type
status
date
slug
summary
tags
category
icon
password
Introduction: The Promise of a Paradigm Shift
The advent of chimeric antigen receptor (CAR) T-cell therapy has undeniably revolutionized the treatment of certain cancers and autoimmune diseases. However, the prevailing model—adoptive cell therapy—requires a complex, bespoke, and costly process. A patient’s T cells must be extracted, genetically engineered ex vivo in specialized manufacturing facilities, expanded, and then reinfused. This logistical marathon translates into treatment delays, costs exceeding hundreds of thousands of dollars, and limited global accessibility. In response, a transformative alternative has emerged: in vivo CAR-T cell engineering. This approach aims to democratize cell therapy by shifting the entire manufacturing process inside the patient's body. Instead of a cellular product, a standardized vial of gene delivery vectors is administered, programming the patient’s own T cells on-site. While the ultimate therapeutic mechanism remains identical, the production paradigm is radically simplified. The thesis of this review is that in vivo CAR-T engineering holds revolutionary potential to increase accessibility, but its success is contingent on overcoming the interconnected challenges of delivery precision, controlling therapeutic durability, and successful clinical translation, as evidenced by the distinct profiles of emerging platform technologies and their early forays into human trials.
The Core Technological Platforms: Mechanisms and Fundamental Trade-Offs
The vision of in vivo engineering is pursued through three primary technological platforms, each presenting a unique set of advantages and compromises centered on efficiency, persistence, and safety.
Viral vectors, primarily lentiviruses, represent a direct translation of ex vivo methodology to an in vivo setting. Engineered for improved T-cell targeting and safety, these vectors offer the significant advantage of stable genomic integration. This leads to long-term CAR expression—potentially persisting for months or years—which is highly desirable for sustaining durable remissions in oncology. However, this benefit is counterbalanced by the persistent risk of insertional mutagenesis, where random integration could disrupt critical genes and potentially lead to malignancy. Furthermore, achieving efficient and specific transduction of T cells in the complex in vivo environment requires sophisticated viral retargeting to avoid off-target effects and immune neutralization.
In contrast, lipid nanoparticle (LNP) platforms, hallmarked by their success in mRNA vaccines, offer a safer and more scalable alternative. LNPs can be engineered to deliver mRNA encoding the CAR directly to T cells. As the mRNA operates in the cytoplasm without genomic integration, the risk of insertional mutagenesis is eliminated, and CAR expression is inherently transient, typically peaking within 24-48 hours and fading over days to a week. This transient nature is a double-edged sword: it provides a built-in safety switch, making the platform attractive for autoimmune conditions where a transient immune reset may be sufficient, but it may be inadequate for eradicating aggressive cancers without repeated dosing. Consequently, a major research focus is extending this window using technologies like self-amplifying mRNA or circular RNA (circRNA).
A third, more physical strategy employs implantable scaffolds or hydrogels. These biomaterial devices are loaded with viral vectors and implanted at or near the disease site. Their function is to recruit circulating T cells into a confined space, creating a local "factory" with a high concentration of vectors, thereby aiming for high local transduction efficiency while minimizing systemic exposure and off-target effects. The duration of CAR expression here is not inherent to the platform but depends on the vector used (lentivirus for persistence, mRNA for transience). This approach elegantly sidesteps many systemic delivery hurdles but introduces the complexity of a medical implant.
The Paramount Challenge: Achieving Cellular Precision
A non-negotiable hurdle for all in vivo platforms is the imperative of cellular precision—ensuring the CAR genetic payload is delivered exclusively to the intended T cells. Off-target delivery could lead to reduced efficacy, unpredictable toxicities, or severe safety events. The strategies to achieve this precision are as diverse as the platforms themselves, relying on molecular recognition and spatial control.
For viral vectors, precision is engineered through molecular retargeting. Scientists modify the viral envelope proteins, such as the VSV-G protein on lentiviruses, by replacing their natural binding domains with antibody fragments or ligands specific to T-cell markers like CD3, CD4, or CD8. Some advanced designs go beyond simple targeting by fusing T-cell costimulatory molecules (e.g., CD80) to the viral coat, providing an activation signal upon binding to enhance transduction and potency. An additional layer of safety is added by using T-cell-specific promoters to ensure CAR expression is active only even if the virus enters another cell type.
Nanoparticle platforms achieve targeting through modular design. The surface of LNPs can be decorated with antibodies or smaller ligands that act as homing devices. For instance, CD5-targeted LNPs have successfully generated CAR-T cells in murine models of fibrosis, and a clinical candidate for lupus employs CD8-targeted LNPs. Beyond conjugated antibodies, researchers are exploring novel lipid chemistries, such as cardiolipin-mimetics, that inherently favor delivery to immune cells.
Implantable scaffolds adopt a strategy of spatial control. By creating a localized depot of vectors, they avoid the distribution problem entirely. These devices either recruit endogenous T cells or are seeded with briefly isolated cells, facilitating conversion in a privileged anatomical site. This method maximizes local concentration at the target tissue (e.g., a tumor) while theoretically reducing systemic risks.
Despite these sophisticated approaches, perfect precision remains elusive. Challenges include eliminating off-target delivery to organs like the liver (a common nanoparticle sink), achieving specificity for the most therapeutically potent T-cell subsets, and managing the immunogenicity of repeated dosing with targeted delivery vehicles. The field must continuously balance the trade-off between exquisitely precise targeting and achieving a therapeutically sufficient level of transduction efficiency.
From Bench to Bedside: The Emerging Clinical and Commercial Landscape
The theoretical promise of in vivo CAR-T is now undergoing the critical test of human clinical trials. Early data serve as both proof-of-concept and a roadmap, highlighting the different therapeutic niches each platform may occupy.
Clinically, functional efficacy has been demonstrated across platforms. In autoimmunity, trials using CD8-targeted LNP-mRNA (e.g., candidate HN2301 for lupus) have shown rapid detection of edited CAR-T cells in patient blood within hours, followed by profound, therapeutic B-cell depletion. This validates that transient, mRNA-based CAR expression can achieve a potent, reset-like effect. In oncology, a lentiviral vector candidate (ESO-T01) has reported clinical responses, including remission, in patients with multiple myeloma, proving that stable, in vivo-generated CAR-T cells can expand and mediate anti-tumor activity. These early successes span both major disease areas and platform types.
Commercially, the landscape has witnessed explosive growth since 2021, with over $2 billion in funding and a pipeline that has surged from a handful of assets to over 70. This momentum underscores the vast perceived potential. Companies are staking claims on different technological solutions. For example, Umoja Biopharma’s VivoVec platform employs a lentiviral system designed for single-dose, long-lasting responses, with its INVICTA-1 trial among the first in vivo CAR-T studies in the U.S. Conversely, Myeloid Therapeutics is advancing intravenous mRNA CAR programs (MT-302, MT-303) delivered via targeted LNPs, focusing on the safety and repeat-dosing profile of transient expression. This commercial diversification mirrors the technological trade-offs, as different strategies are pursued to solve the core challenges of precision and durability.
While in vivo CAR-T therapies constitute a small fraction of the overall CAR-T clinical trial landscape dominated by ex vivo approaches, their entry into human testing marks a pivotal transition. It moves the conversation from "is it possible?" to "how and for which diseases will it be most effective?"
Conclusion: Synthesis and Future Directions
In vivo CAR-T cell engineering represents a bold re-imagining of cell therapy, aiming to compress a complex, weeks-long logistical chain into a single, off-the-shelf injection. As this review has outlined, this paradigm shift does not simplify the underlying biology but rather transfers the technical challenges from the cleanroom to the patient's body. The central hurdles become achieving precise cellular delivery and controlling the duration and activity of the resulting CAR-T cells.
The choice of platform—viral vector or nanoparticle—fundamentally dictates the therapeutic profile, creating a core trade-off between persistence and safety. Lentiviral strategies offer the prospect of durable, "one-and-done" remissions but carry long-term genomic safety concerns. LNP-mRNA strategies offer a safer, tunable, and potentially repeatable intervention but must prove sufficient potency, especially in oncology. Scaffold-based approaches offer a clever hybrid, using spatial control to enhance the efficacy of either vector type.
The future trajectory of the field will involve not only refining these platforms—through next-generation technologies like saRNA or smarter gene circuits—but also strategically matching them to clinical contexts. Transient modulation may be ideal for autoimmune reset or solid tumor microenvironments requiring precise control, while persistent CAR expression may remain the goal for durable eradication of liquid tumors. With continued innovation focused on targeting precision and temporal control, in vivo CAR-T therapy is poised to evolve from a promising concept into a more accessible and versatile pillar of the immunotherapeutic arsenal, ultimately striving to deliver on the original promise of cell therapy for a much broader population of patients.
References
- Chen Y, Xin Q, Qiu J, Zhu M, Li Z, Qiu J, Tu J, Li R. In vivo CAR-T cell engineering: concept, research progress, potential challenges and enhancement strategies. Exp Hematol Oncol. 2025 Nov 18;14(1):133. doi: 10.1186/s40164-025-00725-5. PMID: 41250215; PMCID: PMC12625078.
- Huang Y, Cao R, Wang S, Chen X, Ping Y, Zhang Y. In vivo CAR-T cell therapy: New breakthroughs for cell-based tumor immunotherapy. Hum Vaccin Immunother. 2025 Dec;21(1):2558403. doi: 10.1080/21645515.2025.2558403. Epub 2025 Sep 11. PMID: 40932272; PMCID: PMC12427527.
- Author:Pao
- URL:https://paoresearch.uk//article/2e17f632-00f0-80b5-93d8-f00f32538b1f
- Copyright:All articles in this blog, except for special statements, adopt BY-NC-SA agreement. Please indicate the source!






