- Review
- Open access
- Published:
The role of genetic diversity, epigenetic regulation, and sex-based differences in HIV cure research: a comprehensive review
Epigenetics & Chromatin volume 18, Article number: 1 (2025)
Abstract
Despite significant advances in HIV treatment, a definitive cure remains elusive. The first-in-human clinical trial of Excision BioTherapeutics’ CRISPR-based HIV cure, EBT-101, demonstrated safety but failed to prevent viral rebound. These outcomes may result from the interplay of several factors. Growing evidence indicates that intricate epigenetic modifications play a major role in the persistence of HIV latency, presenting a significant barrier to eradication efforts and causing viral rebound after ART discontinuation. Current strategies to purge the latent reservoir involve LRAs that reactivate latent proviruses. However, their clinical success is hindered by the heterogeneity of HIV reservoirs and the virus’s diverse pathways. Additionally, RNA modifications like N6-methyladenosine (m^6 A) methylation influence HIV biology beyond transcriptional control, affect RNA stability, splicing, and translation, which could enhance therapeutic efficacy. The regulatory framework of chromatin dynamics is also key to understanding viral latency and reactivation, such as Vpr’s role in reactivating latent HIV by targeting HDACs. Sex-specific factors were also shown to play an important role with females, showing stronger early immune responses and higher representation among elite controllers. This review addresses the multifaceted challenges of HIV cure research, focusing on genetic diversity, epigenetic regulation, RNA modifications, chromatin remodeling, and sex-specific factors. By integrating insights into these aspects, this paper aims to advance our understanding of HIV cure strategies and highlight directions for future research.
Background
Excision BioTherapeutics, a California-based biotech company, recently unveiled data from the pioneering first-in-human (FIH) clinical trial of a CRISPR-based HIV cure, EBT-101 (NCT05144386) [1]. This trial, which represents a significant advancement, highlighted the complexity of translating preclinical success to clinical efficacy. EBT-101 was administered intravenously using an adenovirus-associated virus vector serotype 9 (AAV9) to aviremic HIV infected adults on stable antiretroviral therapy (ART), followed by treatment interruption at Week 12 [2, 3]. Two guide RNAs were employed to excise three specific sites targeting the viral long terminal repeats (LTRs) and Gag in the latently integrated HIV genome, generating three possible deletions: 5′LTR to Gag, Gag to 3′LTR, and 5′LTR to 3′LTR. While pre-clinical studies in mice and macaques demonstrated promising curative potential, recent data presented at the 27th ASGCT meeting in Baltimore indicated that EBT-101 did not prevent viral rebound in three participants who paused ART during the Phase 1/2 trial, necessitating the resumption of ART [4].
One plausible reason as to why EBT-101 was not able to sufficiently reach its expected potential could be due to insufficient delivery of AAV9. However, the findings of the FIH prompt a thorough discussion of the complexities of HIV. Learning from previous efforts, various research underlines the relevance of epigenetics in HIV cure research. One key challenge is the establishment of HIV latency, which is significantly controlled by epigenetic modifications such as DNA methylation and histone alterations. Ye et al. (2022) demonstrated that the recruitment of transcriptional repressor complexes can promote HIV silencing in specific cell types, such as astrocytes and microglial cells [5]. These complexes create a tightly packed, inactive chromatin structure at the HIV promoter, making the viral DNA inaccessible to gene-editing tools, unless delivered through suitable vectors. Another study showed that CRISPR was effective against transcriptionally active viral particles in latently infected J-Lat 10.6 cells, but the transcriptionally inert HIV-1 DNA remained inaccessible, likely due to modified chromatin structures [6]. This presents a significant challenge to efforts aimed at reactivating and eradicating HIV from latent reservoirs. Other than epigenetics, this paper also dives into additional factors that may be crucial to future human clinical trials by examining the impact of genetic diversity on CRISPR therapies, how post-transcriptional modifications affect viral persistence, and how hormonal and immune responses across genders influence treatment outcomes.
Evolution of antiretroviral therapy (ART) in HIV therapy
The advent of ART in late 1980s marked a significant leap in managing HIV/AIDS, which continues to pose a significant global health challenge, with ~ 39 million people living with HIV (PLHIV) and 40.4 million deaths to date [7]. In 1987, the United States Food and Drug Administration (FDA) sanctioned the usage of Azidothymidine (AZT) as an antiretroviral medication for HIV/AIDS. AZT was effective in inhibiting viral replication and disease advancement but caused significant adverse effects, including the development of resistance to this single medication [8]. In 1996, drug combinations known as highly active antiretroviral therapy (HAART) were first outlined.
Although these combinations significantly decreased mortality and transmission risk, high adherence to daily ART remained essential [9]. Long-acting formulations of cabotegravir (CAB) and rilpivirine (RPV), administered through monthly intramuscular injections, have demonstrated HIV suppression efficacy comparable to standard once-daily oral therapy [10, 11]. Additionally, Gilead’s twice-yearly lenacapavir (LEN), a promising long-acting injectable HIV medication is currently being evaluated for its efficacy in treating HIV-infected individuals and its potential as an injectable pre-exposure prophylaxis (PrEP) for HIV-negative individuals [12].
While ART reduces the viral load to undetectable levels, it does not eradicate HIV, merely forces the virus into a dormant state. Interruption of ART can result in HIV rebound in as short as a few weeks in some patients due to the reactivation of replication competent virus [1]. While the sharp increase in HIV viral load is predominantly driven by clonal expansions from the latent reservoirs, the different rates of rebound events across different subtype infections also demonstrate the importance of having a thorough understanding of HIV genetic diversity [13, 14].
HIV genetic diversity
HIV genetic diversity can be traced to multiple cross-species transmissions of simian immunodeficiency viruses (SIVs) from nonhuman primates to humans. This zoonotic transmission led to the emergence of distinct viral groups: M (Major), O (Outlier), N (non-M, non-O), and the recent group P. The propagation of this virus contributed to the geographical distribution of genetically distinct HIV, with multiple subtypes and recombinant forms circulating globally. As of now, there are 157 HIV circulating recombinant form (CRFs) documented in the Los Alamos National Laboratory HIV sequence database with the three most prevalent HIV subtypes in PLHIV worldwide being B (54.7%), subtype C (14.7%), and CRF01_AE (7.5%) [15].
Although ART appears effective in controlling HIV infections, increasing evidence indicates that subtype-specific drug resistance mutations (DRMs) can influence treatment efficacy [16, 17]. During HIV replication, genetic mutations vary in pattern and frequency among subtypes due to genetic differences, which in turn affect the distribution of DRMs, ART effectiveness and consequently, treatment and vaccine development [18]. There is also disparity in developing effective treatments as most research focuses on Subtype B, which may not apply to regions with other prevalent subtypes. Molecular studies have shown that proteins from different HIV subtypes, notably with glycoprotein gp120 from Subtypes A and B, affect human cells differently, emphasizing the importance of investigating molecular variations between HIV subtypes for treatment development [19].
A crucial example of this was in a study with Malawian HIV subtype C-positive virologic failure patients who were either initiating or already on first-line ART. Viruses with mutations such as K101E, Y181C, and G190A displayed significantly increased resistance to nevirapine (NVP) by up to 893-fold. H221Y mutation, in combination with Y181C/H221Y and other mutations like K103N or K101Q, further increased resistance levels to NVP, up to 100-fold (K103N) or 3000-fold (K101Q). These DRMs not only heightened resistance to the prescribed antiretroviral drugs but also expanded resistance to first-line drugs such as tenofovir [20], once again stressing the importance of genetic diversity of HIV subtypes when optimizing HIV treatment.
Epigenetics and HIV latency
HIV reverse transcribes its RNA and integrates viral DNA into the host genome. The integrated HIV-derived double-stranded DNA, known as provirus, serves as a template for all viral-derived replication components [21, 22]. In most cases, acute infection causes replication-associated cytopathic consequences that result in cell death. However, a small subset of infected long-lived resting memory T cells harbor integrated HIV DNA which persists indefinitely [23, 24]. Since latency is transcriptionally silent, but replication-competent, ART is incapable of targeting latently infected cells.
HIV latency is sustained by epigenetic changes in its promoter region, such as histone deacetylation in proximity to the promoter and DNA methylation in CpG islands, which suppresses viral transcription. HIV integration, similar to DNA transposition, can cause genetic and epigenetic alterations, notably genomic integration-induced host genomic instability, host epigenetic status changes, and altered gene transcription and splicing regulation [25]. Studies have shown that HIV integration sites can lead to aberrant host gene transcription downstream of the integration site. HIV frequently integrates into certain genes, like the BTB domain and CNC homology 2, BACH2 gene, crucial genomic loci that have been identified as key sites of clonal expansion that played a huge role in the silencing of HIV promoters [26]. As HIV integrates into the host cell genome, it relies on the cell’s own epigenetic and transcriptional machinery, including RNAP-II and p-TEFb/CDK9, to transcribe the integrated provirus [27]. In addition to contributing to the development and maintenance of latency, these modifications have the potential to impair regular cellular processes, including prevention of transcription of important immune markers and co-factors that could inhibit viral infection and Tat-dependent transcription [28, 29].
A promising approach to eliminate HIV reservoirs is the “shock and kill” strategy, which utilizes latency-reversing agents (LRAs) to reactivate latent proviruses in the presence of HAART, making them susceptible to immune responses and viral cytopathic effects. Various classes of LRAs, including histone deacetylase inhibitors (HDACis), histone methyltransferase inhibitors (HMTis), DNA methylation inhibitors (DNMTis), and protein kinase C (PKC) agonists, have been extensively studied for their potential to disrupt latency [30]. Among these, HDACis have garnered significant attention by specifically targeting cellular histone deacetylases (HDACs), which maintain HIV latency within CD4 + T-cells. By inhibiting HDACs, HDACis can potentially reverse latency by activating HIV transcription and increasing the expression of viral proteins and virions, enabling the immune system to clear infected cells or cytolysis [31].
Several examples of HDACis include panobinostat, and romidepsin. When administered, these LRAs significantly increase cell-associated unspliced HIV RNA from CD4 + T cells. While panobinostat and romidepsin enhance total and elongated HIV transcripts, they have minimal impact on polyadenylated or multiply spliced transcripts [32, 33]. In clinical trials, these agents stimulate the production of unspliced HIV transcripts, thereby increasing HIV RNA levels in infected cells. However, integrated HIV DNA, including the size of the latent reservoir, remained unchanged, making it essential to generate peptide antigens crucial for T-cell recognition and the elimination of infected cells [26, 27, 34].
PKC agonists have also shown great promise in triggering latent viral expression via NF-κB signalling [35]. PKC agonists, such as prostratin, and bryostatin stimulate transcription factors like NF-κB to bind to the HIV long terminal repeat (LTR) region and initiate transcription [36]. However, Bryostatin-1’s ability to completely eradicate latent reservoir is limited and does not affect latent HIV transcription in vivo [37]. Another challenge is the heterogeneity of latent reservoirs as evident in the range of infected cell types, along with the varying molecular mechanisms controlling latency, which likely differ between cells [38]. Despite several LRAs being developed, their development has been hampered due to the inability to reduce latent reservoirs and significant side effects [39, 40]. Valuable lessons were gained from LRAs which were effective at impacting the epigenetic pathways. However, novel strategies are still needed to induce apoptosis or manipulate cell check points to successfully eradicate cells harboring latent reservoirs [41, 42].
RNA modifications and their impact on HIV replication
The role of epigenetic mechanisms in gene expression and cellular identity is fundamental. While these processes often lead to long-lasting changes, gene regulation extends beyond transcription [43]. Parallel to epigenetic mechanisms, RNA-level regulation, epitranscriptomics, involves over 170 chemical modifications in almost all RNA [44, 45]. The translation of RNA into proteins is further diversified through post-transcriptional modifications (PTMs) including methylation and acetylation, which are crucial for regulating gene expression and affecting protein stability. PTM-induced conformational changes influence proteins’ interacting partners and subsequent signaling cascades [46]. In the context of HIV, these alterations are critical to the viral life cycle and have a substantial impact on the treatment efficacy.
HIV infection has been shown to modify RNA methylation patterns, impacting the efficacy of treatment. One prevalent modification is N6-methyladenosine (\(\:{m}^{6}A\)), found abundantly on eukaryotic mRNA [47]. The addition of a methyl group to the N6 position of adenosine is facilitated by a multi-subunit methyltransferase complex known as the \(\:{m}^{6}A\) writer complex [48]. In brief, \(\:{m}^{6}A\) writer complexes catalyse methyl group addition to adenosine residues within a specific motif on mRNA molecules known as the DRACH motifs. Thereafter, RNA binding proteins, acting as \(\:{m}^{6}A\) readers, recognize methylated residues and influence mRNA metabolism, including secondary structure, nuclear export, stability, splicing, and degradation [49].
From previous studies, 14 \(\:{m}^{6}A\) modification locations were identified in HIV RNA, including coding and noncoding regions, splicing junctions, and splicing regulatory sequences. Notably, methylation of the Rev response-element (RRE) region enhanced Rev protein binding, facilitating viral RNA export from the nucleus, and boosting HIV replication. This shows that viral infection triggers a significant increase in \(\:{m}^{6}A\) in both host and viral mRNAs [47].
Besides that, the presence of \(\:{m}^{6}A\) within the 5’UTR of HIV RNA was discovered in multiple studies [49,50,51]. This 5’UTR region contains essential regulatory components required for effective HIV replication as it enables complete RNA genomes to be incorporated into progeny virions. Two DRACH motifs in the 5’UTR are of particular interest. One is located near the primer-binding site (PBS), which is necessary for initiating reverse transcription (RT). The other is located near the dimer initiation sequence (DIS), which is involved in a monomer-dimer switch that determines whether a given full-length HIV RNA is translated (monomer) or selected for packaging (dimer). When one of these two adenosine residues was mutated, p24 levels in HEK293T producer cells increased, but the virions’ ability to infect target cells decreased [52]. Furthermore, \(\:{m}^{6}A\) modifications exhibit notable conservation within the HIV genome, despite its susceptibility to mutation. Only a small fraction of the nearly 250 DRACH motifs in full-length HIV RNA seem to harbor \(\:{m}^{6}A\), indicating an exceptionally high specificity in these sites [53, 54]. These findings imply the critical role of \(\:{m}^{6}A\) modifications in enhancing efficient virus replication.
Given this context, the presence of HIV-induced \(\:{m}^{6}A\) modifications could significantly decrease the efficacy of therapies targeting HIV replication. Specifically, modifications within RRE enhance the binding of Rev protein, which is crucial for forming nuclear export complexes. Without these modifications, the efficiency of viral RNA export and subsequent replication could be reduced, potentially leading to a decrease in viral load. This could affect the effectiveness of therapies that rely on viral replication as a target. For instance, METTL3/14 knockdown, decreased \(\:{m}^{6}A\) methylation at conserved adenosines A7877 and A7883 within the RRE hairpin structure. Consequently, impaired Rev protein binding to the RRE reduces the export of viral RNA from the nucleus and inhibits viral replication [47]. Thus, the absence of \(\:{m}^{6}A\) modifications demonstrate the effectiveness of therapies that target viral replication, which can have implications for the pharmacodynamics and pharmacokinetics of ART drugs. Although direct evidence linking \(\:{m}^{6}A\) modifications to HIV therapy are limited, studies on other viruses suggest that targeting \(\:{m}^{6}A\) modifications can make drug-resistant cells more responsive to therapy and boost immunotherapeutic outcomes [55]. CRISPR technology, with its ability to precisely target specific sequences, can be utilized to manipulate \(\:{m}^{6}A\) levels, thereby offering novel therapeutic interventions against HIV.
Histone modification and its role on HIV transcription
RNA modifications are crucial for regulating HIV replication and the effectiveness of antiviral therapies. However, chromatin dynamics, which control how DNA is accessed for transcription, also play a significant role. Failure to adapt to this interplay can result in cell death, developmental defects, and disease. Adaptive cellular responses are frequently achieved through inducible changes in gene expression [56, 57]. Chromatin modification is an ideal mechanism for dynamic gene expression, allowing rapid, adaptable, and reversible changes that facilitate DNA-templated processes such as transcription, chromosome segregation, DNA replication, and repair. Examples include histone-modifying enzymes and ATP-dependent chromatin remodelers, which adjust nucleosome position and content. These chromatin modifiers are preserved throughout evolution and control a variety of processes necessary for healthy cell division and organism growth [57, 58].
In the case of HIV, chromatin remodeling is closely linked to viral latency, replication, and the effectiveness of therapy. In the latent reservoir, the provirus remains silent, making it difficult for the immune system and therapeutic interventions to detect these cells. Many studies have shown that chromatin structure and composition are key to establishing and maintaining proviral latency, as well as the reactivation potential of latently infected cells [59,60,61]. The status of chromatin condensation can be influenced by a variety of mechanisms, including post-translational covalent changes of histone tails and the recruitment of repressive proteins on methylated DNA [62]. Through interactions with chromatin-associated proteins and changes in DNA accessibility, these alterations affect patterns of gene expression. The latent state of proviral DNA in infected cells is maintained by a range of enzymes that change histones [63].
One such histone modification that is crucial in regulating gene expression is histone H3 lysine 9 trimethylation (H3K9me3). H3K9me3 is associated with gene repression and is involved in establishing and maintaining heterochromatin [64, 65]. H3K9me3 has been linked to the establishment of viral latency and HIV transcription suppression [59, 64]. A recent study that investigated the early events following the entry of HIV found that core histones, including the variant H3.3, as well as linker histone H1 and the post-translational H3K9me3 modification, are rapidly loaded onto unintegrated HIV DNA. In recent years, the role of unintegrated HIV DNA has become increasingly apparent. Despite no integration, Vpr is able to promote viral transcription from unintegrated DNA, resulting in a high abundance of multiply spliced transcript, an indicator of the frequency of intact virus [66,67,68].
This loading of core histones occurs shortly after the virus engages with nuclear pore proteins upon nuclear entry. The significance of H3.3 loading on viral DNA processing or expression remains unclear, but it is speculated that it may contribute to the low expression of these DNAs. Furthermore, the study detected high levels of the silencing mark H3K9me3 and low levels of the activation mark H3K9Ac on unintegrated HIV DNA. These modifications correlate with the observed silencing phenotype of unintegrated HIV DNAs. Therefore, the potent silencing of unintegrated HIV DNAs by chromatinization presents a major challenge for the use of non-integrating vectors in gene therapy applications [69].
Along with histone methylation, histone acetylation is also a key factor in the dynamic mechanism of chromatin remodeling. This major post-translational modification (PTM) involves the covalent addition of an acetyl moiety to the lysine ε-amino group on histone protein tails. This process is mediated by the counteracting actions of two enzymes: histone acetyltransferases (HATs) and histone deacetylases (HDACs) [33, 70]. By removing acetyl groups from histone tails, HDACs render the chromatin structure into a more compact form of heterochromatin, making the genes inaccessible for transcription and causing transcriptional repression [71]. This mechanism is especially significant because LRAs that include HDAC inhibitors (HDACis) are meant to stimulate the transcriptional activation of latent HIV [72]. Different in vitro models of HIV latency have been used to identify LRAs, yet, no single or combination of LRAs has been shown to significantly reduce the reservoir size in clinical settings [73].
This lack of efficacy may stem from the complexity of the multiple signaling pathways involved in establishing the latent reservoir including the NF-κB, MAPK/ERK, PI3K/AKT/mTOR, Wnt/β-Catenin, TGF-β, JAK/STAT, and Notch pathways, in addition to mechanisms of chromatin modification and remodeling [74,75,76]. Moreover, the reactivation of the entire HIV reservoir by LRAs is often incomplete due to the heterogeneous nature of the reservoirs [77, 78], which result in the limited and inconsistent reactivation spectrum by LRAs. Therefore, the heterogeneity of cells and tissues constituting HIV reservoirs, coupled with the complex molecular pathways, creates substantial challenges in the development of a single LRA capable of fully reactivating latent virus [79]. To effectively overcome the many blockages to HIV gene expression in resting CD4 + T cells, it is commonly anticipated that combinations of LRAs will be required.
Several studies have shown that hyperacetylation of core histones on the HIV long terminal repeat (LTR) is associated with active HIV genome transcription while hypoacetylation is associated with HIV latency. An interesting observation was that the reactivation of latent HIV provirus occurred with Vpr-induced degradation of HDAC1 and HDAC3 in a VprBP-dependent manner [80], implying that the virus has developed a way to combat class I HDACs to reactivate latently infected cells. Additionally, Vpr facilitates the depletion of the cellular repressor CTIP2, which supports the establishment and persistence of HIV latency post-integration in microglial cells. By counteracting this viral gene silencing, Vpr ultimately facilitates the reactivation of HIV expression in a microglia latent model [81]. Additionally, a novel recombinant zinc finger protein transactivator, ZFPb-362-VPR, significantly enhances proviral HIV transcription in latency models, indicating the potential of Vpr-guided transcriptional activation for reactivation [82]. It is worth noting that translating the enhancement of Vpr expression into a therapeutic strategy utilizing CRISPR technology could lead to more effective reactivation of latent reservoirs, potentially resulting in improved eradication strategies. Chromatin remodeling is an important element in the regulation and reactivation of HIV latency; hence a complete understanding of these events is necessary to achieve a viable cure for HIV.
Sex-based disparities and hormonal influence in HIV research and treatment
While understanding chromatin remodeling and epigenetic mechanisms is crucial for developing effective HIV treatments, it is equally important to consider sex-based disparities and hormonal differences in HIV research and treatment outcomes. Sensitivity to hormones can be modified due to epigenetic factors modifying hormone receptor expression. Concurrently, by recruiting epigenetic regulators, hormones can influence epigenetic factors. Taken together, the interplay between epigenetics and hormonal factors can play a key role in regulating viral transcription, and understanding the differences between both sexes would shed light on HIV treatment [83].
Despite accounting for more than half (53%) of the global HIV population, women continue to be substantially underrepresented in HIV clinical trials [84]. This generates gaps in scientific understanding regarding how HIV affects the safety and efficacy of treatment in women and men. Male and female patients may have distinct risks of acquisition, rate of illness progressions and treatment responses due to physiological and hormonal variations [85]. In the past, concerns about possible fetal injury and the need to reduce variability in study results imposed by hormonal cycles led to the exclusion of women of reproductive age from clinical research studies [86]. This exclusion resulted in a huge data gap for analyzing sex-based differences in HIV transmission and progression.
Female HIV infection is distinguished by a more robust early immune response, as demonstrated by a high CD4 + T cell count, low viral load, and high CD8 + T cell activity. On the contrary, a high viral load, a decreased CD4 + T cell count, and a decreased CD8 + T cell cytotoxic function are indicative of HIV infection in males [87, 88]. Aside from that, biological differences between males and females influence HIV susceptibility. Males’ foreskin and females’ vaginal mucosal microbiomes both contain CD4 + T cells that are susceptible to HIV infection following sexual intercourse with an infected partner. However, males are more likely than females to contract HIV during a single sexual encounter with high viral particles because male CD4 + T cells express more CCR5 receptors in their foreskin. Circumcised males have a 60% lower chance of being infected due to the loss of the foreskin. This partially explains the higher viral loads seen in males during primary HIV infection [89, 90].
In addition to that, hormones that control the menstrual cycle have an impact on the cells, bacteria, and mucosal membrane structure of the female genital tract [91]. One such hormone that thickens mucosal membranes and strengthens immune responses to infections is estrogen. As a result, during estrogen-influenced menstrual phases, women are less likely to contract HIV than during progesterone-dominated periods [92]. Estrogen receptors have also been shown to inhibit HIV DNA reactivation in T cells, while receptor antagonists are capable of activating latent HIV proviruses [93]. The differences in hormone and its effects on latency is also extremely important for key populations such as transgender people on gender-affirming hormone therapy (GAHT). Interestingly, GAHT also induces a unique methylation profile, raising an additional epigenetic perspective to be taken into consideration [94]. Learning from the hormonal differences between sexes could be a powerful tool in strategies to tackle the viral reservoir.
Another contributing factor is the presence of sex chromosomes. Females have two X chromosomes, with one of them inactivated at random in each cell during the embryonic stage. However, this inactivation is not complete and errors in this process may occur; hence certain genes on the second X chromosome remain active, potentially offering an immunological benefit. It has been reported that 15–23% of X-linked genes escape inactivation [95]. Several immune-related genes, such as some toll-like receptors (TLRs), are found on the X chromosome [96]. TLRs are important pathogen-sensing receptors that respond to a variety of microbial ligands and initiate both innate and adaptive immune responses to infection. For example, TLR-7 and TLR-8, located on the X chromosome, may recognize single-stranded viral RNA (ssRNA), including HIV genomic RNA. Genes such as TLR-7 that express themselves biallelically produce larger quantities of protein, which may contribute to a stronger innate immune response [96,97,98]. Studies have shown that female plasmacytoid dendritic cells (pDCs) exhibit an enhanced TLR7-mediated interferon-alpha (IFN-α) response compared to male pDCs, commonly observed in the first week of HIV infection [99]. This increased immune response could contribute to the observed differences in disease progression and treatment outcomes between sexes.
Elite controllers and the key for a functional cure
On top of everything, when exploring HIV curative avenues, it is essential to consider the unique characteristics of elite controllers. Elite controllers comprise of less than 1% of PLHIV across several multicenter cohorts, who maintain undetectable viral loads and stable CD4 + T cell counts without treatment [21, 100]. Holding the key to an HIV cure, elite controllers have been proven to have a unique genetic makeup, a potent T cell response, and keep HIV at bay in ‘gene deserts’, regions of the chromosomes which prevent the virus from being transcribed. These traits differentiate elite controllers from progressors of HIV [101,102,103].
Intriguingly, studies indicate a higher proportion of elite controllers are females, underscoring the importance of investigating gender-specific factors in HIV research [101]. In the case of Loreen Willenberg, often referred to as the San Francisco patient, researchers sampled over 184 million CD4 + T cell genomes from her body and were unable to find any replication-competent or intact HIV DNA [104]. Similarly, the Esperanza patient from Argentina, diagnosed in 2013, was on ART only briefly during her two pregnancies. Despite this, no intact or replication-competent virus has been detected in her body [105]. These cases highlight the potential role of sex-specific immune responses in achieving functional cures and the need for further research into the underlying biological mechanisms. Therefore, the underrepresentation of women in HIV clinical trials may prevent crucial information in developing effective treatments for everyone affected by HIV.
Conclusions
Finding a definitive cure for HIV remains one of the most complex issues in modern medicine. EBT-101 marks a significant advancement, yet recent clinical trials reveal the complexities of translating preclinical success into clinical outcomes, with viral rebound after ART interruption. Addressing these complexities necessitates a multifaceted approach that incorporates genetic diversity, epigenetic regulation, post-transcriptional modifications, chromatin remodeling, and sex-specific factors, as summarized in Table 1.
With diverse subtypes and recombinant forms, the genetic variability of HIV presents profound implications on treatment efficacy and disease progression. For example, a different treatment strategy might need to be considered for those with fast-progressing subtypes. The genetic heterogeneity of HIV, with its various subtypes and recombinant forms, emphasizes the need for a personalized approach to HIV treatment. This strategy is especially important for fast-progressing subtypes, which may respond differentially to ART. A personalized treatment strategy that considers pharmacogenomic parameters, such as host genetic variability, can improve ART efficacy and reduce side effects. For example, the HLA-B5701 screening test is a notable pharmacogenomic technique in clinical practice for identifying patients at risk of hypersensitivity to the antiretroviral medication abacavir [106]. Research also indicates that other host genetic factors, including sex, BMI, heredity, and genetic markers, impact ART efficacy and toxicity, leading to variable treatment outcomes among patients [107]. Integrating these pharmacogenomic insights with knowledge of specific HIV subtypes could enhance treatment outcomes, potentially reducing mortality and improving quality of life for individuals, especially those with fast-progressing forms of the virus.
Additionally, it is crucial to have a nuanced understanding of the intricate mechanisms governing epigenetic regulation, particularly chromatin remodeling and RNA modifications. These mechanisms play a pivotal role in elucidating HIV latency, viral replication dynamics, and immune evasion strategies. Given these findings, adding epigenetic profiling into HIV treatment regimens represents a possible path towards personalized therapy. Patients with high levels of m6A alterations may benefit from approaches that reverse these modifications, increasing ART efficacy by reactivating latent HIV and making it more susceptible to immune clearance. This strategy might be strengthened by using m6A inhibitors in addition to traditional ART, which would reactivate latent HIV reservoirs by increasing viral protein expression, making it easier for the immune system to detect and eradicate the virus [108]. Furthermore, developing small molecule inhibitors that target the writers, erasers, and readers involved in the m6A modification process may boost treatment outcomes by modifying the epigenetic mechanisms that affect HIV latency [109].
Furthermore, sex-specific biological and hormonal variances distinctly affect susceptibility, immune responses, and treatment outcomes. Given these findings, a promising therapeutic approach could involve using estrogen receptor modulators, such as agonists or antagonists, to enhance ART outcomes for women living with HIV. For instance, the modulation of estrogen receptors, particularly ERα, has shown potential in reducing HIV replication and adjusting immune responses. Research suggests that ERα can activate antiviral pathways, potentially explaining sex-specific changes in viral load and immunological responses. Targeting estrogen receptor signaling may be a novel approach to addressing these sex-specific variations, with the goal of improving treatment efficacy and reducing HIV persistence [110, 111]. The success of estrogen receptor antagonists and agonists could be incorporated into future therapies. Similarly, lessons learned from elite controllers suggests that ensuring that there is a higher rate of women representation in trials might provide more clues to an HIV cure. To effectively tackle these challenges, a holistic and inclusive approach to HIV research and treatment is paramount. By integrating genetic, epigenetic, and sex-specific considerations into clinical trial designs, a better development of personalized and equitable therapeutic interventions can be achieved. This comprehensive understanding of HIV will serve as the cornerstone for innovative strategies that cater to the diverse needs of affected populations, advancing the collective effort in the global fight against HIV/AIDS.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- AAV:
-
Adeno-associated virus
- ART:
-
Antiretroviral therapy
- ATP:
-
Adenosine triphosphate
- AZT:
-
Azidothymidine
- BACH2:
-
BTB and CNC homology 2
- BTB domain:
-
Broad-complex, tramtrack, and bric-à-brac domain
- CAB:
-
Cabotegravir
- CART:
-
Combination antiretroviral therapy
- CCR5:
-
C-C chemokine receptor type 5
- CDK9:
-
Cyclin-dependent kinase 9
- CNC homology:
-
Cap’n’collar homology
- CRF:
-
Circulating recombinant form
- CRISPR:
-
Clustered regularly interspaced short palindromic repeats
- DIS:
-
Dimerization initiation site
- DNA:
-
Deoxyribonucleic acid
- DNMTs:
-
DNA methyltransferases
- DRACH:
-
D = A/G/U, R = A/G, A = adenine, C = cytosine, H = A/C/U (m6A RNA modification motif)
- DRMs:
-
Drug resistance mutations
- FDA:
-
Food and drug administration
- GAG:
-
Group-specific antigen
- H3k9me3:
-
Histone 3 lysine 9 trimethylation
- HATs:
-
Histone acetyltransferases
- HDAC:
-
Histone deacetylase
- HDACis:
-
Histone deacetylase inhibitors
- HDACs:
-
Histone deacetylases
- HEK293T:
-
Human embryonic kidney 293T cells
- HIV:
-
Human immunodeficiency virus
- HMTis:
-
Histone methyltransferase inhibitors
- IFNα:
-
Interferon alpha
- JAK/STAT:
-
Janus kinase/signal transducer and activator of transcription
- LEN:
-
Lenacapavir
- LRA:
-
Latency-reversing agent
- LTR:
-
Long terminal repeat
- MAPK/ERK:
-
Mitogen-activated protein kinase/extracellular signal-regulated kinase
- m6A:
-
N6-methyladenosine
- METTL3/14:
-
Methyltransferase like protein 3/14
- mRNA:
-
Messenger RNA
- mTOR:
-
Mechanistic target of rapamycin
- NVP:
-
Nevirapine
- NF-κB:
-
Nuclear factor kappa B
- PBS:
-
Primer binding site
- PDCS:
-
Plasmacytoid dendritic cells
- PI3K/AKT/mTOR:
-
Phosphoinositide 3-kinase/protein kinase b/mechanistic target of rapamycin
- PKC:
-
Protein kinase C
- p-TEFb/CDK9:
-
Positive transcription elongation factor b/cyclin-dependent kinase 9
- PREP:
-
Pre-exposure prophylaxis
- PTM:
-
Post-translational modification
- RNA:
-
Ribonucleic acid
- RNAP-II:
-
RNA polymerase II
- RRE:
-
Rev response element
- RPV:
-
Rilpivirine
- RT:
-
Reverse transcription
- SIV:
-
Simian immunodeficiency virus
- ssRNA:
-
Single-stranded RNA
- TGF-β:
-
Transforming growth factor beta
- TLRs:
-
Toll-like receptors
- VPR:
-
Viral protein R
- VPR BP:
-
VPR binding protein
- WNt/β-Catenin:
-
Wnt signaling pathway/beta-catenin
- ZfPB-362-VPR:
-
Zinc finger protein binding to VPR
References
aidsmap.com. 2024 [cited 2024 Aug 21]. CRISPR gene therapy EBT-101 does not prevent HIV viral rebound. https://www.aidsmap.com/news/may-2024/crispr-gene-therapy-ebt-101-does-not-prevent-hiv-viral-rebound
Burdo TH, Chen C, Kaminski R, Sariyer IK, Mancuso P, Donadoni M, et al. Preclinical safety and biodistribution of CRISPR targeting SIV in non-human primates. Gene Ther. 2024;31:5–6.
ClinicalTrials.gov. 2024 [cited 2024 Aug 21]. Study of EBT-101 in Aviremic HIV-1 Infected Adults on Stable ART. https://clinicaltrials.gov/study/NCT05144386?a=13
Cohrt KO, News. Excision’s EBT-101 demonstrates safety in clinical trial but does not cure HIV - CRISPR Medicine. [cited 2024 Aug 21]. https://crisprmedicinenews.com/news/excisions-ebt-101-demonstrates-safety-in-clinical-trial-but-does-not-cure-hiv/#:~:text=EBT-101 is well tolerated,not prevent HIV-1 rebound&text = Interim clinical data from the,biodistribution data to 48 weeks.
Ye F, Alvarez-Carbonell D, Nguyen K, Leskov K, Garcia-Mesa Y, Sreeram S et al. Recruitment of the CoREST transcription repressor complexes by Nerve Growth factor IB-like receptor (Nurr1/NR4A2) mediates silencing of HIV in microglial cells. PLoS Pathog. 2022;18(7).
Ophinni Y, Inoue M, Kotaki T, Kameoka M. CRISPR/Cas9 system targeting regulatory genes of HIV-1 inhibits viral replication in infected T-cell cultures. Sci Rep. 2018;8(1).
UNAIDS. Core Epidemiology slides. 2024 [cited 2024 Aug 21]. https://www.unaids.org/en/resources/documents/2023/core-epidemiology-slides
Forsythe SS, McGreevey W, Whiteside A, Shah M, Cohen J, Hecht R et al. Twenty years of antiretroviral therapy for people living with hiv: global costs, health achievements, economic benefits. Health Aff. 2019;38(7).
Iacob SA, Iacob DG, Jugulete G. Improving the adherence to antiretroviral therapy, a difficult but essential task for a successful HIV treatment-clinical points of view and practical considerations. 8, Frontiers in Pharmacology. 2017.
Rutstein SE, Sibley AL, Huffstetler HE, Nguyen TT, Do, Tran HV, Le Minh G et al. Acceptability and feasibility of long-acting injectable antiretroviral therapy for HIV-infected persons who inject drugs in Vietnam: a qualitative study. Lancet Reg Heal - West Pac. 2023;31.
Swindells S, Andrade-Villanueva JF, Richmond GJ, Rizzardini G, Baumgarten A, Masiá M et al. Long-Acting Cabotegravir and Rilpivirine for maintenance of HIV-1 suppression. N Engl J Med. 2020;382(12).
Bekerman E, Yant SR, VanderVeen L, Hansen D, Lu B, Rowe W et al. Long-acting lenacapavir acts as an effective preexposure prophylaxis in a rectal SHIV challenge macaque model. J Clin Invest. 2023;133(16).
De Scheerder MA, Vrancken B, Dellicour S, Schlub T, Lee E, Shao W et al. HIV Rebound is predominantly fueled by genetically identical viral expansions from Diverse reservoirs. Cell Host Microbe. 2019;26(3).
Geretti AM, Harrison L, Green H, Sabin C, Hill T, Fearnhill E et al. Effect of HIV-1 subtype on virologic and immunologic response to starting highly active antiretroviral therapy. Clin Infect Dis. 2009;48(9).
HIV Sequence databases. [cited 2024 Aug 21]. https://www.hiv.lanl.gov/components/sequence/HIV/geo/geo.comp
Ndashimye E, Reyes PS, Arts EJ. New antiretroviral inhibitors and HIV-1 drug resistance: more focus on 90% HIV-1 isolates? Vol. 47, FEMS microbiology reviews. 2023.
Loosli T, Hossmann S, Ingle SM, Okhai H, Kusejko K, Mouton J et al. HIV-1 drug resistance in people on dolutegravir-based ART: a collaborative cohort analysis. The Lancet HIV. 2023;10(11):e733–41.
Posada-Céspedes S, van Zyl G, Montazeri H, Kuipers J, Rhee SY, Kouyos R et al. Comparing mutational pathways to lopinavir resistance in HIV-1 subtypes B versus C. 17, PLoS Comput Biol. 2021.
Suh AJ, Suzuki DI, Brelidze TI, Suzuki YJ. Gp120 envelope glycoproteins of HIV-1 Group M subtype A and subtype B differentially affect gene expression in human vascular endothelial cells. J Immunol. 2023;210(1_Supplement).
Zhou Z, Tang K, Zhang G, Wadonda-Kabondo N, Moyo K, Rowe LA et al. Detection of minority drug resistant mutations in Malawian HIV-1 subtype C-positive patients initiating and on first-line antiretroviral therapy. Afr J Lab Med. 2018;7(1).
Deeks SG, Lewin SR, Ross AL, Ananworanich J, Benkirane M, Cannon P et al. International AIDS Society global scientific strategy: towards an HIV cure 2016. 22, Nat Med. 2016.
Zhang J, Crumpacker C. HIV UTR, LTR, and epigenetic immunity. Viruses. 2022;14(5).
Ospelt C. A brief history of epigenetics. Immunol Lett. 2022;249.
Sengupta S, Siliciano RF. Targeting the Latent Reservoir for HIV-1. Vol. 48, Immunity. 2018.
Chen L, Zhang S, Pan X, Hu XH, Zhang YH, Yuan F et al. HIV infection alters the human epigenetic landscape. Gene Ther. 2019;26(1–2).
Lu K, Yip YM. Therapeutic potential of Bioactive flavonoids from Citrus Fruit peels toward obesity and diabetes Mellitus. Futur Pharmacol. 2023;3(1).
Alamer E, Zhong C, Hajnik R, Soong L, Hu H. Modulation of BRD4 in HIV epigenetic regulation: implications for finding an HIV cure. Vol. 18, Retrovirology. 2021.
Asamitsu K, Fujinaga K, Okamoto T. HIV Tat/P-TEFb interaction: A potential target for novel anti-HIV therapies. Vol. 23, Molecules. 2018.
Mori L, Valente ST. Key players in HIV-1 transcriptional regulation: Targets for a functional cure. Vol. 12, Viruses. 2020.
Rodari A, Darcis G, Van Lint CM. The Current Status of Latency Reversing Agents for HIV-1 Remission. Vol. 8, Annual Review of Virology. 2021.
Campbell GR, Spector SA. Current strategies to induce selective killing of HIV-1-infected cells. 112, J Leukoc Biol. 2022.
McMahon DK, Zheng L, Cyktor JC, Aga E, Macatangay BJ, Godfrey C et al. A phase 1/2 Randomized, Placebo-Controlled Trial of Romidespin in persons with HIV-1 on suppressive antiretroviral therapy. J Infect Dis. 2021;224(4).
Rasmussen TA, Tolstrup M, Winckelmann A, Østergaard L, Søgaard OS. Eliminating the latent HIV reservoir by reactivation strategies: advancing to clinical trials. Hum Vaccines Immunother. 2013;9(4).
Yukl SA, Kaiser P, Kim P, Telwatte S, Joshi SK, Vu M et al. HIV latency in isolated patient CD4 + T cells may be due to blocks in HIV transcriptional elongation, completion, and splicing. Sci Transl Med. 2018;10(430).
Jiang G, Dandekar S. Targeting NF-κB signaling with protein Kinase C agonists as an emerging strategy for combating HIV latency. Vol. 31, AIDS Research and Human Retroviruses. 2015.
Margolis DM. Confronting proviral HIV infection. Curr HIV/AIDS Rep. 2007;4(2).
Gutiérrez C, Serrano-Villar S, Madrid-Elena N, Pérez-Eĺas MJ, Martń ME, Barbas C et al. Bryostatin-1 for latent virus reactivation in HIV-infected patients on antiretroviral therapy. AIDS. 2016;30(9).
Ait-Ammar A, Kula A, Darcis G, Verdikt R, De Wit S, Gautier V et al. Current status of latency reversing agents facing the heterogeneity of HIV-1 Cellular and tissue reservoirs. 10, Front Microbiol. 2020.
Xu X, Lin Y, Zeng X, Yang C, Duan S, Ding L et al. PARP1 Might Substitute HSF1 to Reactivate Latent HIV-1 by Binding to Heat Shock Element. Cells. 2022;11(15).
Zhao M, De Crignis E, Rokx C, Verbon A, van Gelder T, Mahmoudi T et al. T cell toxicity of HIV latency reversing agents. Pharmacol Res. 2019;139.
Kim Y, Anderson JL, Lewin SR. Getting the kill into shock and kill: strategies to Eliminate Latent HIV. Volume 23. Cell Host and Microbe; 2018.
Abner E, Jordan A. HIV shock and kill therapy: in need of revision. 166, Antiviral Res. 2019.
Chmielowska-Bąk J, Searle IR, Wakai TN, Arasimowicz-Jelonek M. The role of epigenetic and epitranscriptomic modifications in plants exposed to non-essential metals. 14, Frontiers in Plant Science. 2023.
Janin M, Davalos V, Esteller M. Cancer metastasis under the magnifying glass of epigenetics and epitranscriptomics. 42, Cancer Metastasis Rev. 2023.
Boccaletto P, Stefaniak F, Ray A, Cappannini A, Mukherjee S, Purta E, et al. MODOMICS: a database of RNA modification pathways. 2021 update. Nucleic Acids Res. 2022;50:D1.
Hashiguchi A, Komatsu S. Posttranslational Modifications and Plant–Environment Interaction. In: Methods in Enzymology. 2017.
Lichinchi G, Gao S, Saletore Y, Gonzalez GM, Bansal V, Wang Y et al. Dynamics of the human and viral m6A RNA methylomes during HIV-1 infection of T cells. Nat Microbiol. 2016;1(4):1–9.
Li H, Guo Y, Qi W, Liao M. N6-methyladenosine modification of viral RNA and its role during the recognition process of RIG-I-like receptors. 13, Frontiers in Immunology. 2022.
Cristinelli S, Angelino P, Janowczyk A, Delorenzi M, Ciuffi A. HIV modifies the m6A and m5C Epitranscriptomic Landscape of the host cell. Front Virol. 2021;1.
Pereira-Montecinos C, Toro-Ascuy D, Ananías-Sáez C, Gaete-Argel A, Rojas-Fuentes C, Riquelme-Barrios S et al. Epitranscriptomic regulation of HIV-1 full-length RNA packaging. Nucleic Acids Res. 2022;50(4).
Tirumuru N, Wu L. HIV-1 envelope proteins up-regulate N6-methyladenosine levels of cellular RNA independently of viral replication. J Biol Chem. 2019;294(9).
Lu W, Tirumuru N, Gelais CS, Koneru PC, Liu C, Kvaratskhelia M et al. N6-Methyladenosine– binding proteins suppress HIV-1 infectivity and viral production. J Biol Chem. 2018;293(34).
Baek A, Lee GE, Golconda S, Rayhan A, Manganaris AA, Chen S et al. Single-molecule epitranscriptomic analysis of full-length HIV-1 RNAs reveals functional roles of site-specific m6As. Nat Microbiol. 2024;9(5):1–6.
Phillips S, Mishra T, Huang S, Wu L. Functional Impacts of Epitranscriptomic m6A Modification on HIV-1 Infection. Vol. 16, Viruses. 2024.
Liu S, Chen S, Tang C, Zhao Y, Cui W, Jia L et al. The emerging therapeutic target of dynamic and reversible N6-methyladenosine modification during cancer development. 12, Front Oncol. 2022.
López-Maury L, Marguerat S, Bähler J. Tuning gene expression to changing environments: from rapid responses to evolutionary adaptation. 9, Nat Rev Genet. 2008.
Morrison AJ. Chromatin-remodeling links metabolic signaling to gene expression. Vol. 38, Molecular Metabolism. 2020.
Alfert A, Moreno N, Kerl K. The BAF complex in development and disease. Vol. 12, Epigenetics and Chromatin. 2019.
Sharma AL, Hokello J, Sonti S, Zicari S, Sun L, Alqatawni A et al. CBF-1 promotes the establishment and maintenance of HIV latency by Recruiting Polycomb Repressive Complexes, PRC1 and PRC2, at HIV LTR. Viruses. 2020;12(9).
Lusic M, Giacca M. Regulation of HIV-1 latency by chromatin structure and Nuclear Architecture. 427, J Mol Biol. 2015.
Battivelli E, Dahabieh MS, Abdel-Mohsen M, Svensson JP, Da Silva IT, Cohn LB et al. Distinct chromatin functional states correlate with HIV latency reactivation in infected primary CD4 + T cells. Elife. 2018;7.
Van Lint C, Bouchat S, Marcello A. HIV-1 transcription and latency: An update. Vol. 10, Retrovirology. 2013.
Venkatesh S, Workman JL. Histone exchange, chromatin structure and the regulation of transcription. 16, Nat Rev Mol Cell Biol. 2015.
Ding J, Wang S, Wang Z, Chen S, Zhao J, Solomon M et al. Schlafen 5 suppresses human immunodeficiency virus type 1 transcription by commandeering cellular epigenetic machinery. Nucleic Acids Res. 2022;50(11).
Ma L, Jiang QA, Sun L, Yang X, Huang H, Jin X et al. X-linked rna-binding motif protein modulates hiv-1 infection of cd4 + T cells by maintaining the trimethylation of histone h3 lysine 9 at the downstream region of the 5 = long terminal repeat of hiv proviral dna. MBio. 2020;11(2).
Sloan RD, Wainberg MA. The role of unintegrated DNA in HIV infection. Vol. 8, Retrovirology. 2011.
Meltzer B, Dabbagh D, Guo J, Kashanchi F, Tyagi M, Wu Y. Tat controls transcriptional persistence of unintegrated HIV genome in primary human macrophages. Virology. 2018;518.
Zerbato JM, Khoury G, Zhao W, Gartner MJ, Pascoe RD, Rhodes A et al. Multiply spliced HIV RNA is a predictive measure of virus production ex vivo and in vivo following reversal of HIV latency. EBioMedicine. 2021;65.
Geis FK, Goff SP. Unintegrated HIV-1 DNAs are loaded with core and linker histones and transcriptionally silenced. Proc Natl Acad Sci U S A. 2019;116:47.
Bondarev AD, Attwood MM, Jonsson J, Chubarev VN, Tarasov VV, Schiöth HB. Recent developments of HDAC inhibitors: emerging indications and novel molecules. 87, Br J Clin Pharmacol. 2021.
Huynh NCN, Everts V, Ampornaramveth RS. Histone deacetylases and their roles in mineralized tissue regeneration. 7, Bone Rep. 2017.
Ngo MH, Pankrac J, Ho RCY, Ndashimye E, Pawa R, Ceccacci R et al. Effective and targeted latency reversal in CD4 + T cells from individuals on long term combined antiretroviral therapy initiated during chronic HIV-1 infection. Emerg Microbes Infect. 2024;13(1).
Bullen CK, Laird GM, Durand CM, Siliciano JD, Siliciano RF. New ex vivo approaches distinguish effective and ineffective single agents for reversing HIV-1 latency in vivo. Nat Med. 2014;20(4).
Cary DC, Fujinaga K, Peterlin BM. Molecular mechanisms of HIV latency. 126, J Clin Invest. 2016.
Khanal S, Schank M, Gazzar M, El, Moorman JP, Yao ZQ. Hiv-1 latency and viral reservoirs: Existing reversal approaches and potential technologies, targets, and pathways involved in hiv latency studies. Vol. 10, Cells. 2021.
Wong LM, Jiang G. NF-κB sub-pathways and HIV cure: A revisit. Vol. 63, EBioMedicine. 2021.
Spivak AM, Planelles V. Novel latency reversal agents for HIV-1 cure. Volume 69. Annual Review of Medicine; 2018.
Zheng T, Chen P, Huang Y, Qiu J, Zhou C, Wu Z et al. CPI-637 as a potential Bifunctional latency-reversing Agent that targets both the BRD4 and TIP60 proteins. Front Cell Infect Microbiol. 2021;11.
Wen J, Li X, Zhao QX, Yang XF, Wu ML, Yan Q et al. Pharmacological suppression of glycogen synthase kinase-3 reactivates HIV-1 from latency via activating Wnt/β-catenin/TCF1 axis in CD4 + T cells. Emerg Microbes Infect. 2022;11(1).
Romani B, Kamali Jamil R, Hamidi-Fard M, Rahimi P, Momen SB, Aghasadeghi MR et al. HIV-1 vpr reactivates latent HIV-1 provirus by inducing depletion of class i HDACs on chromatin. Sci Rep. 2016;6.
Forouzanfar F, Ali S, Wallet C, De Rovere M, Ducloy C, El Mekdad H et al. HIV-1 vpr mediates the depletion of the cellular repressor CTIP2 to counteract viral gene silencing. Sci Rep. 2019;9(1).
Scott TA, O’Meally D, Grepo NA, Soemardy C, Lazar DC, Zheng Y et al. Broadly active zinc finger protein-guided transcriptional activation of HIV-1. Mol Ther Methods Clin Dev. 2021;20:18–29.
Baumbach JL, Zovkic IB. Hormone-epigenome interactions in behavioural regulation. Vol. 118, Hormones and Behavior. 2020.
UNAIDS. No Title. 2023. https://www.unaids.org/en/resources/fact-sheet
Curno MJ, Rossi S, Hodges-Mameletzis I, Johnston R, Price MA, Heidari S. A systematic review of the inclusion (or Exclusion) of women in HIV Research: from Clinical studies of antiretrovirals and vaccines to cure strategies. J Acquir Immune Defic Syndr. 2016;71(2).
Parekh A, Fadiran EO, Uhl K, Throckmorton DC. Adverse effects in women: implications for drug development and regulatory policies. 4, Expert Review of Clinical Pharmacology. 2011.
Santinelli L, Ceccarelli G, Borrazzo C, Innocenti G, Pietro, Frasca F, Cavallari EN et al. Sex-related differences in markers of immune activation in virologically suppressed HIV-infected patients. Biol Sex Differ. 2020;11(1).
Ziegler S, Altfeld M. Sex differences in HIV-1-mediated immunopathology. Vol. 11, Current Opinion in HIV and AIDS. 2016.
Masenga SK, Mweene BC, Luwaya E, Muchaili L, Chona M, Kirabo A. HIV–Host Cell Interactions. Vol. 12, Cells. 2023.
Prodger JL, Gray R, Kigozi G, Nalugoda F, Galiwango R, Hirbod T et al. Foreskin T-cell subsets differ substantially from blood with respect to HIV co-receptor expression, inflammatory profile, and memory status. Mucosal Immunol. 2012;5(2).
Plesniarski A, Siddik AB, Su RC. The Microbiome as a Key Regulator of female genital tract barrier function. 11, Frontiers in Cellular and Infection Microbiology. 2021.
Hall OJ, Klein SL. Progesterone-based compounds affect immune responses and susceptibility to infections at diverse mucosal sites. Vol. 10, Mucosal Immunology. 2017.
Das B, Dobrowolski C, Luttge B, Valadkhan S, Chomont N, Johnston R, et al. Estrogen receptor-1 is a key regulator of HIV-1 latency that imparts gender-specific restrictions on the latent reservoir. Proc Natl Acad Sci U S A. 2018;115:33.
Shepherd R, Bretherton I, Pang K, Mansell T, Czajko A, Kim B et al. Gender-affirming hormone therapy induces specific DNA methylation changes in blood. Clin Epigenetics. 2022;14(1).
Souyris M, Cenac C, Azar P, Daviaud D, Canivet A, Grunenwald S et al. TLR7 escapes X chromosome inactivation in immune cells. Sci Immunol. 2018;3(19).
Fathi A, Addo MM, Dahlke C. Sex differences in immunity: implications for the development of Novel vaccines against emerging pathogens. 11, Front Immunol. 2021.
Browne EP. The role of toll-like receptors in retroviral infection. Vol. 8, Microorganisms. 2020.
Souyris M, Mejía JE, Chaumeil J, Guéry JC. Female predisposition to TLR7-driven autoimmunity: gene dosage and the escape from X chromosome inactivation. 41, Seminars in Immunopathology. 2019.
Guéry JC. Sex differences in primary HIV infection: revisiting the role of TLR7-Driven type 1 IFN Production by Plasmacytoid dendritic cells in women. 12, Frontiers in Immunology. 2021.
Okulicz JF, Marconi VC, Landrum ML, Wegner S, Weintrob A, Ganesan A et al. Clinical outcomes of elite controllers, viremic controllers, and long-term nonprogressors in the US department of defense HIV natural history study. J Infect Dis. 2009;200(11).
Mastrangelo A, Banga R, Perreau M. Elite and posttreatment controllers, two facets of HIV control. Vol. 17, Current Opinion in HIV and AIDS. 2022.
Bendenoun M, Samri A, Avettand-Fènoël V, Cardinaud S, Descours B, Carcelain G et al. What Is the most Important for Elite Control: Genetic Background of Patient, Genetic Background of Partner, both or neither? Description of Complete Natural History within a Couple of MSM. EBioMedicine. 2018;27.
Jiang C, Lian X, Gao C, Sun X, Einkauf KB, Chevalier JM, et al. A unique viral reservoir landscape in HIV-1 elite controllers. Nature. 2020;585:7824.
Dubé K, Willenberg L, Dee L, Sylla L, Taylor J, Roebuck C et al. Re-examining the HIV ‘functional cure’ oxymoron: time for precise terminology? J Virus Erad. 2020;6(4).
amfAR, The Foundation for AIDS Research. 2024 [cited 2024 Aug 21]. Curing HIV—How far have we come? - AMFAR, the Foundation for AIDS Research. https://www.amfar.org/news/how-many-have-been-cured/#:~:text=San Francisco patient – Loreen Willenberg,to control her HIV infection.
Aceti A. Pharmacogenetics as a tool to tailor antiretroviral therapy: a review. World J Virol. 2015;4(3).
Dalal B, Shankarkumar A, Ghosh K. Individualization of antiretroviral therapy - pharmacogenomic aspect. 142, Indian J Med Res. 2015.
Chen S, Kumar S, Espada CE, Tirumuru N, Cahill MP, Hu L et al. N6-methyladenosine modification of HIV-1 RNA suppresses type-I interferon induction in differentiated monocytic cells and primary macrophages. PLoS Pathog. 2021;17(3).
Wu F, Cheng W, Zhao F, Tang M, Diao Y, Xu R. Association of N6-methyladenosine with viruses and related diseases. 16, Virol J. 2019.
Griesbeck M, Scully E, Altfeld M. Sex and gender differences in HIV-1 infection. Clin Sci. 2016;130(16).
Szotek EL, Narasipura SD, Al-Harthi L. 17β-Estradiol inhibits HIV-1 by inducing a complex formation between β-catenin and estrogen receptor α on the HIV promoter to suppress HIV transcription. Virology. 2013;443(2).
Funding
Not applicable.
Author information
Authors and Affiliations
Contributions
PL wrote the manuscript. KTD made equal contributions in the development and revision of the manuscript. All authors reviewed the manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Both authors consent to publication.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Letchumanan, P., Theva Das, K. The role of genetic diversity, epigenetic regulation, and sex-based differences in HIV cure research: a comprehensive review. Epigenetics & Chromatin 18, 1 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13072-024-00564-4
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13072-024-00564-4