Loading

Commentary Open Access
Volume 6 | Issue 1

Stem cell–based strategies for HIV-1 remission: Emerging frontiers and translational challenges

  • 1Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
  • 2Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
  • 3Member Cancer Therapeutics Program
+ Affiliations - Affiliations

*Corresponding Author

Giovannino Silvestri, gsilvestri@ihv.umaryland.edu

Received Date: September 22, 2025

Accepted Date: October 21, 2025

Introduction

Antiretroviral therapy (ART) has transformed HIV-1 from a fatal infection into a manageable chronic condition. Yet ART cannot eradicate latent viral reservoirs, necessitating lifelong adherence and leaving more than 38 million people worldwide without a definitive cure [1]. In recent years, stem-cell–based strategies have gained prominence as potential curative approaches, particularly those inspired by the CCR5Δ32 transplantation cases that provided proof-of-concept for long-term viral remission in the absence of ART.

The following sections synthesize lessons from transplantation, advances in gene editing, integration with immunotherapy, and the persistent challenge of HIV reservoirs and microenvironments, concluding with translational and ethical considerations essential for equitable global implementation.

Lessons from Transplantation

The “Berlin Patient” provided the first clinical proof that durable HIV remission is possible [2]. After two allogeneic hematopoietic stem-cell transplants from a CCR5Δ32 homozygous donor, Timothy Ray Brown achieved long-term viral remission without ART. Subsequent cases - the “London Patient” and the “New York Patient”- confirmed this principle, showing that reconstituting an HIV-resistant immune system can enable remission [3,4]. However, allogeneic transplantation carries major risks, including graft-versus-host disease and high treatment-related mortality [5]. Because CCR5Δ32 donors are rare [6], this strategy is impractical for population-scale use. Nevertheless, these landmark cases redirected HIV-cure research toward engineering autologous hematopoietic stem/progenitor cells (HSPCs) capable of resisting infection—forming the foundation for gene-editing approaches.

Gene Editing and Engineered Stem Cells

Genome-editing tools—Zinc-Finger Nucleases (ZFNs), Transcription Activator-Like Effector Nucleases (TALENs), and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR-Cas9) —allow precise disruption of HIV entry co-receptors such as CCR5 (C-C chemokine receptor type 5) in HSPCs. Clinical trials have shown safety and partial engraftment of CCR5-edited cells [7,8]. CRISPR-Cas9 offers superior efficiency and scalability but poses off-target risks that are being mitigated through high-fidelity Cas variants and prime-editing platforms [9]. Targeting CXCR4 is a complementary approach. Although CXCR4 editing can block X4-tropic strains, the receptor’s role in hematopoiesis and stem-cell homing prevents complete knockout [10]. Partial suppression or transient repression via CRISPR interference or small-molecule antagonists reduces viral entry while preserving hematopoietic function [11,12]. Induced pluripotent stem cells (iPSCs) extend gene-based therapies by providing renewable sources of HIV-resistant immune cells. CRISPR-modified iPSCs differentiated into T cells, macrophages, and NK cells have shown robust resistance in pre-clinical models [13]. The last two years have seen major progress: Fang et al. and Alidadi et al. outlined how iPSC-derived CAR-T (Chimeric Antigen Receptor T-cell therapy) and CAR-NK (Chimeric Antigen Receptor-Natural Killer cells) cells can be standardized for large-scale manufacturing and personalized therapy [14,15]. Concepts for universal/hypo-immunogenic donor lines are maturing [16]. Madrid et al. summarized translational and regulatory considerations for allogeneic iPSC-derived immune products [17]. Together these advances support an integrated strategy in which precise gene editing and stem-cell biology converge to create durable, HIV-resistant hematopoietic and immune systems.

Stem-Cell and Gene-Editing Strategies for HIV Remission

Key gene-editing and stem-cell strategies under development for HIV remission are outlined in Table 1.

Table 1. Summarizes the major gene-editing and stem-cell approaches currently under development for HIV remission.

Strategy

Target / Mechanism

Platform

Stage / Status

Key References

CCR5 gene knockout in HSPCs

Blocks HIV entry via CCR5 loss

ZFN / CRISPR

Phase I clinical (ongoing)

[7–9]

CXCR4 modulation

Partial suppression of CXCR4 expression

CRISPRi / antagonists / epigenetic repressors

Pre-clinical (2024)

[10–12]

iPSC-derived immune cells

Differentiation into HIV-resistant T, NK, or macrophage lineages

CRISPR-iPSC platforms

Pre-clinical (2024– 2025)

[13–17]

HSPC-derived CAR-T cells

Persistent anti-HIV adaptive immunity

Lentiviral CAR integration into HSPCs

NHP model and Phase I

[18,19]

HSPC-derived CAR-NK cells

Innate antiviral response with minimal GvHD risk

Retroviral CAR transduction of CD34+ cells

Pre-clinical and pilot trial (2025)

[20]

EBT-101 therapy

CRISPR-based excision of integrated HIV provirus

AAV-CRISPR (Excision Bio Therapeutics)

Phase I/II (2024)

[21]

Immunotherapy Integration

Stem-cell–based immunity can be enhanced by CAR-T and CAR-NK engineering. CAR-T cells targeting HIV Env or Gag have demonstrated safety but limited persistence [18]. Recent developments show that adoptively transferred mature CAR-T cells often exhibit exhaustion and fail to provide durable antiviral surveillance. Engineering hematopoietic stem/progenitor cells (HSPCs) to express CAR constructs can generate a renewable source of CAR-expressing effector cells that continually differentiate in vivo [19]. CAR-NK therapies provide complementary advantages, including MHC-independent recognition, lower graft-versus-host risk, and potent innate cytotoxicity [20]. Pilot trials using HSPC-derived CAR-NK cells are evaluating persistence, safety, and the potential to clear viral reservoirs in ART-suppressed individuals. Combining CAR-based immunity with latency-reversing agents (LRAs) could synergistically expose and eradicate latent HIV. This “shock-and-kill” paradigm, paired with gene-edited stem-cell–derived immunity, may convert transient remission into long-term functional cure [22].

Reservoirs and Microenvironmental Challenges

Viral reservoirs persist in lymphoid tissues, the gut, and the central nervous system [22]. Within these niches, stromal interactions, cytokine gradients, and hypoxic conditions sustain latency and blunt immune clearance. Hypoxia modulates HIF-1α signaling, dampening antiviral immunity and supporting viral persistence [23]. Analogous to oncology, the HIV reservoir microenvironment can be therapeutically targeted. Agents modulating oxygen tension or cellular metabolism and 3-D organoid or tissue-chip systems now enable ex vivo testing of latency reversal and immune-clearance strategies [24]. These models provide physiologically relevant platforms to validate gene-edited immune interventions and guide translation to patients.

Ethical and Regulatory Considerations

Gene editing for HIV cure raises ethical and safety concerns. Off-target mutagenesis, insertional oncogenesis, and long-term clonal drift remain the primary risks [9]. Current mitigation measures include high-fidelity Cas variants, base editing, and transient delivery vectors to limit nuclease exposure. The “CRISPR-baby” incident underscored the line between somatic and germline editing [25,26]. WHO and NIH bioethics frameworks [27,28] emphasize transparent governance, layered review, and community engagement. Equity must also remain central to implementation, as underscored by the UNAIDS 2024 report on fairness in the HIV response [29]. Equity must also remain central. Without deliberate access planning, advanced gene and cell therapies could widen global health disparities. Ensuring affordability, scalable manufacturing, and trial inclusion in high-burden regions will be critical to achieve a just HIV-cure landscape [29].

Future Directions

Future HIV-cure paradigms will combine genetic resistance, immune reprogramming, and microenvironment modulation. We propose integrating (i) CCR5-edited autologous HSPCs, (ii) durable HSPC-derived CAR-T/NK cells, and (iii) LRAs to flush reservoirs, alongside oxygen- and metabolism-targeted adjuncts [18–23]. Developing universal hypo-immunogenic iPSC lines requires precise HLA editing (B2M/CIITA knockout with HLA-E/G retention) and strategies to avoid NK-cell “missing-self” rejection. Parallel efforts are refining differentiation protocols and engraftment durability [14–17]. Lessons from oncology—microenvironment targeting, combination immunotherapies, and adaptive trial design—can accelerate HIV translation. Precision-medicine frameworks incorporating viral tropism, HLA/KIR genotype, and reservoir profiling will personalize therapy and improve success rates.

Conclusion

The CCR5Δ32 transplantation cases proved that HIV cure is achievable. Advances in gene editing, stem-cell engineering, and immunotherapy now transform this insight into actionable strategies. The field’s success will depend on simultaneous progress in safety, microenvironment targeting, and equitable access. Stem-cell–based therapies are no longer experimental outliers—they form a cornerstone of the HIV-cure agenda aiming to make durable remission broadly attainable.

Conflicts of Interest

The authors declare no conflicts of interest.

Funding and Author Contributions

Supported by the National Cancer Institute—Cancer Center Support Grant (CCSG) P30CA134274 and the UMGCCC American Cancer Society Institutional Research Grant IRG-24-1290479-19 (GS). GS conceived, drafted the manuscript and reviewed criticisms; AC performed literature review and table construction.

References

1. Deeks SG, Archin N, Cannon P, Collins S, Jones RB, de Jong MAWP, et al. Research priorities for an HIV cure: International AIDS Society Global Scientific Strategy 2021. Nat Med. 2021 Dec;27(12):2085–98.

2. Hütter G, Nowak D, Mossner M, Ganepola S, Müssig A, Allers K, et al. Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N Engl J Med. 2009 Feb 12;360(7):692–8.

3. Gupta RK, Abdul-Jawad S, McCoy LE, Mok HP, Peppa D, Salgado M, et al. HIV-1 remission following CCR5Δ32/Δ32 haematopoietic stem-cell transplantation. Nature. 2019 Apr;568(7751):244–8.

4. Hsu J, Van Besien K, Glesby MJ, Pahwa S, Coletti A, Warshaw MG, et al. HIV-1 remission and possible cure in a woman after haplo-cord blood transplant. Cell. 2023 Mar 16;186(6):1115–26.e8.

5. Appelbaum FR. Hematopoietic-cell transplantation at 50. N Engl J Med. 2007 Oct 11;357(15):1472–5.

6. Samson M, Libert F, Doranz BJ, Rucker J, Liesnard C, Farber CM, et al. Resistance to HIV-1 infection in caucasian individuals bearing mutant alleles of the CCR-5 chemokine receptor gene. Nature. 1996 Aug 22;382(6593):722–5.

7. Tebas P, Stein D, Tang WW, Frank I, Wang SQ, Lee G, et al. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N Engl J Med. 2014 Mar 6;370(10):901–10.

8. Xu L, Wang J, Liu Y, Xie L, Su B, Mou D, et al. CRISPR-Edited Stem Cells in a Patient with HIV and Acute Lymphocytic Leukemia. N Engl J Med. 2019 Sep 26;381(13):1240–7.

9. Porteus MH. A New Class of Medicines through DNA Editing. N Engl J Med. 2019 Mar 7;380(10):947–59.

10. Lee C. CRISPR/Cas9-Based Antiviral Strategy: Current Status and the Potential Challenge. Molecules. 2019 Apr 5;24(7):1349.

11. Saotome K, McGoldrick LL, Ho JH, Ramlall TF, Shah S, Moore MJ, et al. Structural insights into CXCR4 modulation and oligomerization. Nat Struct Mol Biol. 2025 Feb;32(2):315–25.

12. Prokopovich AK, Litvinova IS, Zubkova AE, Yudkin DV. CXCR4 Is a Potential Target for Anti-HIV Gene Therapy. Int J Mol Sci. 2024 Jan 18;25(2):1187.

13. Allen AG, Chung CH, Atkins A, Dampier W, Khalili K, Nonnemacher MR, et al. Gene Editing of HIV-1 Co-receptors to Prevent and/or Cure Virus Infection. Front Microbiol. 2018 Dec 17;9:2940.

14. Fang Y, Chen Y, Li YR. Engineering the next generation of allogeneic CAR cells: iPSCs as a scalable and editable platform. Stem Cell Reports. 2025 Jul 8;20(7):102515.

15. Alidadi M, Barzgar H, Zaman M, Paevskaya OA, Metanat Y, Khodabandehloo E, et al. Combining the induced pluripotent stem cell (iPSC) technology with chimeric antigen receptor (CAR)-based immunotherapy: recent advances, challenges, and future prospects. Front Cell Dev Biol. 2024 Nov 18;12:1491282.

16. Simpson A, Hewitt AW, Fairfax KA. Universal cell donor lines: A review of the current research. Stem Cell Reports. 2023 Nov 14;18(11):2038–46.

17. Madrid M, Lakshmipathy U, Zhang X, Bharti K, Wall DM, Sato Y, et al. Considerations for the development of iPSC-derived cell therapies: a review of key challenges by the JSRM-ISCT iPSC Committee. Cytotherapy. 2024 Nov;26(11):1382–99.

18. Riley JL, Montaner LJ. Cell-Mediated Immunity to Target the Persistent Human Immunodeficiency Virus Reservoir. J Infect Dis. 2017 Mar 15;215(suppl_3):S160–71.

19. Zhen A, Peterson CW, Carrillo MA, Reddy SS, Youn CS, Lam BB, et al. Long-term persistence and function of hematopoietic stem cell-derived chimeric antigen receptor T cells in a nonhuman primate model of HIV/AIDS. PLoS Pathog. 2017 Dec 28;13(12):e1006753.

20. Liu E, Marin D, Banerjee P, Macapinlac HA, Thompson P, Basar R, et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N Engl J Med. 2020 Feb 6;382(6):545–53.

21. Excision BioTherapeutics. Excision BioTherapeutics announces data from the Phase 1/2 trial of EBT‑101 in HIV and in vivo efficacy data in herpes virus and hepatitis B [press release 13 May 2024]. Available from: https://www.excision.bio/news/press-releases/detail/43/excision-iotherapeutics-announces-data-from-the-phase-12.

22. Cohn LB, Chomont N, Deeks SG. The Biology of the HIV-1 Latent Reservoir and Implications for Cure Strategies. Cell Host Microbe. 2020 Apr 8;27(4):519–30.

23. Zhuang X, Pedroza-Pacheco I, Nawroth I, Kliszczak AE, Magri A, Paes W, et al. Hypoxic microenvironment shapes HIV-1 replication and latency. Commun Biol. 2020 Jul 14;3(1):376.

24. 3D Human Tissue Models for HIV Working Group. 3D Human Tissue Models for HIV Working Group. 3D human tissue models and microphysiological systems for HIV and related comorbidities. Trends Biotechnol. 2024 May;42(5):526–43.

25. Psomas CK, Waters L, Barber T, Fidler S, Macartney M, Alagaratnam J, et al. Highlights of the 10th International AIDS Society (IAS) Conference on HIV Science, 21-25 July 2019, Mexico City, Mexico. J Virus Erad. 2019 Nov 4;5(4):245–52.

26. Greely HT. Human Germline Genome Editing: An Assessment. CRISPR J. 2019 Oct;2(5):253–65.

27. World Health Organization (WHO). Human genome editing: recommendations. Report of the WHO Expert Advisory Committee on Developing Global Standards for Governance and Oversight of Human Genome Editing: Geneva. World Health Organization; 2021. Available at: https://www.who.int/publications/i/item/9789240030381.

28. National Institutes of Health, Office of Science Policy (OSP). Policy and governance of gene-editing research in the United States: Ethical and oversight considerations. Washington, DC: NIH; 2023. Available at: https://osp.od.nih.gov/.

29. Joint United Nations Programme on HIV/AIDS (UNAIDS). Equity in the HIV Response: Assessing progress and charting a way forward. Geneva: UNAIDS; 2024. Available at: https://www.unaids.org/sites/default/files/media_asset/equity-in-the-hiv response_en.pdf.

Author Information X