2025.08.16
Chimeric Antigen Receptor T cell (CAR-T) therapy, a groundbreaking immunotherapy, has demonstrated remarkable efficacy in treating hematological cancers such as B-cell lymphoma[1]. While traditional CAR-T therapy relies on an "ex-vivo engineering" model, the emerging "in-vivo engineering" (in-vivo CAR-T) technology aims to generate CAR-T cells directly within the patient’s body, addressing critical bottlenecks of the ex-vivo approach. Based on relevant literature, this article focuses on the technical principles of in-vivo CAR-T, detailing its differences from traditional ex-vivo CAR-T, advantages, challenges, and future prospects, providing a comprehensive analysis of advancements and hurdles in this emerging field.
I. Technical Principles: Mechanisms of In-Vivo CAR-T
The core of in-vivo CAR-T technology lies in directly modifying T cells within the patient to express chimeric antigen receptors (CARs), enabling targeted cancer cell elimination. This is achieved via engineered vector delivery, eliminating the need for ex-vivo isolation and modification steps[2][3]. Its mechanisms involve the following key steps:
Vector Delivery and T Cell Targeting: Non-viral or viral-based vectors are used to deliver CAR genes directly into the patient. These vectors include lipid-based nanoparticles (LNPs)[4], fusogenic nanovesicles (FuNVs)[5], engineered exosomes[6][7], or injectable viral vectors (e.g., lentiviruses)[8][9]. Designed to specifically target circulating T cells, these vectors deliver CAR-encoding genetic material into T cells via membrane fusion or endocytosis[5][10]. For example, lipid nanoparticles can stably deliver CAR genes in vivo, inducing CAR expression in T cells within days to form functional CAR-T cells[10].
Genetic Engineering and CAR Expression: The delivered genetic material (e.g., mRNA or DNA) is expressed as CAR proteins in T cells. CARs typically consist of an antigen-binding domain (e.g., scFv targeting CD19 or BCMA), a transmembrane domain, and intracellular signaling domains[11][1]. In vivo, this process relies on vector optimization to maximize transduction efficiency. For instance, non-viral vectors reduce virus-related safety risks while achieving high transduction efficiency[12][4]. Studies have shown that specific vector delivery systems can induce sustained CAR expression in vivo, enhancing T cell anti-tumor activity[9][5].
Immune Activation and Tumor Killing: In-vivo generated CAR-T cells recognize cancer cell surface antigens, activate T cell signaling pathways, and release cytokines (e.g., IFNγ, TNF) and granzymes to directly eliminate tumor cells[13][10]. Additionally, the system can be designed as a "universal platform" (e.g., combined with immunomodulators like selinexor) to optimize T cell function and reduce microenvironmental suppression[13][14].
The principle of in-vivo CAR-T is essentially "in-situ engineering," simplifying processes to enable real-time immune reprogramming[3][4]. Current technologies are primarily validated in mouse models, with success dependent on vector design ensuring targeting accuracy and gene expression stability[8][9]. This approach avoids cumbersome ex-vivo manufacturing steps, generating CAR-T cells directly within the biological environment[2][8].
II. Differences from Traditional Ex-Vivo CAR-T
Ex-vivo CAR-T, the current clinical mainstream, involves collecting T cells from patients or donors, engineering them via viral transduction or electroporation in laboratories, expanding them, and infusing them back into patients[1][1][15]. The in-vivo model differs significantly in the following aspects:
Manufacturing Process:
1. Ex-vivo CAR-T: Requires multiple ex-vivo steps: leukapheresis, viral transduction (e.g., lentivirus), ex-vivo activation and expansion (2–3 weeks), and final infusion[16][8][15]. This process demands strict laboratory control and carries risks of T cell exhaustion and functional loss[16][17].
2. In-vivo CAR-T: Eliminates all ex-vivo steps; patients receive direct vector injection, with in-vivo systems automatically completing T cell targeting, genetic engineering, and CAR expression[3][9][5]. This drastically simplifies the process, reducing treatment time to days[10].
Cost and Accessibility:
1. Ex-vivo CAR-T: High manufacturing costs (hundreds of thousands of dollars) and reliance on specialized centers (e.g., cell therapy units) limit global accessibility[18][8][19]. For example, FDA-approved CAR-T therapies face manufacturing bottlenecks, restricting use to specific patient groups[18][1].
2. In-vivo CAR-T: Reduces production costs due to simpler vector manufacturing compared to cell culture; can be administered in general medical facilities without specialized centers[18][20]. Studies indicate potential for widespread adoption, particularly in resource-limited regions[2][15].
Efficacy and Toxicity:
1. Ex-vivo CAR-T: Ex-vivo activation often leads to T cell exhaustion, characterized by increased surface markers (e.g., PD-1, LAG-3), impairing persistence and function[13][16][17]. Common side effects include cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), resulting from excessive immune activation by ex-vivo engineered cells[8][8].
2. In-vivo CAR-T: In-vivo generated cells may reduce T cell exhaustion (e.g., via vector optimization to enhance mitochondrial respiratory function)[21][10] and lower systemic toxicity (e.g., CRS); the in-vivo environment naturally regulates immune responses, minimizing hyperactivation[8][8]. For example, animal models show in-vivo produced CAR-T cells achieve anti-tumor efficacy comparable to traditional ex-vivo CAR-T[10].
Technical Dependence and Limitations:
1. Ex-vivo CAR-T: Primarily effective for hematological cancers; limited efficacy in solid tumors due to microenvironmental suppression and physical barriers[17][22]. Its engineering methods are rigid, lacking flexibility[23][17].
2. In-vivo CAR-T: Flexible design (e.g., combining nanoparticles or exosomes) facilitates solid tumor targeting by overcoming tumor microenvironment barriers[12][4][24]. However, vector optimization is required to address uncontrollable in-vivo factors[9][3].
In summary, the core distinction of in-vivo CAR-T lies in "decentralized manufacturing" and "in-situ engineering," emphasizing convenience and biocompatibility[3][20].
III. Advantages of In-Vivo CAR-T
Based on research, in-vivo CAR-T offers significant advantages by addressing ex-vivo limitations:
Simplified Preparation and Reduced Costs: Eliminating expensive ex-vivo steps reduces production costs (e.g., lower equipment, personnel, and culture medium expenses); vectors (e.g., nanoparticles) enable mass production, improving scalability[18][20]. Studies confirm its ability to "bypass CAR-T manufacturing bottlenecks," making therapy more economical[18][8]. For example, lipid nanoparticle systems achieve efficacy comparable to traditional CAR-T in animal models at significantly lower costs[10].
Shorter Timeline and Improved Accessibility: Reducing preparation time from weeks to days enables timely treatment, particularly benefiting patients with progressive cancers[15][5][18][2]. Treatment can be administered in outpatient settings, reducing hospitalization needs and improving global accessibility[18][8].
Reduced Toxicity: Ex-vivo engineering often causes T cell exhaustion and hyperactivation, leading to CRS and other side effects. In contrast, in-vivo generated cells integrate more naturally into the immune system, lowering risks of systemic toxicity (e.g., CRS and neurotoxicity)[8][8][10]. For example, optimized vectors (e.g., sustained-release delivery) mitigate inflammatory responses in animal models[21].
Enhanced Functionality and Flexibility: Vector platforms support customizable designs (e.g., integrating immunomodulators) to boost CAR-T anti-tumor activity[4][13]. In-vivo T cells exhibit reduced phenotypic exhaustion (e.g., lower PD-1 expression), maintaining long-term tumor-killing capacity[13][12][21]. For solid tumors, targeted delivery (e.g., bioinstructive scaffolds) overcomes microenvironmental suppression[22][24].
Expanded Application Potential: As a "breakthrough technology," its low cost and simplicity facilitate development of new solid tumor therapies[25][17]. For example, combinations with exosomes or nanoparticles enable application across multiple cancer types[6][7][24].
In summary, the core advantages of in-vivo CAR-T—"simplified manufacturing, low cost, high accessibility, and reduced toxicity"—pave the way for widespread clinical application[20][3].
IV. Challenges of In-Vivo CAR-T
Despite promising prospects, in-vivo CAR-T faces significant technical hurdles:
Precise Targeting and Delivery Efficiency: The complex in-vivo environment requires vectors to specifically target T cells (avoiding other tissues) for efficient transduction. Current vectors (e.g., nanoparticles) suffer from off-target effects and low transduction rates; studies report uneven delivery in animal models[8][9][3]. For example, non-viral vector immunogenicity requires optimization to reduce side effects[12][9].
CAR Expression Stability and Persistence: Compared to ex-vivo engineering, in-vivo generated CAR-T cells may exhibit unstable or transient CAR expression; efficacy is affected by in-vivo immunosuppressive factors (e.g., M2 macrophages in the tumor microenvironment)[25][9][3]. Studies highlight the need for vector design (e.g., RNA-based constructs) to extend gene expression duration[20][10].
Safety and Toxicity Control: Vectors may induce toxicity (e.g., oncogenic risks with viral vectors) or unintended immune responses[8][9]. Optimizing in-vivo dosing to avoid hyperactivation remains challenging[8][3].
Technical Optimization and Lack of Standards: Absence of unified in-vivo engineering protocols results in variable efficacy across platforms (e.g., LNPs vs. viral vectors)[12][4]. Targeted delivery for solid tumors requires overcoming physical barriers and immunosuppression[17][22][24].
Scalability and Clinical Translation: Transitioning from animal models to human trials requires addressing scalability and standardized production[26][3]. Regulatory approval (e.g., FDA) for in-vivo therapies remains pending, indicating gaps in clinical validation[18][8].
These challenges reflect technical immaturity but are expected to be overcome with continued research[12][3].
V. Future Outlook
In-vivo CAR-T is regarded as the next generation of immunotherapy, with prospects rooted in technological evolution and application expansion:
Technological Innovation and Vector Development: Research focuses on optimizing delivery platforms, such as "multifunctional nanoparticles" or "engineered exosomes," to enhance targeting and safety[12][6][4]. For example, lipid-based vectors combined with immunomodulators (e.g., rapamycin) improve CAR-T mitochondrial function and persistence[21][10]. Novel "bioinstructive scaffolds" may enable local delivery to address solid tumor challenges[24][3].
Expanded Applications: Current in-vivo CAR-T focuses on hematological cancer models; future efforts will prioritize solid tumor treatment by overcoming microenvironmental barriers via targeted delivery[25][17][24]. Combination therapies (e.g., with selinexor or Axitinib) may enhance synergistic effects and reduce immunosuppression[13][27].
Accessibility and Industrialization: Simplified production will lower costs, transforming in-vivo CAR-T into a "versatile paradigm" for global patient access[12][20][15]. Biomaterial-based strategies may revolutionize CAR-T manufacturing[24].
Regulatory and Clinical Progress: Following animal model success (e.g., extended survival in leukemia models[10]), human clinical trials will advance, requiring adaptive regulatory frameworks[26][8].
Long-Term Vision: The goal is to transform CAR-T into an "off-the-shelf" therapy, revolutionizing cancer treatment via "bridging technological insights"[12][4]. Ultimately, in-vivo CAR-T may extend beyond oncology to treat autoimmune diseases[26][20].
In conclusion, in-vivo CAR-T represents a shift from "complex customization" to "convenient broad-spectrum therapy," poised to reshape immunotherapy[12][3].
Conclusion
In-vivo CAR-T therapy, centered on direct in-vivo engineering, addresses ex-vivo limitations via vector delivery systems, offering advantages such as low cost, high accessibility, and reduced toxicity. Despite technical challenges (e.g., delivery efficiency and safety), ongoing innovations (e.g., nanoparticle optimization and expanded applications) promise a bright future. In-vivo CAR-T is expected to become a mainstream cancer immunotherapy, advancing treatment democratization. While current progress relies on preclinical models, its "decentralized manufacturing" paradigm demonstrates transformative potential[18][12][20][3].
Reference:
1. Luo, Yan et al. “A new sort of cells for chimeric antigen receptor T-cell therapies-isolating CD14-CD127+ T cells for chimeric antigen receptor T-cell manufacture.” Cytotherapy vol. 27,8 (2025): 980-990. doi:10.1016/j.jcyt.2025.04.068
2. Coradin, Tiziana et al. “Efficient in vivo generation of CAR T cells using a retargeted 4th generation lentiviral vector.” Molecular therapy : the journal of the American Society of Gene Therapy, S1525-0016(25)00542-8. 16 Jul. 2025, doi:10.1016/j.ymthe.2025.07.006
3. Li, Yan-Ruide et al. “In vivo CAR engineering for immunotherapy.” Nature reviews. Immunology, 10.1038/s41577-025-01174-1. 16 May. 2025, doi:10.1038/s41577-025-01174-1
4. Albalawi, Yasmeen A. “Applications of nanoparticles in CAR-T cell therapy: non-viral manufacturing, enhancing in vivo function, and in vivo generation of CAR-T cells.” Medical oncology (Northwood, London, England) vol. 42,9 378. 26 Jul. 2025, doi:10.1007/s12032-025-02928-6
5. Zhao, Gui et al. “In vivo production of CAR-T cells using virus-mimetic fusogenic nanovesicles.” Science bulletin vol. 69,3 (2024): 354-366. doi:10.1016/j.scib.2023.11.055
6. Si, Ke et al. “Engineered exosome-mediated messenger RNA and single-chain variable fragment delivery for human chimeric antigen receptor T-cell engineering.” Cytotherapy vol. 25,6 (2023): 615-624. doi:10.1016/j.jcyt.2023.01.005
7. Zhang, Si-Heng et al. “Focusing on exosomes to overcome the existing bottlenecks of CAR-T cell therapy.” Inflammation and regeneration vol. 44,1 45. 4 Nov. 2024, doi:10.1186/s41232-024-00358-x
8. Pinto, E et al. “From ex vivo to in vivo chimeric antigen T cells manufacturing: new horizons for CAR T-cell based therapy.” Journal of translational medicine vol. 23,1 10. 4 Jan. 2025, doi:10.1186/s12967-024-06052-3
9. Bui, Thuy Anh et al. “Advancements and challenges in developing in vivo CAR T cell therapies for cancer treatment.” EBioMedicine vol. 106 (2024): 105266. doi:10.1016/j.ebiom.2024.105266
10. Zhou, Jing-E et al. “Lipid nanoparticles produce chimeric antigen receptor T cells with interleukin-6 knockdown in vivo.” Journal of controlled release : official journal of the Controlled Release Society vol. 350 (2022): 298-307. doi:10.1016/j.jconrel.2022.08.033
11. Billingsley, Margaret M et al. “In Vivo mRNA CAR T Cell Engineering via Targeted Ionizable Lipid Nanoparticles with Extrahepatic Tropism.” Small (Weinheim an der Bergstrasse, Germany) vol. 20,11 (2024): e2304378. doi:10.1002/smll.202304378
12. Meng, Sikun et al. “In Vivo Engineered CAR-T Cell Therapy: Lessons Built from COVID-19 mRNA Vaccines.” International journal of molecular sciences vol. 26,7 3119. 28 Mar. 2025, doi:10.3390/ijms26073119
13. Luo, Wenjing et al. “Selinexor Reduces the Immunosuppression of Macrophages and Synergizes With CD19 CAR-T Cells Against B-Cell Lymphoma.” Cancer science, 10.1111/cas.70123. 17 Jun. 2025, doi:10.1111/cas.70123
14. Karmali, Reem et al. “Impact of race and social determinants of health on outcomes in patients with aggressive B-cell NHL treated with CAR-T therapy.” Blood advances vol. 8,10 (2024): 2592-2599. doi:10.1182/bloodadvances.2023011996
15. Fan, Mingliang et al. “Nanoparticle-mediated universal CAR-T therapy.” International journal of pharmaceutics vol. 666 (2024): 124779. doi:10.1016/j.ijpharm.2024.124779
16. Marchais, Maude et al. “FOXO1 Inhibition Generates Potent Nonactivated CAR T Cells against Solid Tumors.” Cancer immunology research vol. 11,11 (2023): 1508-1523. doi:10.1158/2326-6066.CIR-22-0533
17. Zhu, Shigui. “CAR-T in cancer therapeutics and updates.” Journal of the National Cancer Center vol. 4,3 189-194. 9 Jan. 2024, doi:10.1016/j.jncc.2024.01.001
18. Le Guen, Camille et al. “Les structures académiques peuvent-elles permettre une amélioration de l’accessibilité aux CAR-T cells ?” [Can academic structures improve access to CAR-T cells?]. Bulletin du cancer vol. 111,1 (2024): 62-72. doi:10.1016/j.bulcan.2023.10.005
19. Cai, Jingsheng et al. “RNA technology and nanocarriers empowering in vivo chimeric antigen receptor therapy.” Immunology vol. 173,4 (2024): 634-653. doi:10.1111/imm.13861
20. Cai, Jingsheng et al. “RNA technology and nanocarriers empowering in vivo chimeric antigen receptor therapy.” Immunology vol. 173,4 (2024): 634-653. doi:10.1111/imm.13861
21. Mu, Wenli et al. “Rapamycin enhances CAR-T control of HIV replication and reservoir elimination in vivo.” The Journal of clinical investigation vol. 135,7 e185489. 11 Feb. 2025, doi:10.1172/JCI185489
22. Chang, Yun et al. “Advancements in adoptive CAR immune cell immunotherapy synergistically combined with multimodal approaches for tumor treatment.” Bioactive materials vol. 42 379-403. 10 Sep. 2024, doi:10.1016/j.bioactmat.2024.08.046
23. Chang, Yun et al. “Advancements in adoptive CAR immune cell immunotherapy synergistically combined with multimodal approaches for tumor treatment.” Bioactive materials vol. 42 379-403. 10 Sep. 2024, doi:10.1016/j.bioactmat.2024.08.046
24. Niu, Huanqing et al. “Biomaterials for chimeric antigen receptor T cell engineering.” Acta biomaterialia vol. 166 (2023): 1-13. doi:10.1016/j.actbio.2023.04.043
25. Li, Tingxuan et al. “Advanced Materials and Delivery Systems for Enhancement of Chimeric Antigen Receptor Cells.” Small methods vol. 7,11 (2023): e2300880. doi:10.1002/smtd.202300880
26. Tomasik, Jaromir et al. “Next generations of CAR-T cells - new therapeutic opportunities in hematology?.” Frontiers in immunology vol. 13 1034707. 28 Oct. 2022, doi:10.3389/fimmu.2022.1034707
27. Wu, Suwan et al. “Inhibition of VEGF signaling prevents exhaustion and enhances anti-leukemia efficacy of CAR-T cells via Wnt/β-catenin pathway.” Journal of translational medicine vol. 23,1 494. 30 Apr. 2025, doi:10.1186/s12967-024-05907-z
28. Wang, Zhengyi et al. “Influencing factors and solution strategies of chimeric antigen receptor T-cell therapy (CAR-T) cell immunotherapy.” Oncology research vol. 32,9 1479-1516. 23 Aug. 2024, doi:10.32604/or.2024.048564
29. Chen, Yaping et al. “Nanoinjection: A Platform for Innovation in Ex Vivo Cell Engineering.” Accounts of chemical research vol. 57,12 (2024): 1722-1735. doi:10.1021/acs.accounts.4c00190
30. Sun, Chao et al. “BATF-mediated regulation of exhausted CD8+ T-cell responses and potential implications for chimeric antigen receptor-T therapy.” Immunotherapy vol. 16,5 (2024): 331-340. doi:10.2217/imt-2023-0170
31. Nezvalova-Henriksen, Katerina et al. “Implementation and operational management of marketed chimeric antigen receptor T cell (CAR-T Cell) therapy-a guidance by the GoCART Coalition Pharmacist Working Group.” Bone marrow transplantation vol. 58,10 (2023): 1069-1074. doi:10.1038/s41409-023-02072-7
32. Meller, Leo et al. “Bilateral Cytomegalovirus Retinitis After Chimeric Antigen Receptor T-cell Therapy for B-cell Lymphoma.” Cureus vol. 16,3 e56637. 21 Mar. 2024, doi:10.7759/cureus.56637
33. Álvarez-Benedicto, Ester et al. “Spleen SORT LNP Generated in situ CAR T Cells Extend Survival in a Mouse Model of Lymphoreplete B Cell Lymphoma.” Angewandte Chemie (International ed. in English) vol. 62,44 (2023): e202310395. doi:10.1002/anie.202310395
34. Zhang, Jiqin et al. “Non-viral, specifically targeted CAR-T cells achieve high safety and efficacy in B-NHL.” Nature vol. 609,7926 (2022): 369-374. doi:10.1038/s41586-022-05140-y