- 1Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- 2Department of Clinical Biochemistry, Faculty of Medical Science, Tarbiat Modarres University, Tehran, Iran
- 3Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
- 4Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
Allogenic chimeric antigen receptor T (CAR-T) cells have advantages compared to autologous T cell therapies such as availability cells for production, a suitable HLA-matched donor (if graft-vs-host-disease and rejection effects are to be avoided and also lower risks associated with transduction methods in process of autologous CAR-T cells). In recent years, the additional editing and non-editing technologies are helping to make allogenic CAR-T therapies a hopeful future treatment. Universal off-the-shelf CAR-T cells can be solved key issues include preventing graft-versus-host disease (GVHD) and time consumption and other challenges faced to allogenic CAR-T cells. Here, we have highlighted the improvement in CAR-T development, particularly in engineering allogenic CAR-T, clinical practices related to these, pre-clinical and clinical studies and their successes which investigated in recent 10 years related to treatment of hematological malignancies and cancers by allogenic CAR-T cells.
1 Introduction
Recent advancements in molecular and cellular biology, coupled with an enhanced understanding of the function and nature of tumors, have led to several innovative strategies in targeted immunotherapy. Key therapeutic approaches in this domain now encompass monoclonal and bi-specific antibodies, antibody-drug conjugates, checkpoint inhibitors, and the latest advancements in adoptive cell therapy (ACT). Over the past decade, CAR-T immunotherapy has emerged as a pioneering approach to ACT. CAR-T cell therapy is not only used for cancer treatment but is also being investigated for various diseases, broadening the potential applications and impacts in the medical field. This innovative form of immunotherapy has demonstrated exceptional efficacy in treating hematopoietic cancers and shows considerable promise for addressing the challenges associated with solid tumors (1, 2). A CAR is a synthetic construct that comprises an extracellular, a spacer or hinge region, a transmembrane domain, and one or more intracellular signaling domains, expressed on the T cell surface and other immune cells through non-viral or viral transduction recognizing and interacting with tumor-associated antigens (3). In addition, to overcome the limitation of lentiviral transduction in primary T cells, scientists use high multiplicities of infection (MOI) of the virus by standard protocols. As shown in Figure 1, MOI represents the ratio of infectious agents, typically viral particles, to target cells in a defined system, such as a cell culture. The use of high MOI can affect the success of process through increasing both the time and cost of producing CAR-T cells (4).

Figure 1. Multiplicity of infection (MOI) represents the ratio of infectious agents, typically viral particles, to target cells in a defined system, such as a cell culture. For instance, adding 10 million viruses to 1 million cells results in a MOI of 10, indicating an average of 10 viral particles per cell.
According to different purposes, the design of CAR-T cells varies significantly, requiring careful consideration and optimization of multiple factors to achieve optimal therapeutic outcomes. Once the CAR genes are successfully incorporated into the T-cell genome, the modified T cells are expanded to achieve the desired therapeutic dosage. The expanded CAR-T cells are administered to the patient, leverage the patient’s own, and employ genetically engineered T cells to direct potent therapeutic responses to target and eradicate cancer cells specifically (5). To date, most FDA-approved CAR-T cell products are derived from autologous immune cells including Tisagenlecleucel (Kymriah, Novartis) highly effective in treating patients with relapsed or refractory CD19-positive hematological malignancies, Axicabtagene ciloleucel (Yescarta, Kite Pharmaceuticals)Approved for the treatment of large B-cell lymphoma, Brexucabtagene autoleucel (Tecartus, Kite Pharmaceuticals) Targets B-cell acute lymphoblastic leukemia (ALL) and mantle cell lymphoma (MCL), Idecabtagene vicleucel (Abecma) and Carvykti® (ciltacabtagene autoleucel) used for the treatment of relapsed or refractory multiple myeloma, Breyanzi® (lisocabtagene maraleucel) for second-line treatment of large B-cell lymphoma (6). These therapies have demonstrated significant efficacy in their respective indications, providing new treatment options for patients with limited therapeutic choices. Although the success of currently approved autologous CAR-T cell products can cure up to 35-40% of patients (7) and have a lower risk of immunologic incompatibility between donor and recipient, their widespread application faces significant clinical and economic challenges. The complex manufacturing process for these therapies begins with collecting peripheral blood mononuclear cells (PBMCs) from each patient through leukapheresis. T cells are genetically engineered to produce CAR-T cells with enhanced antitumor functions, subsequently reinfused into the patient. Treatment preparation is a time-consuming process that typically takes about three weeks, which can be particularly problematic for patients with rapidly advancing conditions, such as acute leukemia. Additionally, patients often undergo lymphodepletion through chemotherapy and radiotherapy, making it challenging to collect T cells that meet the required standards for both quantity and quality and can result in a manufacturing failure rate of 2% to 10%. Also, the effectiveness of CAR-T cells can be diminished by T cell exhaustion, a common T cell dysfunction observed in cancer patients (8–10). On the other hand, the heterogeneity in tumor antigen expression and the immune evasion strategies employed by tumor cells, necessitate the use of CAR-T cell products that can target multiple antigen specificities. Finally, autologous cell therapy is designed for individual patients, incurring significant costs and restricting its broader application allogenic CAR T-cell therapy, sourced from healthy donors who are human leukocyte antigen (HLA)-matched or gene-edited cells modified for use in non-HLA-matched patients, offers several advantages over autologous approaches (11). Healthy donors provide a high yield of cells from a single individual, and their PBMCs are in optimal condition, having not been subjected to chemotherapy or radiotherapy. The immediate availability of cryopreserved batches and the potential for standardizing CAR-T cell products allow multiple modifications and combinations to target various tumor antigens, leading to more predictable outcomes. This “off-the-shelf” immunotherapeutic approach enables a single manufacturing run to produce doses for multiple patients or multiple doses for a single patient. By scaling up production and establishing a bank of CAR immune cells from healthy donors, the cost per patient can be reduced, and access to the therapy can be broadened, simplifying supply chain logistics and eliminating the need for bridging therapies (12, 13). Figure 2 compares two steps in autologous and allogeneic CAR-T cell therapy, including cell sourcing and genetic modification.
Autologous CAR-T therapy uses a patient’s T cells, genetically modified to express a CAR, minimizing immune rejection but facing challenges like high cost, time delays, and variable T cell quality. Allogeneic CAR-T therapy, sourced from healthy donors or iPSCs, offers scalable, off-the-shelf treatment with faster delivery but risks complications like GvHD and immune rejection. Genetic modifications, including TCR disruption, HLA ablation, and NK cell inhibitory ligand overexpression, aim to reduce these risks and improve persistence and efficacy.
Allogenic CAR-T cell therapy faces significant challenges. One major issue arises when the T-cell receptor (TCR) on the surface of allogenic CAR-T cells recognizes and attacks the patient’s healthy tissues, leading to GvHD risk. Additionally, host-versus-graft (HvG) reactions can occur, in which the patient’s immune system identifies the CAR-T cells as foreign and eliminates them. To address these issues, researchers can choose T-cell sources with lower TCR signaling capacity to minimize them. Furthermore, CAR-T cells genetically edited (using ZFN, TALEN, CRISPR/Cas9) are being developed to reduce the risk of immune rejection and improve their effectiveness. However, these technologies come with significant challenges and limitations, such as Double-Strand Breaks (DSBs) in DNA leading to unintended mutations, genomic instability, potential safety risks, and lack of modulation resulting in off-target effects and unpredictable outcomes (14–16). Also, CAR T-cell therapy can have several side effects. Immediate effects include cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) and long-term side effects can involve hypogammaglobulinemia and cytopenias (17). Despite these obstacles, continuous research and progress in gene-editing technologies show promise for enhancing the safety and efficacy of allogenic CAR T-cell therapies. In this review, we aim to provide a thorough overview of the latest advancements and challenges in CAR-T cell therapy, its clinical applications, and recent innovations in gene editing technologies. We will also discuss the challenges and side effects associated with CAR-T cell therapy, along with strategies to mitigate these effects. Our focus is to highlight the remarkable potential of CAR-T cell therapy to transform cancer treatment and significantly improve patient outcomes.
2 Source of allogeneic cells
Given the rising demand for CAR therapy, exploring novel immunotherapy strategies is crucial. Both autologous and allogeneic T cells face limitations, including restricted expansion and exhaustion. Allogeneic CAR-T cells also pose a risk of GvHD. While PBMCs are the primary source for allo-CAR-Ts, iPSCs, and umbilical cord blood offer a scalable, antigen-specific T-cell supply with optimized therapeutic features, addressing these challenges.
2.1 Peripheral blood mononuclear cells
PBMCs are typically obtained from the blood of healthy donors, presenting an opportunity to create a cell bank that includes various subtypes of the HLA complex. It allows for selecting batches that match the HLA types to produce multiple vials from a single apheresis product, facilitating a rapid and standardized manufacturing protocol (18).
2.2 Umbilical cord blood cells
While there are challenges, such as the limited availability of UCB cells, their benefits are substantial. UCB cells are a valuable source of hematopoietic stem cells (HSCs) with distinct advantages over adult T cells. UCB cells are particularly “antigen-naïve,” which reduces their alloreactivity and allows for greater flexibility in HLA matching (19). Additionally, reduced NFAT signaling and lower NF-κB activation lead to decreased production of pro-inflammatory cytokines, which helps reduce the frequency and severity of GvHD (20). Furthermore, UCB cells exhibit much lower exhaustion markers like PD-1, TIM-3, and LAG-3 than peripheral blood T cells, enhancing their long-term persistence and effectiveness (21).
2.3 Induced pluripotent stem cells
iPSCs can proliferate indefinitely while retaining pluripotency, enabling the generation of diverse, genetically modified CAR-T cells with superior proliferation capacity and longer telomeres than mature T cells. This feature allows for the feasible generation of a diverse range of genetically modified CAR-T cells, which enhances antitumor effectiveness, significantly reduces immunogenicity, improves compatibility, and effectively mitigates the risk of allorejection (22).
The generation of iPSC-derived CAR-T cells from adult somatic cells generally involves two key steps:(a) Reprogramming somatic cells into iPSCs via the introduction of transcription factors such as Oct4, Sox2, Nanog, Lin28, Klf4, and c-Myc (23), and TCR modification to generate hypoimmunogenic iPSC lines;(b) Genetic engineering, where CAR transgenes are introduced into iPSC-derived T cells to direct tumor-specific targeting (Figure 3) (24).
A significant advantage of iPSCs is that CAR-T cells can be derived from a single clone, allowing for uniform clonal expansion and stable genetic modifications for reliable and effective treatment. In this regard, Jing et al. successfully generated mature T cells from human iPSCs using G9a/GLP inhibition and exhibited characteristics similar to mature alpha-beta T cells from peripheral blood, including effector and memory-like subpopulations (25). Kinoshita et al. introduced GD2-CARrejTs, a new iPSC-derived CAR-T cell therapy for small cell lung cancer (SCLC). These rejuvenated T cells showed more potent cytotoxic effects than conventional GD2-CARTs (26). In the realm of clinical applications, fate therapeutics is a leading company developing off-the-shelf iCAR-T products for various therapies, and its products, including FT819 (anti-CD19) and FT825/ONO-8250 (anti-HER2), are currently in Phase I clinical trials (27, 28). While stem cell-based approaches hold great promise, their successful clinical application will depend on ongoing innovation and rigorous evaluation.
Peripheral blood T cells, retaining their rearranged TCR gene, can be reprogrammed into iPSCs. These T-iPSCs can be engineered with CAR for enhanced tumor specificity in adoptive cell therapy or derived from antigen-specific T cell clones to produce targeted T cells.
3 Mechanisms of action: how allogenic CAR-T cells work
When human T cells are equipped with a CAR, they can effectively eradicate tumor cells in a MHC- and Fas-independent manner, making them versatile and powerful in targeting cancer cells (29). CAR-T cells engage with target cells by creating a unique immunological synapse. Once the immunological synapse is established, the CAR-T cells can induce tumor cell lysis through various pathways (30).
CAR-T cells primarily achieve tumor lysis through direct interactions with tumor cells. CAR-T cell initial activation was strongly dependent on the expression level of ICAM1 Cytokine production by activated CAR-T cells,enhances their anti-tumoral capabilities and is crucial in mediating tumor lysis via secondary mechanisms (31). CAR-T cells exhibit a disorganized lymphocyte-specific protein tyrosine kinase (Lck) arrangement and recruit lytic granules more quickly than TCRs, leading to a more rapid killing of tumor target cells and faster detachment from dying tumor cells by CAR-T cells (32). The perforin pathway and FasL (Fas Ligand) pathway can collaborate to kill target cells. This synergy is essential for achieving complete and durable tumor control by CAR-T cells (shown in Figure 4) (33).

Figure 4. (A) Structure of CAR T-cells. The structure of Chimeric Antigen Receptors (CARs) in CAR T-cells comprises several essential components: a single chain variable fragment (scFv), a hinge region along with a transmembrane (TM) region, costimulatory domains (such as 4-1BB, ICOS, CD28, and OX40), and a signaling domain (CD3ζ). First-generation CARs included only CD3ζ as the intracellular domain. In contrast, second- and third-generation CARs feature one or two costimulatory domains connected to CD3ζ, respectively. Fourth-generation CARs, referred to as “TRUCK” CARs, are structurally similar to second-generation CARs but incorporate an inducible cytokine expression (such as IL-12) via an NFAT-responsive promoter. Fifth-generation CAR-T cells include a JAK/STAT activation domain derived from IL-2RB, positioned between CD28 and CD247. This addition promotes cells proliferation, prevents terminal differentiation, and enhances persistence. (B) The mechanism of action of CAR T-cells involves the binding of CARs to specific antigens present on the surface of tumor cells through the scFv recognition domain. This interaction triggers anticancer effects by promoting the inflammatory cytokines’ secretion, such as IFN-γ, and facilitates cytolytic effector function via granzyme and perforin in an ICAM-1-dependent manner.
CAR-T cell therapies exhibit greater sensitivity to target antigens than antibody or antibody-drug conjugate therapies. They can detect and respond to lower levels of antigens present in tumor cells (34). In dealing with a tumor that changes over time, multifunctional CAR-T cells, which can be bispecific, switchable, or capable of delivering other therapeutic agents directly, can perform multiple actions simultaneously and be effective (35, 36).
4 Food and drug administration approval of CARTs
Since 2017, the FDA has authorized six CAR-T cells in the USA and other nations, and the National Medical Products Administration of China has approved two CAR-T products. These products target patients suffering from acute lymphoblastic leukemia (ALL), multiple myeloma (MM), or advanced/resistant large B-cell lymphoma (LBCL), in which significant results are acquired with total response rates that may reach as high as 100% measurable response rates in a number of cases (37, 38).
It was for the first time that the FDA approved CAR-T cells therapy on Aug 30th, 2017, to treat acute lymphoblastic leukemia in young adults and children. Later, the FDA approved three additional CD19-specific CAR-T cells for the purpose of treating various B cell malignancies, i.e., Breyanzi, Tecartus, and Yescarta. Even though the CAR expressed in Tecartus is the same as that in Yescarta, they have different manufacturing processes because while Yescarta does not involve the enrichment of T cells, Tecartus necessitates such an enrichment. Furthermore, february 2022 and april 2021 were the dates on which two BCMA-specific CAR-T cell therapies could successfully get approval for the treatment of a number of myelomas, i.e., ciltacabtagene autoleucel (Carvykti) and idecabtagene vicleucel (Abecma) (37). Table 1 offers detailed information regarding all FDA-approved CAR-T cells therapies (51).
5 Advances in CAR-T development: focus on off-the-shelf allogenic CAR-T
CAR-T therapy is an innovative treatment for certain cancers, especially hematologic malignancies, but traditional autologous CAR-T therapy has several limitations. Autologous CAR-T involves extracting and engineering T-cells from individual patients, making it time-consuming and expensive, with potential functional impairments in the engineered T-cells. Additionally, the process can take weeks, delaying treatment (52, 53). This main challenge has led to interest in off-the-shelf allogenic CAR-T cells (alloCAR-T), which use T-cells from a single healthy donor, offering faster availability, broader applicability, and cost-effectiveness. However, challenges remain, such as avoiding GVHD and host-mediated graft rejection (53, 54). Advances in genome-editing techniques and non-gene-editing technologies are helping to address these challenges, making alloCAR-T therapies a promising future treatment. Here, we have highlighted the progress in CAR-T development, particularly in engineering alloCAR-T cells.
5.1 Methods to engineer off-the-shelf allogenic CAR-T
As mentioned above, creating a universal off-the-shelf allo-CAR-T product requires overcoming key issues include preventing HvG, avoiding GvHD, preventing fratricide (where CAR-T cells attack each other), and enhancing the persistence of the infused allo-CAR-T cells. These challenges can be addressed through gene editing technologies and non-gene editing approaches. Additionally, supporting the expansion and persistence of CAR-T cells can be achieved by producing proinflammatory cytokines, such as IL-15 or IL-18, through transgenic methods (55, 56).
5.1.1 Gene editing technology
Gene editing technologies have become crucial for modifying allogenic CAR-T cells to reduce immune rejection, enhance functionality, and improve safety. Methods like homing endonucleases (also called meganucleases) (57), transcription activator-like (TAL) effector nucleases (TALENs) (52, 58–60), zinc-finger nucleases (ZFNs) (61), and CRISPR-Cas9 (62), are used to create precise genetic changes. However, multiple modifications can increase the risk of off-target mutations and chromosomal translocations, with CRISPR-Cas9, TALENs, and ZFNs posing higher risks than newer techniques like base editing and prime editing (52, 63, 64). Additionally, making several edits to CAR-T cells could lead to other unwanted effects, such as reduced in vivo persistency (64). Homing endonucleases, while highly specific, are challenging to engineer, limiting their use (65). MegaTALs, which combine homing endonucleases with TAL effector arrays, offer enhanced specificity and editing efficiency (66). However, both homing endonucleases and MegaTALs require custom engineering for specific targeting, which limited their large-scale adoption of these gene-editing platforms (67). TALENs and ZFNs induce double-strand breaks (DSBs) to disrupt target genes, but are costly and time-consuming (68–73). Despite these challenges, TALEN-edited CAR-T cells have been used in clinical trials to knock out αβTCR and CD52 (52, 58, 59). CRISPR-Cas9, using guide RNAs, is more flexible, cost-effective, and precise, and is widely used in clinical trials for modifying CAR-T cells (68, 74–77). Newer techniques like base editing and prime editing, which do not induce DSBs, offer fewer off-target effects and are being tested in clinical trials for CAR-T therapies. These advancements hold promise for safer, more effective off-the-shelf allo-CAR-T treatments (64, 78, 79) (Table 2). Genome-edited allogeneic CAR-T cells offer therapeutic potential but pose risks like chromosomal abnormalities and secondary malignancies, requiring long-term monitoring. CRISPR/Cas9 and other editing techniques may cause unintended genetic changes, emphasizing the need for vigilant safety assessments (53).
5.1.2 Non-gene editing approaches
Non-gene editing approaches to reduce GvHD in CAR-T therapy focus on inhibiting TCR signaling, using virus-specific T cells (VSTs), memory T cells, and γδ T cells. Truncated CD3 can inhibit TCR signaling to minimize GvHD while preserving CAR activity, though solid tumors present challenges due to antigen heterogeneity and immunosuppressive environments. VSTs, particularly those from HLA-matched donor libraries, have shown low GvHD risk, with CAR-transduced VSTs demonstrating anti-tumor effects. Another strategy is using emory T cells that being less alloreactive than naïve T cells, although their role in allogeneic CAR-T therapy remains underexplored. γδ T cells, which function independently of MHC complexes, provide a unique advantage by reducing GvHD and tumor escape risks. Overall, these non-gene editing strategies present safer, promising alternatives for improving CAR-T therapy efficacy while minimizing GvHD (80).
Although gene editing technologies have many advantages, their main challenge is safely removing multiple genes (multiplexing) while minimizing risks. As an alternative, non-gene-editing technologies provide an interesting approach to support the development of allogenic CAR-Ts in the future. Nowadays, three strategies have been developed as non-gene-edited approaches: 1) A TCR inhibitory molecule (TIM) that, when incorporated into T-cell DNA, competes with TCR elements, rendering the TCR unresponsive. This method was tested with an NKG2D-based CAR in metastatic colorectal cancer (81). 2) A miRNA scaffold targeting CD3ζ, which completely eliminates TCR expression, was assessed in a phase I clinical trial with a BCMA-targeting CAR-T for relapse/refractory multiple myeloma (82). 3) Third strategy involves retaining TCR/HLA-I in the endoplasmic reticulum (ER) to prevent GvHD and host-versus-graft (HvG) reactions. This is achieved by using a peptide like KDEL, combined with an scFv targeting the TCR, to retain all TCRs in the ER (83).
Removing the TCR from T-cells is beneficial, but factors influencing cellular persistence in an allogenic setting are still unclear. While HLA-I/II proteins play a role in HvG reactions, other factors like metabolic regulation may also affect persistence. Non-gene-editing approaches, such as combining miRNA or siRNA sequences to target multiple genes at once, make it easier to address these complexities (84–86). A new multiplex shRNA platform allows the targeting of up to four genes simultaneously, including those involved in GvHD (CD3ζ), HLA-I/HvG (B2M, CIITA), apoptosis (CD95), immune checkpoints (LAG-3), and co-stimulation, reduction/persistence (CD28) (87).
Gene-edited allogeneic CAR-T cells reduce GvHD risk through TCR knockout but face challenges like immune rejection, reduced persistence, and complex manufacturing. In contrast, non-edited CAR-T cells, including virus-specific, memory, and γδ T cells, offer safer alternatives with lower immune rejection and better persistence but may still pose some risk of allo-reactivity. While gene editing provides precise control, it also introduces regulatory and cost challenges. Non-edited approaches leverage natural T cell subsets for safety and efficacy, making them promising but requiring further optimization for tumor targeting (53).
5.2 Clinical experience with off-the-shelf allogenic CAR-T
5.2.1 Achievements so far
The clinical development of off-the-shelf allogenic CAR-T therapies is still in its early stages, with no approvals yet. Early-phase trials show promising results, but challenges remain, including the risk of GvHD, graft rejection, and infections. Unlike autologous CAR-T treatments, which are costly and time-consuming due to patient-specific manufacturing, allogenic CAR-T therapies aim to offer a more accessible alternative, especially for patients who cannot afford personalized therapies. Progress has been slower than expected due to safety concerns and technical challenges in gene editing, though recent advancements are improving their potential. The first use of allogenic CAR T cells in humans was reported in 2017, with UCART19, a TALEN-edited, αβTCR/CD52-knockout product. It was evaluated in two Phase I studies (PALL and CALM) for relapsed or refractory B-cell acute lymphoblastic leukemia (R/R B-ALL) in pediatric and adult populations. The trials showed promising results, especially in infants, who were successfully bridged to hematopoietic stem cell transplantation (HSCT) (52, 58, 59). In 2020, combined data from two Phase I trials of UCART19 in children (7 patients) and adults (14 patients) with relapsed/refractory B-cell acute lymphoblastic leukemia (R/R B-ALL) demonstrated a promising complete response (CR/CRi) rate of 67%, although treatment-related deaths occurred (58). Updated results from the adult trial in 2022 (25 patients) showed an overall response rate (ORR) of 48%, with a median response duration of 7.4 months and median overall survival of 13.4 months. Importantly, GvHD was rare, with only two patients experiencing mild grade 1 acute skin-related GvHD (59) (Supplementary Table 3).
In the ALPHA and ALPHA2 studies, an anti-CD19 CAR-T therapy, a second dose of ALLO-501/ALLO-501A was given approximately 30 days after the first infusion to enhance the persistence of CAR-T cells in peripheral blood beyond day 28, aiming to improve response duration (88, 89). In 2023, a Phase I trial of ALLO-715, a TALEN-edited CAR-T product targeting BCMA for relapsed/refractory multiple myeloma (R/R MM), showed an overall response rate (ORR) of 55.8% among 43 patients. Although no cases of GvHD were reported, 88% of patients experienced grade ≥3 adverse events, and 53.5% had infections, with 23.3% experiencing severe infections, including 7% fatal cases (60). Additionally, ALLO-316, an anti-CD70 CAR-T product, is being tested in the Phase 1 TRAVERSE study for patients with advanced or metastatic clear cell renal cell carcinoma (CcRCC) (90). TALEN-edited CAR-T products like UCART123 (anti-CD123) and UCART22 (anti-CD22) are also being tested in Phase I trials for R/R B-ALL and AML (BALLI-01 and AMELI-01 studies respectively), with 70.8% of patients showing objective responses and no cases of GvHD (91, 92). A clinical hold was imposed on UCART123 in the ABC study following a fatality, resulting in dose adjustments and a recommendation to restrict the total cyclophosphamide dose (87, 93).
Supplementary Table 3 summarizes key clinical trials applying different gene-editing and non-gene editing approaches in the development of allogeneic CAR-T cells therapies.
GRm13Z40-2 is a CAR-T cell therapy targeting the IL-13 receptor α2 (IL13Rα2) in glioblastoma patients. In its manufacturing, ZFNs were used to knock out the glucocorticoid receptor in T cells, enabling patients to receive high-dose glucocorticoids without compromising CAR T-cell function. The therapy was administered intracranial in four doses, along with recombinant IL-2 to enhance T-cell growth and persistence. The treatment was generally well tolerated, with no signs of GVHD. In terms of efficacy, 66% of patients showed temporary clinical responses, including tumor size reductions or necrosis (61).
The ARCUS genome-editing technology is used in the anti-CD19 allogenic CAR-T product PBCAR0191, currently being tested in a Phase I/II study for R/R non-Hodgkin’s lymphoma (R/R NHL) and B-ALL (NCT03666000). Another allogenic CAR-T product, PBCAR19B, designed to reduce immune rejection by host T-cells and NK cells, indicated a 71% ORR and a 43% CR rate in a Phase I study (NCT04649112) during 2021-2023 years in 13 patients (94).
CRISPR/Cas9 is being utilized in several promising CAR-T therapies for various cancers. CTX110, targeting CD19, is currently being evaluated in the Phase 1 CARBON trial for relapsed/refractory non-Hodgkin’s lymphoma (r/r NHL), demonstrating a 67% overall response rate (ORR) at the highest dose, with no GvHD despite significant HLA mismatch. A second infusion provided additional benefit (95, 96). CTX-130 (as an anti-CD70 CAR-T) has been tested in the COBALT-LYM (T-cell lymphoma) and COBALT-RCC (advanced clear cell renal cell carcinoma) studies. In T-cell lymphoma, the ORR was 71% with no GvHD, while in RCC, the ORR was 8%, though one patient achieved a durable complete response (97, 98). CB-010 (as an anti-CD19 CAR-T) with CRISPR-edited TRAC and PD-1 knockouts, demonstrated a 94% ORR in the ANTLER Phase 1 trial for B-NHL patients, which is comparable to autologous CAR-T therapies, and showed no GvHD (99). CRISPR-edited CD7-targeted CAR-T cells for T-ALL exhibited high efficacy, with complete response rates ranging from 71% to 91%, and no GvHD or neurotoxicity (62, 77). Early trials have also shown promising results for NK/T lymphomas and AML (75, 100).
However, alternative gene-editing methods have also been explored. P-BCMA-ALLO1, an allogenic CAR-T targeting BCMA for relapsed/refractory multiple myeloma (RRMM), is manufactured using the piggyBac® DNA Delivery System and the Cas-CLOVER™ gene editing system, which eliminates TCR expression and reduces MHC class I expression to prevent GvHD. It showed strong activity in MM xenografts and is being tested in a Phase I study. P-MUC1C-ALLO1, targeting MUC1-C for common solid tumors, is also produced with piggyBac® and Cas-CLOVER™, leading to TCR and MHC class I knockout for an enriched T stem cell memory product (101, 102). Another approach used base-pair editing in a Phase I trial of CD7 CAR-T cells. Preliminary results showed mixed outcomes, including one patient achieving leukemic remission, another undergoing stem cell transplantation, and a third developing a fatal fungal infection. Adverse events included cytokine release syndrome and multilineage cytopenia (79).
CAR-T products using non-gene-editing technologies have also been tested in clinical trials. CYAD-101, an allogenic CAR-T designed to co-express an NKG2D-based CAR and an inhibitory peptide to interfere with TCR signaling, was tested in the alloSHRINK Phase I study for unresectable metastatic colorectal cancer. After standard chemotherapy, 25 patients received three infusions of CYAD-101. There were no dose-limiting toxicities (DLT) or graft-versus-host disease (GvHD), and the overall response rate (ORR) was 13%, with 60% achieving stable disease (81, 103). CYAD-211, using a miRNA-based shRNA approach to silence the CD3ζ component of the TCR while co-expressing an anti-BCMA CAR, was tested in the IMMUNICY-1 Phase I trial for relapsed/refractory multiple myeloma. Early results from 12 patients showed 3 partial responses and 8 stable diseases, with no DLT, GvHD, or neurotoxicity (104). A CD19-targeting allogenic CAR-T, using intracellular retention to prevent TCR expression, was tested in a Phase I study for non-Hodgkin’s lymphoma, showing no GvHD and a 75% ORR in the first eight patients (83). Lastly, FT819, an iPSC-derived CD19-targeting CAR-T, was evaluated in a Phase I study for B-cell malignancies, showing a tolerable safety profile with no DLT or GvHD in 12 patients (105) (Supplementary Table 3).
6 The major challenges that must be resolved for the purpose of allogenic CAR-T therapy
In spite of their many favorable traits, a number of challenges are also ascribable to allogenic CAR-T cell. In fact, allogenic T cells may result in severe GVHD, which is predominantly caused by a TCR-mediated immune response to the life-threatening host tissues. In turn, the host immune system may also lead to the induction of allorejection, which can result in impeded anti-tumor activity and limited effectiveness of the therapy. This is attributable to the fact that through the detection of nonself- HLA class I and class II proteins found on the membrane of donor T-cells, the host immune cells can mainly detect and eliminate allogeneic cells. This is the primary objective of allogeneic CAR-Ts, which limits their duration of response and activity (38).
When designing allogeneic CAR-Ts, one of the most commonly used strategies to prevent GVHD is the creation of TCR-deficient T cells by employing genome editing tools, including as TALENs (106, 107), CRISPR/Cas9 (107, 108), and ZFN (3, 4).
A number of allogeneic candidates have attained the same objective response rates as the ones witnessed in their autologous counterparts on the clinical scale, and irrespective of two patients (one adult and one infant) presenting with acute skin GvHD Grade I, which was controlled easily (59), the preliminary data obtained from those investigations totally indicated no evidence of acute GvHD. As a result, in spite of earlier concerns, the modifications that were conducted in order to avoid GvHD in allogeneic ‘off-the-shelf’ CAR-Ts apparently suffice to decrease such a risk drastically. In addition, the strategies used to decrease allorejection are also analyzed, which test repeated rounds of infusion by employing chemotherapy-resistant CAR-Ts. This allows for deeper/prolonged lymphopenia or genetic elimination of the main molecules that govern the immunogenicity of the CAR-T cells (38).
For instance, the studies on CALM indicated that even though the expansion of CAR-T cells took place 8 to 14 days after injection, a rapid decline was experienced by most participants by day 28, which indicated a limited duration of response (109). Different lymphodepleting regimens were tested that showed the intensity was positively correlated with the cellular persistence; however, the risk of infections was increased, as observed in the UNIVERSAL investigation with a fifth-grade fungal pneumonia incident (60). In order to address this issue, the subsequent investigations suggested a second dose of 501A and ALLO-501 in order to maintain the levels of CAR-T (89). Nonetheless, the safety concerns resulted in dose adjustment recommendations in investigations such as ABC and incidents of grade 5 pneumonia associated with conditioning regimens (87).
One of the popular techniques employs genome editing of MHC class I proteins through the disruption of the β2-microglobulin (β2M) locus (38). The engineering of the PBCAR19B cells was targeted to knock-down(human β2M) and express a HLA-E transgene in order to avoid allorejection (87). The proof-of-concept provided by the preliminary record results indicated that such modifications were apparently successful in delaying the host NK-and T- cells’ recovery. In much the same way, it has been reported that CTX-130, which is a modified anti-CD70 allo -CAR-T used for the disruption of the CD70 and β2M genes so as to decrease the fratricide and allorejection, elicits a durable complete response in an individual suffering from Renal Cell Carcinoma (RCC) (110), This may indicate that, even in solid tumors, the technique is actually enhancing the allogeneic CAR-Ts’ activity. Currently, CB-011, which is an anti-BCMA allo-CAR-Ts engineered with CRISPR/CAS12a to knock out both TRAC and β2M while co-expressing a β2M-HLA-E fusion peptide, is under evaluation in the CaMMouflage Phase 1 trial and has presented encouraging preclinical data, resulting in noticeably enhanced durability of anti-tumor activity (87).
In addition, the creation of a bank of allogeneic T cells is also an alternative, which has been predominantly utilized for non-modified and virus-specific T cells but also for anti-CD123 retrovirally transduced CAR T for the purpose of treating acute myeloid leukemia (38, 111).
One of the big concerns is the safety risks associated with the application of gene-editing technologies. In one patient receiving a consolidation dose of ALLO-501A, a chromosomal abnormality was recorded, which resulted in stopping all the investigations with the same technology for several months (89). The studies found that the observed chromosomal abnormality was not associated with the TALEN gene manufacturing or editing process; however, it questioned the safety consideration of genome-edited cellular therapies.
In recent years, the FDA has noted a risk of T-cells malignancies, which is applicable to all currently authorized therapies using CAR-Ts. Such concerns are raised regarding autologous CAR T-cells that can persist for a long duration. Given that allogenic CAR-Ts lack persistence, such a safety concern does not apply at present. As the technology is enhanced progressively, the increased persistence of the CAR-T cells may result in the same safety concerns (37, 87).
7 Conclusions and future perspective
Comprehensive investigations are essential to establish the efficacy of allogeneic CAR-T therapies compared to approved autologous CAR-Ts, particularly for treating solid tumors with limited therapeutic options. Current findings suggest that allogeneic CAR-Ts have the potential to overcome significant barriers to access for a broader patient population. This advancement stems from innovative approaches that disrupt TCRs and mitigate GvHD, which poses a major toxicity risk in allogeneic T-cells therapies. Genetic TCRα ablation using targeted genome-editing techniques has gained popularity, yet challenges remain, including the risk of DNA double-strand breaks and complexities in manufacturing that may impact cell yield and fitness. Non-genome-editing tools present a promising alternative, offering potentially safer and more adaptable methods for developing next-generation CAR-Ts. Despite the encouraging prospects of these techniques in reducing GvHD risks, a critical challenge remains: managing the HvG reaction post-infusion. Therefore, it is imperative for “off-the-shelf” allogeneic CAR-Ts to address both GvHD and HvG reactions. This can be achieved through various modifications, such as the disruption or downregulation of genes involved in allorejection, including CIITA, CD52, and B2M.To enhance the development of allogeneic CAR-T cells, we recommend focusing on the following strategies:
● Investing in alternative genetic modifications that minimize GvHD and HvG risks.
● Exploring non-genome-editing approaches to simplify manufacturing and improve cell viability.
● Conducting preclinical and clinical trials to assess the safety and efficacy of modified allogeneic CAR-Ts in diverse patient populations.
● Developing robust monitoring systems to track post-infusion reactions and optimize patient management.
In conclusion, continued investment in innovative technologies is essential for the safe infusion of allogeneic CAR-T cells while enhancing their efficacy, persistence, and safety profiles.
Author contributions
MA: Writing – original draft, Writing – review & editing. AS: Writing – original draft, Writing – review & editing. MS: Writing – original draft. SH: Writing – original draft. MA: Writing – review & editing. MV: Conceptualization, Writing – review & editing.
Funding
The author(s) declare that no financial support was received for the research and/or publication of this article.
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The author(s) declare that no Generative AI was used in the creation of this manuscript.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
Supplementary material
The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fimmu.2025.1557157/full#supplementary-material
References
1. Ayala Ceja M, Khericha M, Harris CM, Puig-Saus C, and Chen YY. CAR-T cell manufacturing: major process parameters and next-generation strategies. J Exp Medicine. (2024) 221:e20230903. doi: 10.1084/jem.20230903
2. Ghorai SK and Pearson AN. Current strategies to improve chimeric antigen receptor T (CAR-T) cell persistence. Cureus. (2024) 16:e65291. doi: 10.7759/cureus.65291
3. Ahmad U, Khan Z, Ualiyeva D, Amissah OB, Noor Z, Khan A, et al. Chimeric antigen receptor T cell structure, its manufacturing, and related toxicities; A comprehensive review. Adv Cancer Biology-Metastasis. (2022) 4:100035. doi: 10.1016/j.adcanc.2022.100035
4. Fichter C, Aggarwal A, Wong AKH, McAllery S, Mathivanan V, Hao B, et al. Modular lentiviral vectors for highly efficient transgene expression in resting immune cells. Viruses. (2021) 13. doi: 10.3390/v13061170
5. Sterner RC and Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. (2021) 11:69. doi: 10.1038/s41408-021-00459-7
6. Bhaskar ST, Dholaria B, Savani BN, Sengsayadeth S, and Oluwole O. Overview of approved CAR-T products and utility in clinical practice. Clin Hematol Int. (2024) 6:93. doi: 10.46989/001c.124277
7. Brudno JN, Maus MV, and Hinrichs CS. CAR T cells and T-cell therapies for cancer: A translational science review. JAMA. (2024). doi: 10.1001/jama.2024.19462
8. Zhang Z, Liu S, Zhang B, Qiao L, Zhang Y, and Zhang Y. T cell dysfunction and exhaustion in cancer. Front Cell Dev Biol. (2020) 8:17. doi: 10.3389/fcell.2020.00017
9. Baguet C, Larghero J, and Mebarki M. Early predictive factors of failure in autologous CAR T-cell manufacturing and/or efficacy in hematologic Malignancies. Blood Advances. (2024) 8:337–42. doi: 10.1182/bloodadvances.2023011047
10. Caldwell KJ, Gottschalk S, and Talleur AC. Allogeneic CAR cell therapy—more than a pipe dream. Front Immunol. (2021) 11:618427. doi: 10.3389/fimmu.2020.618427
11. Depil S, Duchateau P, Grupp S, Mufti G, and Poirot L. ‘Off-the-shelf’allogeneic CAR T cells: development and challenges. Nat Rev Drug discovery. (2020) 19:185–99. doi: 10.1038/s41573-019-0051-2
12. Aparicio C, Acebal C, and González-Vallinas M. Current approaches to develop “off-the-shelf” chimeric antigen receptor (CAR)-T cells for cancer treatment: a systematic review. Exp Hematol Oncology. (2023) 12:73. doi: 10.1186/s40164-023-00435-w
13. Aftab BT, Sasu B, Krishnamurthy J, Gschweng E, Alcazer V, and Depil S. Toward “off-the-shelf” allogeneic CAR T cells. Adv Cell Gene Ther. (2020) 3:e86. doi: 10.1002/acg2.v3.3
14. Valton J, Guyot V, Marechal A, Filhol J-M, Juillerat A, Duclert A, et al. A multidrug-resistant engineered CAR T cell for allogeneic combination immunotherapy. Mol Ther. (2015) 23:1507–18. doi: 10.1038/mt.2015.104
15. Moradi V, Khodabandehloo E, Alidadi M, Omidkhoda A, and Ahmadbeigi N. Progress and pitfalls of gene editing technology in CAR-T cell therapy: a state-of-the-art review. Front Oncology. (2024) 14:1388475. doi: 10.3389/fonc.2024.1388475
16. Mohty R and Lazaryan A. Off-The-Shelf” allogeneic chimeric antigen receptor T-cell therapy for B-cell Malignancies: current clinical evidence and challenges. Front Oncology. (2024) 14:1433432. doi: 10.3389/fonc.2024.1433432
17. Schroeder T, Martens T, Fransecky L, Valerius T, Schub N, Pott C, et al. Management of chimeric antigen receptor T (CAR-T) cell-associated toxicities. Intensive Care Medicine. (2024) 50:1459–69. doi: 10.1007/s00134-024-07468-7
18. Chen S and van den Brink MR. Allogeneic “Off-the-Shelf” CAR T cells: challenges and advances. Best Pract Res Clin Haematology. (2024) 101566. doi: 10.1016/j.beha.2024.101566
19. Caël B, Galaine J, Bardey I, Marton C, Fredon M, Biichle S, et al. Umbilical cord blood as a source of less differentiated T cells to produce CD123 CAR-T cells. Cancers. (2022) 14:3168. doi: 10.3390/life13091794
20. Devi S, Bongale AM, Tefera MA, Dixit P, and Bhanap P. Fresh umbilical cord blood—a source of multipotent stem cells, collection, banking, cryopreservation, and ethical concerns. Life. (2023) 13:1794. doi: 10.3390/bioengineering12010060
21. Lin Y, Lin J, Huang J, Chen Y, Tan J, Li Y, et al. Lower T cell inhibitory receptor level in mononuclear cells from cord blood compared with peripheral blood. Stem Cell Investigation. (2019) 6:35. doi: 10.1097/MOH.0000000000000195
22. 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) 12:1491282. doi: 10.3389/fcell.2024.1491282
23. Sadeqi Nezhad M, Abdollahpour-Alitappeh M, Rezaei B, Yazdanifar M, and Seifalian AM. Induced pluripotent stem cells (iPSCs) provide a potentially unlimited T cell source for CAR-T cell development and off-the-shelf products. Pharmaceutical Res. (2021) 38:931–45. doi: 10.1007/s11095-021-03067-z
24. Zong J and Li Y-R. iPSC technology revolutionizes CAR-T cell therapy for cancer treatment. Bioengineering. (2025) 12:60. doi: 10.3390/bioengineering12010060
25. Jing R, Falchetti M, Han T, Najia M, Hensch L, Meader E, et al. Generation of functional iPSC-derived CAR-T cells for cancer immunotherapy via G9a/GLP inhibition. Blood. (2024) 144:2043. doi: 10.1182/blood.2024024029
26. Kinoshita S, Ishii M, Ando J, Kimura T, Yamaguchi T, Harada S, et al. Rejuvenated iPSC-derived GD2-directed CART cells harbor robust cytotoxicity against small cell lung cancer. Cancer Res Communications. (2024) 4:723–37. doi: 10.1158/2767-9764.CRC-23-0414
27. Kirouac DC, Zmurchok C, and Morris D. Making drugs from T cells: The quantitative pharmacology of engineered T cell therapeutics. Appl. (2024) 10:31. doi: 10.1038/s41540-024-00355-3
28. Hosking MP, Arimori T, Shirinbak S, Omilusik K, Chandra S, Bickers C, et al. 269 Preferential targeting of HER2-expressing cancer cells by FT825/ONO-8250, an off-the-shelf iPSC-derived CAR-T cell incorporating novel synthetic mechanisms for enhanced solid tumor activity. BMJ Specialist Journals;. (2024). doi: 10.1136/jitc-2024-SITC2024.0269
29. Yang Y, Jacoby E, and Fry TJ. Challenges and opportunities of allogeneic donor-derived CAR T cells. Curr Opin hematology. (2015) 22:509–15. doi: 10.1097/MOH.0000000000000195
30. Xiong Y, Libby KA, and Su X. The physical landscape of CAR-T synapse. Biophys J. (2024) 123:2199–210. doi: 10.1016/j.bpj.2023.09.004
31. Yeku OO and Brentjens RJ. Armored CAR T-cells: utilizing cytokines and pro-inflammatory ligands to enhance CAR T-cell anti-tumour efficacy. Biochem Soc Trans. (2016) 44:412–8. doi: 10.1042/BST20150291
32. Davenport A, Cross R, Watson K, Liao Y, Shi W, Prince H, et al. Chimeric antigen receptor T cells form nonclassical and potent immune synapses driving rapid cytotoxicity. Proc Natl Acad Sci. (2018) 115:E2068–E76. doi: 10.1073/pnas.1716266115
33. Hassin D, Garber OG, Meiraz A, Schiffenbauer YS, and Berke G. Cytotoxic T lymphocyte perforin and Fas ligand working in concert even when Fas ligand lytic action is still not detectable. Immunology. (2011) 133:190–6. doi: 10.1111/j.1365-2567.2011.03426.x
34. Hosen N. Identification of cancer-specific cell surface targets for CAR-T cell therapy. Inflammation Regeneration. (2024) 44:17. doi: 10.1186/s41232-024-00324-6
35. Fu R, Li H, Li R, McGrath K, Dotti G, and Gu Z. Delivery techniques for enhancing CAR T cell therapy against solid tumors. Advanced Funct Materials. (2021) 31:2009489. doi: 10.1080/21645515.2024.2378543
36. Liu D, Zhao J, and Song Y. Engineering switchable and programmable universal CARs for CAR T therapy. J Hematol Oncology. (2019) 12:69. doi: 10.1186/s13045-019-0762-1
37. Mitra A, Barua A, Huang L, Ganguly S, Feng Q, and He B. From bench to bedside: the history and progress of CAR T cell therapy. Front Immunol. (2023) 14:1188049. doi: 10.3389/fimmu.2023.1188049
38. Martínez Bedoya D, Dutoit V, and Migliorini D. Allogeneic CAR T cells: an alternative to overcome challenges of CAR T cell therapy in glioblastoma. Front Immunol. (2021) 12:640082. doi: 10.3389/fimmu.2021.640082
39. Vandergriff A, Huang K, Shen D, Hu S, Hensley MT, Caranasos TG, et al. Targeting regenerative exosomes to myocardial infarction using cardiac homing peptide. Theranostics. (2018) 8:1869–78. doi: 10.1080/21645515.2024.2378543
40. Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. (2017) 377:2531–44. doi: 10.1056/NEJMoa1707447
41. Wang M, Munoz J, Goy A, Locke FL, Jacobson CA, Hill BT, et al. KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma. N Engl J Med. (2020) 382:1331–42. doi: 10.1056/NEJMoa1914347
42. Abramson JS, Palomba ML, Gordon LI, Lunning MA, Wang M, Arnason J, et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet. (2020) 396:839–52. doi: 10.1016/S0140-6736(20)31366-0
43. Munshi NC, Anderson LD Jr., Shah N, Madduri D, Berdeja J, Lonial S, et al. Idecabtagene vicleucel in relapsed and refractory multiple myeloma. N Engl J Med. (2021) 384:705–16. doi: 10.1056/NEJMoa2024850
44. Berdeja JG, Madduri D, Usmani SZ, Jakubowiak A, Agha M, Cohen AD, et al. Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): a phase 1b/2 open-label study. Lancet. (2021) 398:314–24. doi: 10.1016/S0140-6736(21)00933-8
45. Schuster SJ, Bishop MR, Tam CS, Waller EK, Borchmann P, McGuirk JP, et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med. (2019) 380:45–56. doi: 10.1056/NEJMoa1804980
46. Jacobson CA, Chavez JC, Sehgal AR, William BM, Munoz J, Salles G, et al. Axicabtagene ciloleucel in relapsed or refractory indolent non-Hodgkin lymphoma (ZUMA-5): a single-arm, multicentre, phase 2 trial. Lancet Oncol. (2022) 23:91–103. doi: 10.1016/S1470-2045(21)00591-X
47. Shah BD, Ghobadi A, Oluwole OO, Logan AC, Boissel N, Cassaday RD, et al. KTE-X19 for relapsed or refractory adult B-cell acute lymphoblastic leukaemia: phase 2 results of the single-arm, open-label, multicentre ZUMA-3 study. Lancet. (2021) 398:491–502. doi: 10.1016/S0140-6736(21)01222-8
48. Kamdar M, Solomon SR, Arnason J, Johnston PB, Glass B, Bachanova V, et al. Lisocabtagene maraleucel versus standard of care with salvage chemotherapy followed by autologous stem cell transplantation as second-line treatment in patients with relapsed or refractory large B-cell lymphoma (TRANSFORM): results from an interim analysis of an open-label, randomised, phase 3 trial. Lancet. (2022) 399:2294–308. doi: 10.1016/S0140-6736(22)00662-6
49. Fowler NH, Dickinson M, Dreyling M, Martinez-Lopez J, Kolstad A, Butler J, et al. Tisagenlecleucel in adult relapsed or refractory follicular lymphoma: the phase 2 ELARA trial. Nat Med. (2022) 28:325–32. doi: 10.1038/s41591-021-01622-0
50. Locke FL, Miklos DB, Jacobson CA, Perales MA, Kersten MJ, Oluwole OO, et al. Axicabtagene ciloleucel as second-line therapy for large B-cell lymphoma. N Engl J Med. (2022) 386:640–54. doi: 10.1056/NEJMoa2116133
51. Goyco Vera D, Waghela H, Nuh M, Pan J, and Lulla P. Approved CAR-T therapies have reproducible efficacy and safety in clinical practice. Hum Vaccin Immunother. (2024) 20:2378543. doi: 10.1056/NEJMoa2116133
52. Qasim W, Zhan H, Samarasinghe S, Adams S, Amrolia P, Stafford S, et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci Trans medicine. (2017) 9:eaaj2013. doi: 10.1126/scitranslmed.aaj2013
53. Qasim W. Genome-edited allogeneic donor “universal” chimeric antigen receptor T cells. Blood. (2023) 141:835–45. doi: 10.1182/blood.2022016204
54. Qasim W. Allogeneic CAR T cell therapies for leukemia. Am J Hematol. (2019) 94:S50–s4. doi: 10.1002/ajh.v94.S1
55. Hu B, Ren J, Luo Y, Keith B, Young RM, Scholler J, et al. Augmentation of antitumor immunity by human and mouse CAR T cells secreting IL-18. Cell reports. (2017) 20:3025–33.
56. Svoboda J, Gerson JN, Landsburg DJ, Chong EA, Barta SK, Dwivedy Nasta S, et al. Interleukin-18 secreting autologous anti-CD19 CAR T-cells (huCART19-IL18) in patients with non-Hodgkin lymphomas relapsed or refractory to prior CAR T-cell therapy. Blood. (. 2022) 140:4612–4. doi: 10.1182/blood-2022-162393
57. MacLeod DT, Antony J, Martin AJ, Moser RJ, Hekele A, Wetzel KJ, et al. Integration of a CD19 CAR into the TCR alpha chain locus streamlines production of allogeneic gene-edited CAR T cells. Mol Ther. (2017) 25:949–61. doi: 10.1016/j.ymthe.2017.02.005
58. Benjamin R, Jain N, Maus MV, Boissel N, Graham C, Jozwik A, et al. UCART19, a first-in-class allogeneic anti-CD19 chimeric antigen receptor T-cell therapy for adults with relapsed or refractory B-cell acute lymphoblastic leukaemia (CALM): a phase 1, dose-escalation trial. Lancet Haematology. (2022) 9:e833–e43. doi: 10.1016/S2352-3026(22)00245-9
59. Benjamin R, Graham C, Yallop D, Jozwik A, Mirci-Danicar OC, Lucchini G, et al. Genome-edited, donor-derived allogeneic anti-CD19 chimeric antigen receptor T cells in paediatric and adult B-cell acute lymphoblastic leukaemia: results of two phase 1 studies. Lancet. (2020) 396:1885–94. doi: 10.1016/S0140-6736(20)32334-5
60. Mailankody S, Matous JV, Chhabra S, Liedtke M, Sidana S, Oluwole OO, et al. Allogeneic BCMA-targeting CAR T cells in relapsed/refractory multiple myeloma: phase 1 UNIVERSAL trial interim results. Nat medicine. (2023) 29:422–9. doi: 10.1038/s41591-022-02182-7
61. Brown CE, Rodriguez A, Palmer J, Ostberg JR, Naranjo A, Wagner JR, et al. Off-the-shelf, steroid-resistant, IL13Rα2-specific CAR T cells for treatment of glioblastoma. Neuro-oncology. (2022) 24:1318–30. doi: 10.1038/s41591-022-02163-w
62. Hu Y, Zhou Y, Zhang M, Zhao H, Wei G, Ge W, et al. Genetically modified CD7-targeting allogeneic CAR-T cell therapy with enhanced efficacy for relapsed/refractory CD7-positive hematological Malignancies: a phase I clinical study. Cell Res. (2022) 32:995–1007. doi: 10.1038/s41422-022-00721-y
63. Fu Y, Foden JA, Khayter C, Maeder ML, Reyon D, Joung JK, et al. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat Biotechnol. (2013) 31:822–6. doi: 10.1038/nbt.2623
64. Diorio C, Murray R, Naniong M, Barrera L, Camblin A, Chukinas J, et al. Cytosine base editing enables quadruple-edited allogeneic CART cells for T-ALL. Blood J Am Soc Hematology. (2022) 140:619–29. doi: 10.1007/s11899-017-0402-1
65. Marcaida MJ, Muñoz IG, Blanco FJ, Prieto J, and Montoya G. Homing endonucleases: from basics to therapeutic applications. Cell Mol Life Sci. (2010) 67:727–48. doi: 10.1007/s00018-009-0188-y
66. Murty T and Mackall CL. Gene editing to enhance the efficacy of cancer cell therapies. Mol Ther. (2021) 29:3153–62. doi: 10.1016/j.ymthe.2021.10.001
67. Diorio C, Teachey DT, and Grupp SA. Allogeneic chimeric antigen receptor cell therapies for cancer: progress made and remaining roadblocks. Nat Rev Clin Oncol. (. 2024), 1–18. doi: 10.1038/s41571-024-00959-y
68. Singh N, Shi J, June CH, and Ruella M. Genome-editing technologies in adoptive T cell immunotherapy for cancer. Curr hematologic Malignancy reports. (2017) 12:522–9. doi: 10.1038/s41571-024-00959-y
69. Provasi E, Genovese P, Lombardo A, Magnani Z, Liu P-Q, Reik A, et al. Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer. Nat medicine. (2012) 18:807–15. doi: 10.1007/s11899-017-0417-7
70. Pattanayak V, Ramirez CL, Joung JK, and Liu DR. Revealing off-target cleavage specificities of zinc-finger nucleases by in vitro selection. Nat Methods. (2011) 8:765–70. doi: 10.1038/nmeth.1670
71. Gautron A-S, Juillerat A, Guyot V, Filhol J-M, Dessez E, Duclert A, et al. Fine and predictab le tuning of TALEN gene editing targeting for improved T cell adoptive immunotherapy. Mol Therapy-Nucleic Acids. (2017) 9:312–21. doi: 10.1016/j.omtn.2017.10.005
72. Poirot L, Philip B, Schiffer-Mannioui C, Le Clerre D, Chion-Sotinel I, Derniame S, et al. Multiplex genome-edited T-cell manufacturing platform for “off-the-shelf” adoptive T-cell immunotherapies. Cancer Res. (2015) 75:3853–64. doi: 10.1158/0008-5472.CAN-14-3321
73. Gupta RM and Musunuru K. Expanding the genetic editing tool kit: ZFNs, TALENs, and CRISPR-Cas9. J Clin investigation. (2014) 124:4154–61. doi: 10.1158/0008-5472.CAN-14-3321
74. Adli M. The CRISPR tool kit for genome editing and beyond. Nat communications. (2018) 9:1911. doi: 10.1172/JCI72992
75. Ottaviano G, Georgiadis C, Gkazi SA, Syed F, Zhan H, Etuk A, et al. Phase 1 clinical trial of CRISPR-engineered CAR19 universal T cells for treatment of children with refractory B cell leukemia. Sci Trans medicine. (2022) 14:eabq3010. doi: 10.1126/scitranslmed.abq3010
76. Guo Y, Tong C, Su L, Zhang W, Jia H, Liu Y, et al. CRISPR/Cas9 genome-edited universal CAR T cells in patients with relapsed/refractory lymphoma. Blood Advances. (2022) 6:2695–9. doi: 10.1038/s41576-018-0059-1
77. Li S, Wang X, Liu L, Liu J, Rao J, Yuan Z, et al. CD7 targeted “off-the-shelf” CAR-T demonstrates robust in vivo expansion and high efficacy in the treatment of patients with relapsed and refractory T cell Malignancies. Leukemia. (2023) 37:2176–86. doi: 10.1038/s41375-023-02018-4
78. Rees HA and Liu DR. Base editing: precision chemistry on the genome and transcriptome of living cells. Nat Rev Genet. (2018) 19:770–88. doi: 10.1038/s41576-018-0059-1
79. Chiesa R, Georgiadis C, Syed F, Zhan H, Etuk A, Gkazi SA, et al. Base-edited CAR7 T cells for relapsed T-cell acute lymphoblastic leukemia. New Engl J Medicine. (2023) 389:899–910. doi: 10.1038/s41576-018-0059-1
80. Graham C, Jozwik A, Pepper A, and Benjamin R. Allogeneic CAR-T cells: more than ease of access? Cells. (2018) 7:155. doi: 10.3390/cells7100155
81. Prenen H, Dekervel J, Hendlisz A, Anguille S, Ahmad A, Cerf E, et al. Updated data from the alloSHRINK phase 1 first-in-Human study evaluating CYAD-101, an innovative non-Gene-Edited allogeneic CAR-T, in metastatic colorectal cancer. J Clin Oncol. (2021) 39:74. doi: 10.1200/JCO.2021.39.3_suppl.74
82. Lonez C, Bolsee J, Huberty F, Nguyen T, Jacques-Hespel C, Demoulin B, et al. 285 Proof-of-concept of a non-gene editing technology using shRNA down-regulation to engineer and optimize CAR T-cell functionality. BMJ Specialist Journals;. (2023). doi: 10.1136/jitc-2023-SITC2023.0285
83. Wang X, Xue L, Li S, Fan Q, Liu K, Jin R, et al. S264: preliminary analyses of A non-gene-editing allogentic car-T in cd19+ Relapsed or refractory non-hodgin’s lymphoma. HemaSphere. (2022) 6:165–6. doi: 10.1097/01.HS9.0000843948.58401.85
84. Liu YP, Haasnoot J, ter Brake O, Berkhout B, and Konstantinova P. Inhibition of HIV-1 by multiple siRNAs expressed from a single microRNA polycistron. Nucleic Acids Res. (2008) 36:2811–24. doi: 10.1093/nar/gkn109
85. Choi J-G, Bharaj P, Abraham S, Ma H, Yi G, Ye C, et al. Multiplexing seven miRNA-Based shRNAs to suppress HIV replication. Mol Ther. (2015) 23:310–20. doi: 10.1038/mt.2014.205
86. Wang T, Xie Y, Tan A, Li S, and Xie Z. Construction and characterization of a synthetic MicroRNA cluster for multiplex RNA interference in mammalian cells. ACS Synthetic Biol. (2016) 5:1193–200. doi: 10.1021/acssynbio.5b00180
87. Lonez C and Breman E. Allogeneic CAR-T therapy technologies: has the promise been met? Cells. (2024) 13:146. doi: 10.3390/cells13020146
88. Neelapu SS, Nath R, Munoz J, Tees M, Miklos DB, Frank MJ, et al. ALPHA study: ALLO-501 produced deep and durable responses in patients with relapsed/refractory non-Hodgkin’s lymphoma comparable to autologous CAR T. Blood. (2021) 138:3878. doi: 10.1182/blood-2021-146038
89. Lekakis LJ, Locke FL, Tees M, Neelapu SS, Malik SA, Hamadani M, et al. ALPHA2 study: ALLO-501A allogeneic CAR T in LBCL, updated results continue to show encouraging safety and efficacy with consolidation dosing. Blood. (2021) 138:649. doi: 10.1182/blood-2021-146045
90. Srour S, Kotecha R, Curti B, Chahoud J, Drakaki A, Tang L, et al. Abstract CT011: a phase 1 multicenter study (TRAVERSE) evaluating the safety and efficacy of ALLO-316 following conditioning regimen in pts with advanced or metastatic clear cell renal cell carcinoma (ccRCC). Cancer Res. (2023) 83:CT011–CT. doi: 10.1158/1538-7445.AM2023-CT011
91. Boissel N, Chevallier P, Curran K, Schiller G, Liu H, Larson R, et al. P1408: updated results of the phase I BALLI-01 trial of UCART22, an anti-CD22 allogeneic CAR-t cell product, in patients with relapsed or refractory (r/r) CD22+ B-cell acute lymphoblastic leukemia (B-ALL). HemaSphere. (2023) 7:e323373f. doi: 10.1097/01.HS9.0000972520.32337.3f
92. Sallman DA, DeAngelo DJ, Pemmaraju N, Dinner S, Gill S, Olin RL, et al. Ameli-01: a phase I trial of UCART123v1. 2, an anti-CD123 allogeneic CAR-T cell product, in adult patients with relapsed or refractory (R/R) CD123+ acute myeloid leukemia (AML). Blood. (2022) 140:2371–3. doi: 10.1182/blood-2022-169928
93. Cellectis Cellectis to Showcase Clinical Data from AMELI-01 and Preclinical Data from UCARTCS1 at ASH (2022). Available online at: https://www.cellectis.com/en/press/cellectis-to-showcase-clinical-data-from-ameli-01-and-preclinical-data-fromucartcs1-at-ash-2022/ (Accessed November 22, 2023).
94. Precision bioSciences provides update on allogeneic CAR T programs and regulatory path forward. In: Precision BioSciences I, editor. Precision BioSciences Provides Update on Allogeneic CAR T Programs and Regulatory Path Forward. Precision BioSciences, Inc.;. Available at: https://investorprecisionbiosciencescom/node/9926/pdf.
95. McGuirk JP, Tam CS, Kröger N, Riedell PA, Murthy HS, Ho PJ, et al. CTX110 allogeneic CRISPR-Cas9-engineered CAR T cells in patients (pts) with relapsed or refractory (R/R) large B-cell lymphoma (LBCL): results from the phase 1 dose escalation carbon study. Blood. (2022) 140:10303–6. doi: 10.1182/blood-2022-166432
96. CRISPR therapeutics corporate presentation (2022). Available online at: https://crisprtx.gcs-web.com/static-files/5c256eb5-4982-4e47-b463-79eff954e622 (Accessed November 23, 2023).
97. Pal S, Tran B, Haanen J, Hurwitz M, Sacher A, Agarwal N, et al. 558 CTX130 allogeneic CRISPR-Cas9–engineered chimeric antigen receptor (CAR) T cells in patients with advanced clear cell renal cell carcinoma: results from the phase 1 COBALT-RCC study. BMJ Specialist Journals;. (2022). doi: 10.1136/jitc-2022-SITC2022.0558
98. Iyer SP, Sica RA, Ho PJ, Hu B, Zain J, Prica A, et al. S262: The COBALT-LYM study of CTX130: A phase 1 dose escalation study of CD70-targeted allogeneic CRISPR-CAS9–engineered CAR T cells in patients with relapsed/refractory (R/R) T-cell Malignancies. HemaSphere. (2022) 6:163–4. doi: 10.1097/01.HS9.0000843940.96598.e2
99. Caribou biosciences corporate presentation (2023). Available online at: https://investor.cariboubio.com/static-files/3168b9f6-48dd-4e44-8103-df5ff9013b6b. (Accessed June 4, 2025).
100. Hu Y, Zhou Y, Zhang M, Ge W, Li Y, Yang L, et al. CRISPR/Cas9-engineered universal CD19/CD22 dual-targeted CAR-T cell therapy for relapsed/refractory B-cell acute lymphoblastic leukemia. Clin Cancer Res. (2021) 27:2764–72. doi: 10.1158/1078-0432.CCR-20-3863
101. Dholaria B, Kocoglu MH, Kin A, Asch AS, Ramakrishnan A, Bachier C, et al. Early safety results of P-BCMA-ALLO1, a fully allogeneic chimeric antigen receptor T-cell (CAR-T), in patients with relapsed/refractory multiple myeloma (RRMM). Blood. (2023) 142:3479. doi: 10.1182/blood-2023-182430
102. Oh D, Henry J, Baranda J, Dumbrava E, Cohen E, Eskew J, et al. 46P Development of an allogeneic CAR-T targeting MUC1-C (MUC1, cell surface associated, C-terminal) for epithelial derived tumors. Immuno-Oncology Technol. (2022) 16. doi: 10.1016/j.iotech.2022.100151
103. Prenen H, Dekervel J, Anguille S, Hendlisz A, Michaux A, Sotiropoulou PA, et al. CYAD-101: An innovative non-gene edited allogeneic CAR-T for solid tumor cancer therapy. Liver. (2020) 8:57.1. doi: 10.1200/JCO.2020.38.15_suppl.3032
104. Al-Homsi A-S, Anguille S, Deeren D, Nishihori T, Meuleman N, Abdul-Hay M, et al. Immunicy-1: targeting BCMA with cyad-211 to establish proof of concept of an shRNA-based allogeneic CAR T cell therapy platform. Blood. (2021) 138:2817. doi: 10.1182/blood-2021-147738
105. Mehta A, Farooq U, Chen A, McGuirk JP, Ly T, Wong L, et al. Interim phase I clinical data of FT819-101, a study of the first-ever, off-the-shelf, iPSC-derived TCR-less CD19 CAR T-cell therapy for patients with relapsed/refractory B-cell Malignancies. Blood. (2022) 140:4577–8. doi: 10.1182/blood-2022-167194
106. Sommer C, Boldajipour B, Kuo TC, Bentley T, Sutton J, Chen A, et al. Preclinical evaluation of allogeneic CAR T cells targeting BCMA for the treatment of multiple myeloma. Mol Ther. (2019) 27:1126–38. doi: 10.1016/j.ymthe.2019.04.001
107. Ren J, Liu X, Fang C, Jiang S, June CH, and Zhao Y. Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition. Clin Cancer Res. (2017) 23:2255–66. doi: 10.1158/1078-0432.CCR-16-1300
108. Georgiadis C, Preece R, Nickolay L, Etuk A, Petrova A, Ladon D, et al. Long terminal repeat CRISPR-CAR-coupled “Universal” T cells mediate potent anti-leukemic effects. Mol Ther. (2018) 26:1215–27. doi: 10.1016/j.ymthe.2018.02.025
109. Dupouy S, Marchiq I, Derippe T, Almena-Carrasco M, Jozwik A, Fouliard S, et al. Clinical pharmacology and determinants of response to UCART19, an allogeneic anti-CD19 CAR-T cell product, in adult B-cell acute lymphoblastic leukemia. Cancer Res Commun. (2022) 2:1520–31. doi: 10.1158/2767-9764.CRC-22-0175
110. Pal SK, Tran B, Haanen J, Hurwitz ME, Sacher A, Tannir NM, et al. CD70-targeted allogeneic CAR T-cell therapy for advanced clear cell renal cell carcinoma. Cancer Discov. (2024) 14:1176–89. doi: 10.1158/2159-8290.CD-24-0102
Keywords: allogeneic CAR T cells, editing technology, non-editing technology, off-the-shelf CART cell, graft-versus-host disease
Citation: Shokati A, Sanjari-Pour M, Akhavan Rahnama M, Hoseinzadeh S, Vaezi M and Ahmadvand M (2025) Allogeneic CART progress: platforms, current progress and limitations. Front. Immunol. 16:1557157. doi: 10.3389/fimmu.2025.1557157
Received: 08 January 2025; Accepted: 22 May 2025;
Published: 12 June 2025.
Edited by:
Peter Hamar, Semmelweis University, HungaryReviewed by:
Antonio Di Stasi, University of Alabama at Birmingham, United StatesMaurizio Chiriva-Internati, University of Texas MD Anderson Cancer Center, United States
Copyright © 2025 Shokati, Sanjari-Pour, Akhavan Rahnama, Hoseinzadeh, Vaezi and Ahmadvand. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Mohammad Ahmadvand, bWFobWFkdmFuZEBzaW5hLnR1bXMuYWMuaXI=; Mahshid Akhavan Rahnama, TWFoc2hpZGFraGF2YW42OUB5YWhvby5jb20=
†These authors have contributed equally to this work and share first authorship