous, involving a single virion with dominant CCR5 viral tropismBoth R5 and X4 variants emerging during infection and associated with rapid disease progression
Preventing escape mutations and limiting viral diversity
Less defective proviral gene insertions, limiting immune activation
Large viral diversity
Lower virologic set point
Best reduction of residual viremia
The treatment quickly restores and protects the immune system: Protection of the different polyfunctionalities of TCD4+ and TCD8+, limit apoptosis by reducing activation and viral diversityMore limited immune restoration: From the surviving TCD4+ and TCD8+ clones to achieve the different polyfunctionalities
Major difference in the quantity and quality of the reservoir
Short Half-Life Reservoir:
TTM infection and TCM protection
Better reduction in total HIV-1 DNA and integrated HIV-DNA
Prevents the establishment of the stable form of integrated proviral HIV-1 DNA that is less prone to elimination
Long Half-Life Reservoir:
TCM Infection
Blood reservoirs are mainly composed of stable proviruses in long-lived quiescent cells and are unaffected by ART
Control of replication in post-treatment, VISCONTI:
A preserved immune system that allows effective control of replication and suppression of the viral reservoir after treatment discontinuation.
No control of replication:
An immune system unable to control replication after treatment discontinuation.
Optimal conditions for future cure trials
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

Chéret, A. Acute HIV-1 Infection: Paradigm and Singularity. Viruses 2025, 17, 366. https://doi.org/10.3390/v17030366

AMA Style

Chéret A. Acute HIV-1 Infection: Paradigm and Singularity. Viruses. 2025; 17(3):366. https://doi.org/10.3390/v17030366

Chicago/Turabian Style

Chéret, Antoine. 2025. "Acute HIV-1 Infection: Paradigm and Singularity" Viruses 17, no. 3: 366. https://doi.org/10.3390/v17030366

APA Style

Chéret, A. (2025). Acute HIV-1 Infection: Paradigm and Singularity. Viruses, 17(3), 366. https://doi.org/10.3390/v17030366

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