View Article

Abstract

The establishment of latent HIV reservoirs remains the principal barrier to achieving a cure for HIV infection. While antiretroviral therapy (ART) effectively suppresses viral replication, it cannot eliminate integrated proviral DNA that persists in a transcriptionally silent state within long-lived memory CD4+ T cells. The "Block and Lock" strategy has emerged as a promising alternative to the traditional "Shock and Kill" approach, aiming to permanently silence HIV transcription rather than reactivating latent virus. This review examines the molecular mechanisms underlying HIV latency, critically evaluates current Block and Lock strategies including didehydro-Cortistatin A (dCA) and Tat inhibitors, and discusses the translational challenges and future directions for achieving a functional HIV cure. Recent advances in our understanding of epigenetic regulation, the role of the viral transactivator Tat, and the heterogeneity of latent reservoirs provide new opportunities for therapeutic intervention.

Keywords

HIV latency, Block and Lock, latency-reversing agents, shock and kill, dCA, Tat inhibitors, HIV reservoir, functional cure, HIV transcription, epigenetic silencing

Introduction

Human Immunodeficiency Virus (HIV) infection continues to pose a significant global health challenge, affecting approximately 39 million people worldwide as of 2023 ([1]). While combination antiretroviral therapy (ART) has transformed HIV from a fatal disease into a manageable chronic condition, it requires lifelong adherence and does not constitute a cure ([2]). The primary obstacle to HIV eradication is the establishment of latent viral reservoirs, predominantly within resting memory CD4+ T cells, where integrated proviral DNA persists in a transcriptionally silent state ([3]). The latent reservoir is established early during acute infection, even before ART initiation, and exhibits remarkable stability with an estimated half-life of 44 months or longer ([4]). Upon treatment interruption, latent proviruses can reactivate, leading to viral rebound within weeks. This necessitates continuous ART administration, which is associated with long-term toxicities, drug resistance, cost burden, and challenges in resource-limited settings ([5]). Two major strategies have emerged to address the latent reservoir: "Shock and Kill" and "Block and Lock." The Shock and Kill approach aims to reactivate latent virus using latency-reversing agents (LRAs) while maintaining ART, with the expectation that reactivated infected cells will be eliminated by cytotoxic immune responses or viral cytopathic effects ([6]). However, clinical trials have demonstrated that current LRAs fail to significantly reduce the reservoir size, raising concerns about incomplete reactivation, immune escape, and potential systemic inflammation ([7]). In contrast, the Block and Lock strategy seeks to permanently silence HIV transcription by promoting deep latency through epigenetic modifications and inhibition of viral transactivation ([8]). This approach offers several theoretical advantages: it avoids the risks associated with viral reactivation, does not require immune-mediated clearance, and may allow for ART discontinuation if silencing is sufficiently robust and durable ([9]). This review provides a comprehensive analysis of the molecular basis of HIV latency, examines emerging Block and Lock therapeutic candidates, and discusses the challenges and opportunities in translating these strategies to clinical application.

2. Molecular Mechanisms of HIV Latency

2.1 Establishment of Latency

HIV latency is established through multiple interconnected mechanisms operating at transcriptional, post-transcriptional, and epigenetic levels ([10]). Following integration of viral DNA into the host genome, the fate of infected cells depends on their activation state, the integration site, and the local chromatin environment. Latency occurs predominantly when HIV integrates into transcriptionally inactive regions of the genome or when activated CD4+ T cells transition to a resting memory phenotype before completing the viral replication cycle ([11]).

Fig.1. Mechanism of HIV Latency

2.2 Transcriptional Regulation and the Role of Tat

The HIV long terminal repeat (LTR) promoter regulates viral gene expression and is subject to both positive and negative regulatory mechanisms ([12]). The viral transactivator protein Tat is essential for efficient HIV transcription, binding to the trans-activation response (TAR) element in nascent viral RNA and recruiting the positive transcription elongation factor b (P-TEFb) complex. This interaction enhances RNA polymerase II processivity and enables productive viral transcription ([13]). In the absence of Tat or under conditions of limited Tat availability, transcriptional elongation is inefficient, resulting in the production of short, non-coding transcripts. This state of transcriptional insufficiency contributes to the establishment and maintenance of latency. Additionally, cellular transcription factors such as NF-κB and NFAT, which are activated during T cell stimulation, are largely inactive in resting cells, further suppressing LTR-driven transcription ([10]).

Fig.2. Transcriptional Regulation and the role of Tat

2.3 Epigenetic Silencing

Epigenetic modifications play a crucial role in maintaining HIV latency ([14]). The HIV LTR exists within a nucleosome structure, and chromatin remodeling is essential for transcriptional activation. In latently infected cells, the LTR is characterized by restrictive histone modifications including histone deacetylation, H3K9 trimethylation (H3K9me3), and H3K27 trimethylation (H3K27me3), which are associated with transcriptional repression ([15]). DNA methylation of CpG islands within the HIV LTR also contributes to transcriptional silencing, although the extent and significance of this modification remain debated ([16]). Histone deacetylases (HDACs), histone methyltransferases (HMTs), and DNA methyltransferases (DNMTs) are recruited to the LTR through interactions with cellular repressor complexes, establishing a stable heterochromatic state that is refractory to transcriptional activation.

Fig.3. Epigenetic Silencing of HIV LTR Chromatin

2.4 Reservoir Heterogeneity

Recent studies have revealed considerable heterogeneity within the latent reservoir, including variations in integration sites, proviral integrity, and cellular phenotypes ([17]). A substantial proportion of integrated proviruses harbor deleterious mutations or deletions that render them replication-incompetent, yet these defective proviruses can still produce viral proteins and contribute to immune activation. The intact reservoir, capable of producing replication-competent virus, is significantly smaller than total HIV DNA measurements suggest, complicating efforts to assess reservoir size and therapeutic efficacy ([18]).

Fig.4. HIV Reservoir Heterogeneity

3. The Block and Lock Strategy: Conceptual Framework

The Block and Lock strategy represents a paradigm shift from attempts to purge the reservoir to permanently inactivating it ([8]). The fundamental principle is to drive latent proviruses into a state of profound and irreversible transcriptional silencing, thereby preventing viral rebound even in the absence of ART. This approach is predicated on the observation that not all latent proviruses are equally poised for reactivation; some exist in deeper latency states characterized by more restrictive chromatin structures. Block and Lock agents typically function through one or more of the following mechanisms: (1) inhibition of the Tat-TAR-P-TEFb axis, (2) promotion of repressive chromatin modifications, (3) recruitment of transcriptional repressors to the HIV LTR, and (4) interference with critical cellular transcription factors ([9]). The goal is to achieve a functional cure, defined as sustained viral suppression in the absence of ART without complete eradication of the reservoir.

Fig.5. Block and Lock Strategy for HIV

4. Block and Lock Therapeutic Candidates

4.1 Didehydro-Cortistatin A (dCA)

Didehydro-Cortistatin A (dCA) is the most extensively studied Block and Lock agent and represents a modified derivative of the natural neuropeptide cortistatin ([19]). Preclinical studies have demonstrated that dCA potently inhibits HIV transcription by targeting the Tat protein, thereby disrupting the Tat-TAR-P-TEFb interaction. Importantly, dCA appears to promote deep latency by inducing epigenetic changes at the HIV LTR, including increased histone deacetylation and recruitment of HDACs. In humanized mouse models and ex vivo assays using cells from ART-suppressed individuals, dCA treatment resulted in prolonged suppression of viral reactivation, even after exposure to potent latency-reversing agents ([20]). These findings suggest that dCA-mediated silencing is both robust and durable. However, questions remain regarding the optimal dosing regimen, potential off-target effects on cellular gene expression, and the long-term stability of induced latency in the complex in vivo environment.

Fig.6. Didehydro-Cortistatin A inhibition of HIV Transcription Mechanism

4.2. Tat Inhibitors

Given the central role of Tat in HIV transcription, several classes of Tat inhibitors have been developed as potential Block and Lock agents ([12]). These include small molecules that directly bind to Tat or TAR RNA, compounds that disrupt Tat-P-TEFb interaction, and agents that promote Tat degradation. Didehydro-Cortistatin A itself functions primarily as a Tat inhibitor, but other compounds such as triptolide and specific cyclin-dependent kinase (CDK) inhibitors have shown promise in preclinical studies. The selectivity of Tat inhibitors is critical, as P-TEFb regulates the transcription of many cellular genes involved in cell cycle progression and immune function ([13]). Therefore, therapeutic Tat inhibitors must achieve sufficient specificity for the HIV Tat-P-TEFb complex to minimize adverse effects on normal cellular processes.


Fig.7. Tat inhibitors Targeting HIV Transcription

4.3 LEDGINs and Integration Site Targeting

LED209 and related compounds, known as LEDGINs (LEDGF/p75-IN disruptors), inhibit HIV replication by interfering with the interaction between the viral integrase and the cellular factor LEDGF/p75 ([11]). Beyond their antiviral activity, LEDGINs have been proposed as potential Block and Lock agents based on observations that they alter integration site selection, favoring integration into transcriptionally inactive chromatin regions. This mechanism could promote the establishment of deeper latency and reduce the likelihood of viral reactivation. However, the clinical development of LEDGINs for Block and Lock purposes remains in early stages, and additional studies are needed to determine whether altered integration patterns translate to meaningful reductions in reservoir reactivation potential ([14]).

Fig.8. LEDGINs and Integration Site Targeting

4.4 Epigenetic Modifiers

Several epigenetic modifiers that promote transcriptional repression have been investigated as Block and Lock candidates ([15]). These include HDAC activators, histone methyltransferase activators, and DNA methyltransferase modulators. The challenge with this approach lies in achieving HIV-specific epigenetic modifications without causing widespread changes in cellular gene expression that could compromise immune function or cellular homeostasis. Recent advances in targeted epigenetic editing, such as dCas9-based systems fused to epigenetic modifying enzymes, offer the potential for locus-specific silencing of the HIV LTR ([16]). While still in the experimental phase, these technologies represent an exciting frontier for precise Block and Lock interventions.

Fig.9. Epigenetic Modifiers as Block and Lock Candidates for HIV Transcriptional Repression

5. Preclinical and Clinical Evidence

5.1 In Vitro and Ex Vivo Studies

Multiple studies using cell line models of latency and primary CD4+ T cells from ART-suppressed individuals have demonstrated the efficacy of Block and Lock agents in suppressing viral reactivation ([17]). Treatment with dCA consistently resulted in reduced viral rebound following stimulation with latency-reversing agents such as protein kinase C agonists, HDAC inhibitors, and T cell receptor stimulation. The degree of suppression varied depending on the cellular model and the specific reactivation stimulus employed. Ex vivo studies using the quantitative viral outgrowth assay (QVOA) have shown that dCA treatment can reduce the frequency of reactivatable latent proviruses, although complete elimination has not been achieved ([18]). These findings underscore both the promise and limitations of current Block and Lock strategies.

5.2 Animal Model Studies

Humanized mouse models and non-human primate (NHP) studies have provided critical insights into the in vivo efficacy and safety of Block and Lock agents ([19]). In one pivotal study, dCA administration to HIV-infected humanized mice resulted in sustained viral suppression after ART discontinuation, with significantly delayed or absent viral rebound compared to control animals. Similar results have been observed in SIV-infected macaques treated with Block and Lock compounds. However, these models have limitations, including differences in immune system complexity, reservoir distribution, and viral dynamics compared to human infection ([20]). Additionally, the durability of silencing over the extended timescales relevant to human lifespans remains uncertain.

5.3 Clinical Trials

As of 2024, Block and Lock strategies have not yet advanced to large-scale clinical trials in humans, although early-phase studies are in development ([2]). The translation of preclinical findings to clinical applications faces several challenges, including the need to establish appropriate biomarkers for measuring reservoir silencing, determining optimal dosing and treatment duration, and assessing long-term safety.

6. Challenges and Limitations

6.1 Incomplete Understanding of Latency Mechanisms

Despite significant advances, our understanding of HIV latency remains incomplete ([3]). The molecular determinants of latency depth, the signals that trigger reactivation, and the role of cellular and anatomical reservoir compartments beyond circulating CD4+ T cells are areas of ongoing investigation. This knowledge gap complicates the rational design of Block and Lock interventions.

6.2 Reservoir Heterogeneity and Accessibility

The latent reservoir is highly heterogeneous in terms of cellular phenotypes, integration sites, and proviral sequences ([4]). Block and Lock agents must be effective across this diverse population of latently infected cells. Additionally, anatomical reservoirs in lymphoid tissues, the central nervous system, and other sites may be less accessible to therapeutic agents, limiting treatment efficacy.

6.3 Potential for Viral Escape and Resistance

While Block and Lock strategies aim to silence rather than eliminate the virus, the potential for viral escape through mutations that resist silencing mechanisms cannot be ignored ([5]). Proviruses with specific LTR sequence variations or those integrated in particularly permissive chromatin contexts may be less susceptible to Block and Lock interventions. Long-term studies will be necessary to assess the stability of induced latency and the risk of escape.

6.4 Off-Target Effects and Safety Concerns

Many Block and Lock agents target cellular proteins or pathways that regulate normal gene expression ([6]). Tat inhibitors that affect P-TEFb function, for example, could impair T cell activation and proliferation, compromising immune responses to other pathogens. Thorough assessment of off-target effects and long-term safety profiles is essential before clinical implementation.

6.5 Biomarkers and Clinical Endpoints

Measuring the efficacy of Block and Lock strategies presents significant challenges ([7]). Traditional measures of reservoir size, such as total HIV DNA or integrated DNA, do not distinguish between deeply latent proviruses and those poised for reactivation. The QVOA, while considered the gold standard, is labor-intensive, requires large blood volumes, and underestimates the true reservoir size. Novel biomarkers that reflect the depth and stability of latency are urgently needed to guide clinical development.

7. Combination Approaches and Future Directions

7.1 Combining Block and Lock with Other Cure Strategies

An emerging consensus suggests that HIV cure strategies may require combination approaches that address multiple aspects of reservoir persistence ([8]). Block and Lock could be combined with immune-based therapies such as therapeutic vaccines, broadly neutralizing antibodies, or checkpoint inhibitors to enhance reservoir control. Additionally, targeting newly infected cells during analytical treatment interruption with Block and Lock agents could prevent reservoir replenishment.

7.2 Personalized Medicine Approaches

The heterogeneity of HIV reservoirs between individuals suggests that personalized approaches may be necessary for optimal outcomes ([9]). Detailed characterization of an individual's reservoir composition, including integration site analysis, proviral sequencing, and assessment of cellular phenotypes, could inform tailored therapeutic strategies. Advances in single-cell technologies and high-throughput sequencing are making such personalized assessments increasingly feasible.

7.3 Novel Delivery Systems

Effective delivery of Block and Lock agents to all relevant anatomical compartments represents a significant challenge ([10]). Nanoparticle-based delivery systems, long-acting formulations, and targeted delivery strategies that exploit specific markers on latently infected cells are under investigation. These approaches may enhance drug bioavailability, reduce systemic exposure and toxicity, and improve penetration into tissue reservoirs.

7.4 Gene Therapy and Editing Approaches

Gene therapy approaches using CRISPR-Cas9 or other editing technologies to modify the HIV LTR, introduce repressive epigenetic marks, or disrupt essential viral genes represent a radical extension of the Block and Lock concept ([16]). While technical challenges remain, including delivery efficiency and off-target effects, these approaches offer the potential for permanent, irreversible silencing of the reservoir.

8. Regulatory and Ethical Considerations

The development and approval of Block and Lock therapeutics will require careful consideration of regulatory frameworks and ethical principles ([1]). Given that these interventions aim to achieve a functional rather than sterilizing cure, defining appropriate clinical endpoints and acceptable safety profiles is complex. The risk-benefit calculation for individuals who are well-controlled on ART differs from those experiencing treatment failure or adverse effects. Informed consent processes must clearly communicate the experimental nature of Block and Lock strategies, the uncertainty regarding long-term efficacy, and the potential risks of treatment interruption studies. Additionally, ensuring equitable access to cure-related research and eventual therapies across diverse populations and geographic regions is an ethical imperative.

Tables

Table 1: Comparison of Shock and Kill vs. Block and Lock Strategies

Parameter

Shock and Kill

Block and Lock

Primary Mechanism

Reactivation of latent virus

Transcriptional silencing

Viral Production

Increased during treatment

Suppressed or eliminated

Immune Response Required

Yes, to clear reactivated cells

No

Risk of Immune Activation

High

Low

ART Requirement

Continued during reactivation

Potentially discontinued

Main Challenges

Incomplete reactivation, immune escape

Durability of silencing, off-target effects

Clinical Status

Multiple trials, limited success

Preclinical development

Reservoir Elimination

Attempted

Not attempted (functional cure)

Table 2: Major Block and Lock Therapeutic Candidates

Agent

Mechanism of Action

Primary Target

Development Stage

Key Findings

Didehydro-Cortistatin A (dCA)

Tat inhibition, epigenetic silencing

Tat-TAR-P-TEFb

Preclinical

Sustained viral suppression in humanized mice; durable latency in ex vivo studies

Tat Inhibitors (general)

Disruption of viral transcription

Tat protein or TAR RNA

Preclinical

Variable efficacy; concerns about P-TEFb off-target effects

LEDGINs (LED209)

Altered integration site selection

LEDGF/p75-IN interaction

Early preclinical

Promotes integration into inactive chromatin

HDAC Activators

Enhanced histone deacetylation

HIV LTR chromatin

Experimental

Risk of widespread gene expression changes

dCas9-Epigenetic Editors

Targeted epigenetic modification

HIV LTR

Proof-of-concept

Potential for precise, locus-specific silencing

Table 3: Key Molecular Mechanisms Maintaining HIV Latency

Mechanism

Molecular Components

Effect on HIV LTR

Therapeutic Target

Transcriptional Insufficiency

Absence/low Tat levels; inactive NF-κB, NFAT

Low RNA Pol II processivity

Tat inhibitors

Chromatin Remodeling

Restrictive histone modifications (H3K9me3, H3K27me3)

Heterochromatin formation

Histone modifiers

Histone Deacetylation

HDACs recruited to LTR

Transcriptional repression

HDAC modulators

DNA Methylation

DNMTs methylate CpG islands

Stable transcriptional silencing

DNA methylation editors

Integration Site

Integration into heterochromatin regions

Low basal transcription

Integration site selection

Cellular Quiescence

Resting T cell phenotype

Inactive cellular transcription factors

N/A (cellular state)

Table 4: Clinical Challenges for Block and Lock Strategy Implementation

Challenge Category

Specific Issues

Potential Solutions

Measurement/Biomarkers

Lack of markers for latency depth; QVOA limitations

Development of novel assays; single-cell technologies

Reservoir Heterogeneity

Variable integration sites; cellular diversity

Combination approaches; personalized medicine

Anatomical Barriers

CNS, lymphoid tissue reservoirs

Advanced delivery systems; long-acting formulations

Safety/Toxicity

Off-target effects on cellular gene expression

Target specificity; careful dose optimization

Durability

Uncertainty of long-term silencing stability

Extended follow-up studies; escape monitoring

Clinical Endpoints

Defining functional cure criteria

Consensus building; analytical treatment interruption studies

Regulatory Approval

Novel therapeutic paradigm

Engagement with regulatory agencies; adaptive trial designs

CONCLUSION

The Block and Lock strategy represents a promising and innovative approach to achieving a functional cure for HIV infection. By promoting deep and durable transcriptional silencing of latent proviruses, this approach offers potential advantages over Shock and Kill strategies, including avoidance of viral reactivation-associated risks and the possibility of safe ART discontinuation. Preclinical studies, particularly with dCA and Tat inhibitors, have demonstrated proof-of-concept efficacy in multiple experimental systems. However, significant challenges remain before Block and Lock can be translated to clinical practice. These include incomplete understanding of latency mechanisms, reservoir heterogeneity, potential for viral escape, off-target effects, and the need for appropriate biomarkers and clinical endpoints. Future research should focus on combination approaches that integrate Block and Lock with immunotherapies, development of novel delivery systems, and exploration of gene editing technologies. As the HIV cure research field continues to evolve, Block and Lock strategies will likely play an important role in the therapeutic arsenal. The ultimate goal—a safe, effective, and durable cure accessible to all people living with HIV—requires sustained scientific innovation, collaborative research efforts, and commitment to addressing the biological, clinical, and societal complexities of HIV persistence.

 

REFERENCE

  1. Joint United Nations Programme on HIV/AIDS (UNAIDS). (2024). Global HIV statistics: 2024 fact sheet. UNAIDS Reports, 1-8.
  2. Deeks, S. G., Archin, N., Cannon, P., Collins, S., Jones, R. B., de Jong, M. A. W. P., Lambotte, O., Lamplough, R., Ndung'u, T., Sugarman, J., Tiemessen, C. T., Vandekerckhove, L., Lewin, S. R., & International AIDS Society (IAS) Working Group. (2023). Research priorities for an HIV cure: International AIDS Society Global Scientific Strategy 2023. Nature Medicine, 29(11), 2738-2753.
  3. Kulpa, D. A., & Chomont, N. (2023). HIV persistence in the setting of antiretroviral therapy: When, where, and how does HIV hide? Journal of Virus Eradication, 9(2), 100317.
  4. Einkauf, K. B., Osborn, M. R., Gao, C., Sun, W., Sun, X., Lian, X., Parsons, E. M., Gladkov, G. T., Seiger, K. W., Blackmer, J. E., Rigau, D. N., Huang, S. H., Hoh, R., Somsouk, M., Deeks, S. G., Kuritzkes, D. R., Cohn, L. B., Yu, X. G., & Lichterfeld, M. (2023). Parallel analysis of transcription, integration, and sequence of single HIV-1 proviruses. Cell, 186(20), 4272-4283.
  5. Margolis, D. M., Archin, N. M., Cohen, M. S., Eron, J. J., Ferrari, G., Garcia, J. V., Gay, C. L., Goonetilleke, N., Joseph, S. B., Swanstrom, R., Turner, A. M. W., & Wahl, A. (2023). Curing HIV: Seeking to target and clear persistent infection. Cell, 183(5), 1341-1357.
  6. Kim, Y., Anderson, J. L., & Lewin, S. R. (2023). Getting the "kill" into "shock and kill": Strategies to eliminate latent HIV. Cell Host & Microbe, 31(4), 513-534.
  7. Ait-Ammar, A., Kula, A., Darcis, G., Verdoni, R., De Wit, S., Gautier, V., Mallon, P. W. G., Marcello, A., Rohr, O., & Van Lint, C. (2023). Current status of latency reversing agents facing the heterogeneity of HIV-1 cellular and tissue reservoirs. Frontiers in Microbiology, 14, 1268024.
  8. Vansant, G., Bruggemans, A., Janssens, J., & Debyser, Z. (2023). Block-and-lock strategies to cure HIV infection. Viruses, 15(1), 84.
  9. Nixon, C. C., & Mavigner, M. (2023). Mechanisms of HIV reservoir establishment and persistence: Contributions from the tissue microenvironment. Current Opinion in HIV and AIDS, 18(5), 245-252.
  10. Sengupta, S., & Siliciano, R. F. (2023). Targeting the latent reservoir for HIV-1. Immunity, 56(6), 1244-1268.
  11. Achuthan, V., Perreira, J. M., Ahn, J., Brass, A. L., & Engelman, A. N. (2023). Capsid-CPSF6 interaction licenses nuclear HIV-1 integration by facilitating chromatin scanning. Nature Communications, 14(1), 4621.
  12. Mousseau, G., Kessing, C. F., Fromentin, R., Trautmann, L., Chomont, N., & Valente, S. T. (2023). The Tat inhibitor didehydro-cortistatin A prevents HIV-1 reactivation from latency. mBio, 14(1), e02122-22.
  13. Mbonye, U., & Karn, J. (2023). The molecular basis for human immunodeficiency virus latency. Annual Review of Virology, 10(1), 271-291.
  14. Molina-París, C., Lythe, G., Gong, C., Hodgkin, P. D., & King, J. R. (2023). Understanding the stochastic dynamics of HIV-1 latency. Mathematical Biosciences, 364, 109069.
  15. Jiang, G., & Dandekar, S. (2023). Targeting chromatin: A promising strategy for the cure of HIV infection. Trends in Microbiology, 31(8), 798-810.
  16. Casazza, R. L., Lazinski, D. W., & Trono, D. (2023). Targeted epigenome editing: Methods and advances in HIV latency. Current Opinion in Virology, 60, 101323.
  17. Abdel-Mohsen, M., Richman, D., Archin, N. M., Routy, J. P., Bosch, R., Lalezari, J. P., & Howell, B. J. (2023). Recommendations for measuring HIV reservoir size in cure-directed clinical trials. Nature Medicine, 29(5), 1068-1077.
  18. Peluso, M. J., Bacchetti, P., Ritter, K. D., Beg, S., Lai, J., Martin, J. N., Hunt, P. W., Henrich, T. J., Palmer, S., Hoh, R., Sanchez, A., Christenson, J., Campbell, P., McCune, J. M., Deeks, S. G., & Hatano, H. (2023). Differential decay of intact and defective proviral DNA in HIV-1-infected individuals on suppressive antiretroviral therapy. JCI Insight, 8(4), e162907.
  19. Cocchi, F., DeVico, A. L., Lu, W., Popovic, M., Latinovic, O. S., Sajadi, M. M., Redfield, R. R., & Gallo, R. C. (2024). Soluble factors from T cells inhibiting X4 strains of HIV are a mixture of β chemokines and RNases. Proceedings of the National Academy of Sciences, 121(3), e2315865121.
  20. Gavegnano, C., Detorio, M., Montero, C., Bosque, A., Planelles, V., & Schinazi, R. F. (2023). Ruxolitinib and tofacitinib are potent and selective inhibitors of HIV-1 replication and virus reactivation in vitro. Antimicrobial Agents and Chemotherapy, 67(2), e01327-22.

Reference

  1. Joint United Nations Programme on HIV/AIDS (UNAIDS). (2024). Global HIV statistics: 2024 fact sheet. UNAIDS Reports, 1-8.
  2. Deeks, S. G., Archin, N., Cannon, P., Collins, S., Jones, R. B., de Jong, M. A. W. P., Lambotte, O., Lamplough, R., Ndung'u, T., Sugarman, J., Tiemessen, C. T., Vandekerckhove, L., Lewin, S. R., & International AIDS Society (IAS) Working Group. (2023). Research priorities for an HIV cure: International AIDS Society Global Scientific Strategy 2023. Nature Medicine, 29(11), 2738-2753.
  3. Kulpa, D. A., & Chomont, N. (2023). HIV persistence in the setting of antiretroviral therapy: When, where, and how does HIV hide? Journal of Virus Eradication, 9(2), 100317.
  4. Einkauf, K. B., Osborn, M. R., Gao, C., Sun, W., Sun, X., Lian, X., Parsons, E. M., Gladkov, G. T., Seiger, K. W., Blackmer, J. E., Rigau, D. N., Huang, S. H., Hoh, R., Somsouk, M., Deeks, S. G., Kuritzkes, D. R., Cohn, L. B., Yu, X. G., & Lichterfeld, M. (2023). Parallel analysis of transcription, integration, and sequence of single HIV-1 proviruses. Cell, 186(20), 4272-4283.
  5. Margolis, D. M., Archin, N. M., Cohen, M. S., Eron, J. J., Ferrari, G., Garcia, J. V., Gay, C. L., Goonetilleke, N., Joseph, S. B., Swanstrom, R., Turner, A. M. W., & Wahl, A. (2023). Curing HIV: Seeking to target and clear persistent infection. Cell, 183(5), 1341-1357.
  6. Kim, Y., Anderson, J. L., & Lewin, S. R. (2023). Getting the "kill" into "shock and kill": Strategies to eliminate latent HIV. Cell Host & Microbe, 31(4), 513-534.
  7. Ait-Ammar, A., Kula, A., Darcis, G., Verdoni, R., De Wit, S., Gautier, V., Mallon, P. W. G., Marcello, A., Rohr, O., & Van Lint, C. (2023). Current status of latency reversing agents facing the heterogeneity of HIV-1 cellular and tissue reservoirs. Frontiers in Microbiology, 14, 1268024.
  8. Vansant, G., Bruggemans, A., Janssens, J., & Debyser, Z. (2023). Block-and-lock strategies to cure HIV infection. Viruses, 15(1), 84.
  9. Nixon, C. C., & Mavigner, M. (2023). Mechanisms of HIV reservoir establishment and persistence: Contributions from the tissue microenvironment. Current Opinion in HIV and AIDS, 18(5), 245-252.
  10. Sengupta, S., & Siliciano, R. F. (2023). Targeting the latent reservoir for HIV-1. Immunity, 56(6), 1244-1268.
  11. Achuthan, V., Perreira, J. M., Ahn, J., Brass, A. L., & Engelman, A. N. (2023). Capsid-CPSF6 interaction licenses nuclear HIV-1 integration by facilitating chromatin scanning. Nature Communications, 14(1), 4621.
  12. Mousseau, G., Kessing, C. F., Fromentin, R., Trautmann, L., Chomont, N., & Valente, S. T. (2023). The Tat inhibitor didehydro-cortistatin A prevents HIV-1 reactivation from latency. mBio, 14(1), e02122-22.
  13. Mbonye, U., & Karn, J. (2023). The molecular basis for human immunodeficiency virus latency. Annual Review of Virology, 10(1), 271-291.
  14. Molina-París, C., Lythe, G., Gong, C., Hodgkin, P. D., & King, J. R. (2023). Understanding the stochastic dynamics of HIV-1 latency. Mathematical Biosciences, 364, 109069.
  15. Jiang, G., & Dandekar, S. (2023). Targeting chromatin: A promising strategy for the cure of HIV infection. Trends in Microbiology, 31(8), 798-810.
  16. Casazza, R. L., Lazinski, D. W., & Trono, D. (2023). Targeted epigenome editing: Methods and advances in HIV latency. Current Opinion in Virology, 60, 101323.
  17. Abdel-Mohsen, M., Richman, D., Archin, N. M., Routy, J. P., Bosch, R., Lalezari, J. P., & Howell, B. J. (2023). Recommendations for measuring HIV reservoir size in cure-directed clinical trials. Nature Medicine, 29(5), 1068-1077.
  18. Peluso, M. J., Bacchetti, P., Ritter, K. D., Beg, S., Lai, J., Martin, J. N., Hunt, P. W., Henrich, T. J., Palmer, S., Hoh, R., Sanchez, A., Christenson, J., Campbell, P., McCune, J. M., Deeks, S. G., & Hatano, H. (2023). Differential decay of intact and defective proviral DNA in HIV-1-infected individuals on suppressive antiretroviral therapy. JCI Insight, 8(4), e162907.
  19. Cocchi, F., DeVico, A. L., Lu, W., Popovic, M., Latinovic, O. S., Sajadi, M. M., Redfield, R. R., & Gallo, R. C. (2024). Soluble factors from T cells inhibiting X4 strains of HIV are a mixture of β chemokines and RNases. Proceedings of the National Academy of Sciences, 121(3), e2315865121.
  20. Gavegnano, C., Detorio, M., Montero, C., Bosque, A., Planelles, V., & Schinazi, R. F. (2023). Ruxolitinib and tofacitinib are potent and selective inhibitors of HIV-1 replication and virus reactivation in vitro. Antimicrobial Agents and Chemotherapy, 67(2), e01327-22.

Photo
Arnab Roy
Corresponding author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Sandeep Prasad Verma
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Nikita Kumari
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Bikash Vishwakarma
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Kristy Kumari
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Sajid Ansari
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Astha Topno
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Shweta Kumari
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Manshi Kumari
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Purnima Kumari
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Ajay Kumar
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Shruti Kumari
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Ananya Mishra
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Priyanka Singh
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Dibya Sahoo
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Abhijit Kumar
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Manu Sharma
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Komal Singh
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Sneha Singh
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Smita Bharti
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Sneha Sinha
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Jasmine Kumari
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Abhinav Kumar
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Priyanshu Kumar Singh
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Photo
Faiz Alam
Co-author

Sai Nath University, Ranchi, Jharkhand-835219, India

Arnab Roy*, Sandeep Prasad Verma, Nikita Kumari, Bikash Vishwakarma, Kristy Kumari, Sajid Ansari, Astha Topno, Shweta Kumari, Manshi Kumari, Purnima Kumari, Ajay Kumar, Shruti Kumari, Ananya Mishra, Priyanka Singh, Dibya Sahoo, Abhijit Kumar, Manu Sharma, Komal Singh, Sneha Singh, Smita Bharti, Sneha Sinha, Jasmine Kumari, Abhinav Kumar, Priyanshu Kumar Singh, Faiz Alam, Targeting and Reversing HIV Latency Using Novel 'Block and Lock' Strategies: A Comprehensive Review, Int. J. Sci. R. Tech., 2025, 2 (12), 16-26. https://doi.org/10.5281/zenodo.17789094

More related articles
Advancing the Radiopharmaceutical Revolution: Inno...
Alok Kumar, Ankita Singh, Mahesh Kumar Yadav, Aryan Singh, Faizan...
Related Articles
Formulation and Evaluation of Herbal Aloevera Lotion...
Tushar Wadekar , Dr. Jayashri Bairagi, Pandurang Pisal, Harshal Hire, Sahil Pawar, ...
Nodular Sclerosis Classical Hodgkin Lymphoma in a Young Adult: A Comprehensive C...
Saira Susan Thomas, Pallippat Thumban Kheif Mamu, Manjima Sunil, ...
Valorization of Tender Coconut Waste into Sustainable Value-Added Products: Chal...
Selva Kumar C., Chinnarasu K., Kavi Nishanthini S., Kaviya Dharshini D., Kanimozhi B., ...
A Review on the Role of Transdermal Drug Delivery: Microneedles, Patches, and Na...
Yash Tambe, Vaishali Pagar, Harshal Pagar, Vaibhav Thakare, ...
Advancing the Radiopharmaceutical Revolution: Innovation, Challenges, and Expand...
Alok Kumar, Ankita Singh, Mahesh Kumar Yadav, Aryan Singh, Faizan Raza, Md Raja Ansari, Md Affan, Ra...
More related articles
Advancing the Radiopharmaceutical Revolution: Innovation, Challenges, and Expand...
Alok Kumar, Ankita Singh, Mahesh Kumar Yadav, Aryan Singh, Faizan Raza, Md Raja Ansari, Md Affan, Ra...