Appearance
This report is written by MaltSci based on the latest literature and research findings
How does CAR-T therapy work?
Abstract
Chimeric Antigen Receptor T-cell (CAR-T) therapy has revolutionized immunotherapy, particularly for hematological malignancies like acute lymphoblastic leukemia (ALL) and certain non-Hodgkin lymphomas. This innovative approach involves the genetic modification of a patient's T cells to express chimeric antigen receptors (CARs) that specifically recognize tumor-associated antigens. Upon reinfusion, these engineered T cells proliferate and exert potent anti-tumor effects. Since the FDA's approval in 2017, CAR-T therapy has demonstrated remarkable efficacy, inducing remission in patients with previously resistant cancers. However, challenges such as severe side effects, including cytokine release syndrome (CRS) and neurotoxicity, necessitate careful management. Current research focuses on several key areas: the mechanism of CAR-T cell action, the engineering processes for T cell modification, and the clinical applications and challenges in treating solid tumors. Ongoing investigations aim to enhance CAR design, explore combination therapies, and improve patient outcomes. Understanding the intricacies of CAR-T therapy is crucial for advancing personalized cancer treatment strategies and expanding its therapeutic potential.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Mechanism of Action of CAR-T Cells
- 2.1 Design and Structure of CARs
- 2.2 Activation and Proliferation of CAR-T Cells
- 3 Engineering T Cells for CAR-T Therapy
- 3.1 T Cell Collection and Genetic Modification
- 3.2 Types of Viral Vectors Used
- 4 Clinical Applications of CAR-T Therapy
- 4.1 Success in Hematological Malignancies
- 4.2 Challenges in Solid Tumors
- 5 Adverse Effects and Management Strategies
- 5.1 Cytokine Release Syndrome (CRS)
- 5.2 Neurotoxicity and Other Side Effects
- 6 Future Directions in CAR-T Therapy
- 6.1 Enhancements in CAR Design
- 6.2 Potential for Combination Therapies
- 7 Summary
1 Introduction
Chimeric Antigen Receptor T-cell (CAR-T) therapy has emerged as a revolutionary advancement in the field of immunotherapy, particularly in the treatment of hematological malignancies such as acute lymphoblastic leukemia (ALL) and certain types of non-Hodgkin lymphoma. This innovative therapeutic approach involves the genetic modification of a patient's own T cells to express chimeric antigen receptors (CARs) that specifically recognize and bind to antigens present on the surface of cancer cells. Following this genetic engineering, the modified T cells are reinfused into the patient, where they can proliferate and exert potent anti-tumor effects. The significance of CAR-T therapy is underscored by its ability to induce remission in patients with previously treatment-resistant cancers, thereby offering new hope for those facing dire prognoses [1][2].
The development of CAR-T therapy has been marked by significant milestones since its initial FDA approval in 2017, with ongoing research expanding its applications beyond hematological malignancies to solid tumors [1]. Despite its promising outcomes, CAR-T therapy is not without challenges. Notably, patients may experience severe side effects, including cytokine release syndrome (CRS) and neurotoxicity, which necessitate careful management and ongoing research to optimize safety and efficacy [3][4]. Understanding the intricate mechanisms of CAR-T cell action, the engineering processes involved, and the clinical implications of these therapies is crucial for advancing personalized cancer treatment strategies.
Current research on CAR-T therapy is rapidly evolving, with significant attention focused on several key areas. The first area is the mechanism of action of CAR-T cells, which encompasses the design and structure of CARs and the subsequent activation and proliferation of CAR-T cells [5]. The second area involves the engineering of T cells for CAR-T therapy, detailing the processes of T cell collection and genetic modification, as well as the types of viral vectors employed in these procedures [6]. The third area highlights the clinical applications of CAR-T therapy, particularly its success in treating hematological malignancies and the challenges it faces in solid tumors [2][3].
Moreover, the adverse effects associated with CAR-T therapy and the strategies for their management represent another critical area of investigation. Key concerns include CRS and neurotoxicity, which require effective monitoring and intervention strategies [7][8]. Lastly, future directions in CAR-T therapy are also being explored, focusing on enhancements in CAR design and the potential for combination therapies that could improve patient outcomes and broaden the therapeutic scope of CAR-T approaches [7][9].
This review aims to provide a comprehensive overview of how CAR-T therapy works, detailing the underlying biological mechanisms, the technological innovations involved, current clinical applications, and future research directions. By synthesizing the latest findings and advancements in the field, we hope to contribute to a deeper understanding of CAR-T therapy and its potential to transform cancer treatment.
2 Mechanism of Action of CAR-T Cells
2.1 Design and Structure of CARs
Chimeric antigen receptor (CAR) T cell therapy is an innovative approach in cancer immunotherapy that utilizes genetically engineered T cells to target and eliminate cancer cells. The mechanism of action of CAR-T cells involves several key components and steps, which can be understood through the design and structure of CARs.
The CAR is composed of three primary structural elements: an extracellular domain, a transmembrane domain, and a cytoplasmic domain. The extracellular domain is responsible for recognizing and binding to specific antigens expressed on the surface of target cells, typically tumor cells. This domain is often derived from the single-chain variable fragment (scFv) of a monoclonal antibody or, increasingly, from nanobodies, which are smaller antibody fragments with high affinity and stability [10]. The transmembrane domain anchors the CAR in the T cell membrane, while the cytoplasmic domain is responsible for initiating intracellular signaling upon antigen recognition.
When CAR-T cells encounter their target antigen, the binding of the CAR's extracellular domain to the antigen triggers a cascade of intracellular signaling events through the cytoplasmic domain. This activation leads to T cell proliferation, cytokine release, and ultimately the destruction of the antigen-expressing target cells. The efficacy of CAR-T cells is enhanced by the inclusion of co-stimulatory signaling domains within the cytoplasmic region, which can augment T cell activation and persistence [2].
CAR-T cells are engineered by transducing T cells with a viral vector that carries the gene encoding the CAR. This process can involve various generations of CARs, each defined by the number and type of signaling domains included. First-generation CARs contained only the signaling domain from the T cell receptor, while second-generation CARs included additional co-stimulatory domains (such as CD28 or 4-1BB), and third-generation CARs incorporated multiple co-stimulatory signals to further enhance T cell activation and survival [11].
Recent advances have seen the development of CARs utilizing nanobodies as the antigen-binding domain. These nanobody-based CAR-T cells have shown comparable or even superior functionality compared to traditional scFv-based CARs in various preclinical and clinical settings, due to their smaller size, which allows for better tissue penetration and a more flexible design [12].
Overall, CAR-T therapy represents a significant advancement in the treatment of cancers, particularly hematological malignancies, by leveraging the specificity of CARs to direct T cells against cancer cells effectively. The ongoing evolution of CAR design continues to enhance the therapeutic potential of this innovative approach, with a focus on improving safety, efficacy, and the ability to target a broader range of malignancies [7].
2.2 Activation and Proliferation of CAR-T Cells
Chimeric Antigen Receptor T (CAR-T) cell therapy represents a transformative approach in cancer treatment, particularly for hematological malignancies. The mechanism of action of CAR-T cells involves several critical steps: T cell engineering, activation, proliferation, and execution of anti-tumor functions.
Initially, T cells are extracted from a patient's blood and genetically modified to express CARs that target specific tumor antigens. This genetic modification allows CAR-T cells to recognize and bind to tumor cells in an MHC-independent manner, which is a significant advantage over traditional T cell receptors. Upon encountering their specific antigen on tumor cells, CAR-T cells undergo a series of activation signals.
The recognition of tumor antigens by CARs leads to the propagation of T cell activation signals, which include a co-stimulatory signal essential for effective CAR-T cell activation. The inclusion of co-stimulatory domains, such as CD28 or 4-1BB, has been pivotal in enhancing the efficacy of CAR-T cells. Early designs lacking these co-stimulatory signals demonstrated limited success, but modern CAR-T therapies universally incorporate these domains to improve activation, proliferation, and functional longevity of the T cells (Honikel and Olejniczak 2022) [5].
Following activation, CAR-T cells proliferate significantly, which is crucial for establishing an effective immune response against the tumor. Studies have shown that co-stimulatory signals are integral not only for T cell activation but also for their persistence and durability in the patient's body. The absence of appropriate co-stimulation can lead to poor persistence and functionality of CAR-T cells, resulting in diminished anti-tumor efficacy (Lainšček et al. 2023) [13].
Moreover, CAR-T cells can be designed to include additional regulatory elements that modulate their activation and proliferation. For instance, engineered systems that utilize downstream transcription factors allow for precise control over CAR-T cell activation, enabling transient or sustained proliferation based on the tumor environment (Lainšček et al. 2023) [13].
The proliferation of CAR-T cells is further influenced by the conditions under which they are expanded ex vivo. Innovative protocols, such as the use of antigen-specific stimulation via membrane vesicles, have been developed to enhance the expansion and functional activity of CAR-T cells. These methods aim to produce populations of CAR-T cells that are not only more numerous but also exhibit superior anti-tumor functions (Ukrainskaya et al. 2021) [14].
In summary, CAR-T therapy operates through a well-orchestrated mechanism involving the genetic modification of T cells to express CARs, subsequent activation via antigen recognition and co-stimulatory signals, and robust proliferation that culminates in effective tumor cell targeting and destruction. The ongoing advancements in CAR-T cell engineering and expansion techniques continue to enhance the efficacy and safety of this promising therapeutic modality.
3 Engineering T Cells for CAR-T Therapy
3.1 T Cell Collection and Genetic Modification
Chimeric Antigen Receptor T-cell (CAR-T) therapy represents a cutting-edge approach in cancer immunotherapy, particularly effective against hematological malignancies. The process of CAR-T therapy involves several critical steps, including the collection of T cells from the patient, their genetic modification, and subsequent reinfusion.
Initially, T cells are harvested from the patient’s peripheral blood through a procedure known as leukapheresis. This process involves separating the T cells from other blood components, allowing for a concentrated collection of the patient's own immune cells. The isolated T cells, specifically CD4+ and CD8+ T cells, are then subjected to genetic modification. This is typically achieved through viral transduction, where genes encoding chimeric antigen receptors (CARs) are introduced into the T cells. These CARs are designed to recognize specific tumor antigens, thus enabling the modified T cells to target and eliminate cancer cells effectively.
The CAR is composed of several key domains: an extracellular antigen recognition domain, often derived from a single-chain variable fragment (scFv) of an antibody, which binds to a specific tumor-associated antigen; a transmembrane domain that anchors the CAR in the T cell membrane; and one or more intracellular signaling domains that initiate T cell activation upon antigen recognition. The inclusion of co-stimulatory signaling domains, such as CD28 or 4-1BB, is crucial for enhancing T cell activation, proliferation, and persistence. This design evolved following early CAR-T constructs that lacked these co-stimulatory signals, which exhibited limited efficacy [5].
Once the T cells are genetically modified to express the CAR, they undergo an expansion phase in the laboratory to increase their numbers. This step is essential to produce a sufficient quantity of CAR-T cells for therapeutic use. Following this expansion, the CAR-T cells are reinfused back into the patient. Upon reinfusion, these engineered T cells seek out and bind to the target tumor cells that express the corresponding antigen, leading to the activation of T cell responses that can result in the destruction of malignant cells [2].
In summary, CAR-T therapy operates by reprogramming a patient’s own T cells to enhance their ability to identify and eliminate cancer cells. The entire process—from T cell collection to genetic modification and reinfusion—highlights the innovative use of synthetic biology in developing personalized cancer treatments, demonstrating significant promise particularly in hematological cancers such as acute lymphoblastic leukemia and certain types of lymphoma [15].
3.2 Types of Viral Vectors Used
Chimeric antigen receptor (CAR) T-cell therapy is an innovative immunotherapeutic approach that involves the genetic engineering of a patient's T cells to express chimeric antigen receptors (CARs). These CARs are designed to recognize specific tumor-associated antigens on cancer cells, enabling the modified T cells to selectively target and eliminate malignant cells. The fundamental process of CAR-T therapy begins with the extraction of T cells from the patient's blood, followed by their modification using viral vectors, which serve as vehicles for introducing the CAR genes into the T cells. After successful genetic modification, the CAR-T cells are expanded in the laboratory and subsequently reinfused into the patient.
The engineering of T cells for CAR-T therapy typically utilizes viral vectors such as lentiviruses and retroviruses. These vectors are employed due to their ability to integrate the CAR gene into the host T cell genome, ensuring stable expression of the CAR. Lentiviral vectors are particularly favored for their capacity to transduce non-dividing cells and for providing long-term expression of the CAR, which is critical for sustained T cell activity against tumors. Retroviral vectors, while also effective, generally require the target cells to be dividing for successful gene transfer.
The CAR constructs themselves can vary in design, often comprising an extracellular domain that binds to the target antigen, a transmembrane domain that anchors the CAR in the T cell membrane, and an intracellular signaling domain that activates T cell effector functions upon antigen recognition. This unique structure allows CAR-T cells to bypass the need for major histocompatibility complex (MHC) antigen presentation, which is a significant advantage in targeting tumor cells that may downregulate MHC expression to evade immune detection.
The therapeutic efficacy of CAR-T therapy has been prominently demonstrated in hematological malignancies, where it has led to remarkable outcomes. However, the application of CAR-T therapy in solid tumors presents additional challenges due to factors such as the immunosuppressive tumor microenvironment and the heterogeneity of tumor antigens. Ongoing research aims to enhance the effectiveness of CAR-T cells in these contexts, including the exploration of next-generation CAR designs and combination therapies that may improve tumor targeting and overcome the limitations of current CAR-T technologies[1][16][17].
4 Clinical Applications of CAR-T Therapy
4.1 Success in Hematological Malignancies
Chimeric antigen receptor T-cell (CAR-T) therapy represents a transformative approach in the treatment of hematological malignancies, leveraging the body’s immune system to target and eliminate cancer cells. This innovative therapy involves the genetic modification of a patient’s T-cells to express a synthetic receptor known as a chimeric antigen receptor (CAR), which specifically recognizes antigens present on the surface of tumor cells. The primary mechanism of action involves the extraction of T-cells from the patient, their subsequent engineering to express CARs, and the reinfusion of these modified cells back into the patient, where they actively seek out and destroy cancer cells.
The clinical application of CAR-T therapy has been particularly successful in treating hematological malignancies such as acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL), and various types of non-Hodgkin lymphoma. The inaugural approval of CAR-T therapy occurred in 2017, specifically for relapsed/refractory ALL, marking a significant milestone in cancer treatment. Since then, various CAR-T products targeting specific antigens, such as CD19 and B-cell maturation antigen (BCMA), have received approval, showcasing remarkable efficacy and leading to high response rates among patients. For instance, FDA-approved CAR-T therapies targeting CD19 are now available for both B-cell acute lymphoblastic leukemia and low- and high-grade B-cell non-Hodgkin lymphoma, while therapies targeting BCMA have been developed for multiple myeloma [18][19].
The success of CAR-T therapy in hematological malignancies can be attributed to several factors. Firstly, the identification of specific tumor-associated antigens, such as CD19 and BCMA, allows for targeted destruction of malignant cells while sparing normal cells. This specificity reduces collateral damage and enhances the therapeutic index of the treatment. Secondly, CAR-T cells can proliferate and persist long-term in the patient’s body, potentially leading to durable remissions and long-term antitumor immunity [20][21].
Despite the promising results, the application of CAR-T therapy is not without challenges. Adverse effects, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), have been reported, which necessitate careful monitoring and management during treatment [9][22]. Furthermore, issues related to therapy resistance, disease relapse, and minimal residual disease (MRD) persistence post-remission remain significant hurdles that require ongoing research and innovative solutions [23][24].
In summary, CAR-T therapy has revolutionized the treatment landscape for hematological malignancies by providing a targeted, effective approach that utilizes the body’s immune system. The high response rates and the potential for long-term remission in patients underscore the clinical success of this therapy, while ongoing research aims to address the associated challenges and expand its application to a broader range of malignancies.
4.2 Challenges in Solid Tumors
Chimeric antigen receptor T-cell (CAR-T) therapy is an innovative immunotherapeutic approach that utilizes genetically modified T-cells to target and eliminate cancer cells. The fundamental mechanism involves extracting T-cells from a patient’s blood, engineering them to express chimeric antigen receptors that recognize specific antigens present on the surface of tumor cells, and subsequently reinfusing these modified T-cells back into the patient’s body. Once reintroduced, these CAR-T cells are designed to seek out and destroy cancer cells, thus providing a potent therapeutic strategy against malignancies.
Initially, CAR-T therapy demonstrated remarkable efficacy in hematological malignancies, such as certain types of leukemia and lymphoma. However, its application in solid tumors has proven to be significantly more challenging. Solid tumors present unique obstacles that hinder the effectiveness of CAR-T therapy. These challenges include tumor antigen heterogeneity, which complicates the identification of universally expressed targets, and the immunosuppressive tumor microenvironment (TME), which can inhibit T-cell function and proliferation. Furthermore, physical barriers such as dense extracellular matrices and abnormal vasculature restrict T-cell infiltration into tumor tissues [25][26][27].
The efficacy of CAR-T therapy in solid tumors is also limited by the presence of suppressive cytokines and regulatory immune cells within the TME, which contribute to immune evasion and T-cell exhaustion [28][29]. Additionally, the relatively low persistence of CAR-T cells in vivo poses a significant challenge, as effective tumor eradication often requires sustained T-cell activity [30][31].
To address these challenges, researchers are exploring various optimization strategies. These include the development of multi-antigen targeting constructs to enhance specificity, the engineering of armored CAR-T cells capable of resisting immunosuppressive signals, and the incorporation of matrix-degrading enzymes to facilitate T-cell access to tumor sites [25][26]. Combination therapies, such as pairing CAR-T therapy with immune checkpoint inhibitors or oncolytic viruses, are also being investigated to improve therapeutic outcomes [27][32].
Clinical trials are actively assessing the application of CAR-T therapy across various solid tumors, including lung, breast, and colorectal cancers, with the goal of expanding its therapeutic horizons and providing new treatment options for patients facing these challenging malignancies [1][26][28]. Despite the current limitations, ongoing research continues to unveil innovative strategies that may ultimately lead to successful CAR-T therapy applications in solid tumors, thus revolutionizing cancer treatment [30][31].
5 Adverse Effects and Management Strategies
5.1 Cytokine Release Syndrome (CRS)
Chimeric antigen receptor T-cell (CAR-T) therapy is a groundbreaking immunotherapeutic approach that harnesses the power of genetically engineered T cells to target and eliminate cancer cells. While this therapy has shown remarkable efficacy, particularly in treating relapsed or refractory hematological malignancies, it is also associated with significant adverse effects, the most notable being cytokine release syndrome (CRS).
CRS occurs when CAR-T cells become activated and proliferate in response to tumor antigens, leading to an excessive release of pro-inflammatory cytokines into the bloodstream. This phenomenon can result in a wide range of clinical symptoms, ranging from mild fever and fatigue to severe manifestations such as hypotension, hypoxia, and multi-organ dysfunction, which can be life-threatening [33][34].
The pathophysiology of CRS is complex and involves the activation of various immune cell types, including T cells, monocytes, and neutrophils. Upon CAR-T cell activation, there is a rapid surge in cytokine levels, including interleukin-6 (IL-6), interferon-gamma (IFN-γ), and tumor necrosis factor-alpha (TNF-α), among others [35][36]. The severity of CRS can vary significantly among patients, influenced by factors such as the tumor burden, the lymphodepletion regimen prior to CAR-T infusion, and the dose of CAR-T cells administered [37].
Management strategies for CRS are critical to ensure patient safety and the continued efficacy of CAR-T therapy. Tocilizumab, an IL-6 receptor antagonist, is commonly employed to mitigate CRS symptoms, particularly in cases of severe CRS [33][38]. The timing of tocilizumab administration is crucial, although optimal protocols are still being refined [39]. In addition to tocilizumab, corticosteroids may also be utilized to manage severe CRS, though their use requires careful consideration due to potential impacts on CAR-T cell efficacy [37].
Emerging strategies are being explored to prevent or mitigate CRS while preserving the therapeutic benefits of CAR-T cells. These include engineering CAR-T cells with mechanisms to regulate cytokine production, such as the double knockdown of IL-6 and IFN-γ, which has shown promise in reducing cytokine release and enhancing the safety profile of CAR-T therapy [35]. Additionally, ongoing research is investigating the use of kinase inhibitors and other pharmacological agents to modulate the immune response and reduce the incidence of CRS [40].
In conclusion, while CAR-T therapy represents a significant advancement in cancer treatment, the associated risk of CRS necessitates vigilant monitoring and proactive management strategies. The development of standardized protocols for grading and managing CRS will be essential as the use of CAR-T therapy expands to a broader patient population [41][42]. Understanding the underlying mechanisms of CRS and implementing innovative approaches to mitigate its effects will be crucial for optimizing patient outcomes in CAR-T therapy.
5.2 Neurotoxicity and Other Side Effects
Chimeric antigen receptor T (CAR-T) cell therapy represents a significant advancement in immunotherapy, particularly for treating hematologic malignancies. This innovative approach involves modifying a patient’s T cells to express a chimeric antigen receptor that specifically recognizes tumor-associated antigens, enabling these engineered T cells to selectively target and eliminate cancer cells. While CAR-T therapy has demonstrated remarkable clinical efficacy, it is also associated with a spectrum of adverse effects, notably neurotoxicity, cytokine release syndrome (CRS), and other complications.
Neurotoxicity is one of the most significant adverse effects observed in patients undergoing CAR-T therapy. Immune effector cell-associated neurotoxicity syndrome (ICANS) encompasses a range of neurological symptoms that can arise after treatment, including cognitive disturbances, seizures, and alterations in consciousness. The incidence of neurotoxicity can vary, with studies indicating that approximately 43% of patients may experience neurological symptoms following CAR-T cell infusion, typically within the first week of treatment [43].
The underlying mechanisms of CAR-T-related neurotoxicity remain largely undefined, although several factors have been proposed. It is suggested that elevated levels of pro-inflammatory cytokines, endothelial dysfunction, and damage to the blood-brain barrier may contribute to the development of neurological symptoms [44]. Additionally, the release of damage-associated molecular patterns (DAMPs) from pyroptotic cells has been identified as a potential mechanism mediating these toxicities [45].
Management strategies for neurotoxicity following CAR-T therapy typically involve symptomatic treatments. Corticosteroids and other immunosuppressive agents, such as tocilizumab, have been employed to mitigate severe neurotoxic effects [46]. Furthermore, antiepileptic drugs may be utilized to manage seizures associated with neurotoxicity [47]. Continuous monitoring and assessment of neurological status are critical to ensure timely intervention and prevent complications [48].
In addition to neurotoxicity, CAR-T therapy is frequently associated with cytokine release syndrome, which can manifest as fever, hypotension, and multi-organ dysfunction due to systemic inflammation. The management of CRS often includes the administration of IL-6 inhibitors, such as tocilizumab, and supportive care [49].
Moreover, nephrotoxicity is another adverse effect that has garnered attention in the context of CAR-T therapy. Acute kidney injury (AKI) can occur in 5% to 33% of patients, with risk factors including pre-existing renal impairment and the use of certain medications [4]. Identifying and managing renal toxicity is essential to maintain overall treatment efficacy and patient safety [49].
In conclusion, while CAR-T cell therapy offers a promising treatment avenue for hematologic malignancies, its associated adverse effects, particularly neurotoxicity and CRS, require careful management. Understanding the pathophysiology behind these toxicities is crucial for developing effective prevention and treatment strategies, ultimately enhancing the safety and efficacy of CAR-T therapy in clinical practice. Ongoing research is necessary to further elucidate the mechanisms involved and to refine management protocols to optimize patient outcomes [6][48].
6 Future Directions in CAR-T Therapy
6.1 Enhancements in CAR Design
Chimeric Antigen Receptor T-cell (CAR-T) therapy is a groundbreaking approach in cancer treatment that leverages the body's immune system to specifically target and eliminate malignant cells. The fundamental mechanism involves extracting T-cells from a patient, genetically modifying them to express chimeric antigen receptors (CARs) that recognize specific antigens on cancer cells, and subsequently reinfusing these modified T-cells back into the patient. Upon reintroduction, the CAR-T cells seek out and destroy cancer cells that express the targeted antigens, leading to potential remission or cure of the malignancy.
Recent advancements in CAR-T therapy have led to the development of next-generation CAR designs aimed at enhancing therapeutic efficacy and safety. These innovations include the incorporation of co-stimulatory domains, which provide additional signals necessary for T-cell activation and proliferation, as well as safety switches that can mitigate severe side effects such as cytokine release syndrome (CRS) and neurotoxicity [50]. The evolution of CAR architecture has transitioned from initial prototypes with limited efficacy to sophisticated designs that integrate various signaling pathways to optimize T-cell functionality [1].
One promising direction in CAR-T therapy is the use of novel genetic engineering tools, such as CRISPR and base editing, to refine CAR constructs further. These technologies enable precise modifications to enhance the persistence and antitumor activity of CAR-T cells while reducing immunogenicity and the risk of graft-versus-host disease (GVHD) [50]. Moreover, researchers are exploring in vivo CAR-T cell engineering, which simplifies the manufacturing process by programming CAR-T cells directly within the patient, potentially lowering costs and improving accessibility [51].
Additionally, the expansion of CAR platforms to include other immune effector cells, such as natural killer (NK) cells and macrophages, is an exciting frontier. This diversification aims to tackle the challenges posed by solid tumors, where traditional CAR-T cells often struggle due to the immunosuppressive tumor microenvironment [8].
To address the limitations of current CAR-T therapies, ongoing research is focusing on enhancing the ability of CAR-T cells to penetrate solid tumors and evade immune suppression. Strategies such as the engineering of CAR-T cells to express additional receptors or cytokines that can counteract the immunosuppressive signals within the tumor microenvironment are being investigated [29]. Furthermore, utilizing advanced 3D culture models for preclinical testing can provide more physiologically relevant insights into CAR-T cell interactions within solid tumors, guiding the design of more effective therapies [7].
In summary, CAR-T therapy operates by genetically modifying T-cells to recognize and attack cancer cells, with ongoing enhancements in CAR design focusing on improving efficacy, safety, and applicability to a broader range of malignancies, including solid tumors. The future of CAR-T therapy appears promising, with innovative approaches poised to overcome existing challenges and expand the therapeutic landscape for cancer treatment [1][50][51].
6.2 Potential for Combination Therapies
Chimeric antigen receptor T-cell (CAR-T) therapy represents a groundbreaking advancement in immunotherapy, particularly in the treatment of hematologic malignancies. This therapeutic approach involves the genetic modification of a patient's T cells to express receptors specific to tumor antigens, enabling these CAR-T cells to recognize and eliminate cancer cells effectively. The efficacy of CAR-T therapy has been particularly notable in conditions such as acute lymphoblastic leukemia (ALL) and various forms of lymphoma, where it has demonstrated significant anti-tumor effects [52].
However, while CAR-T therapy has shown promise in hematologic cancers, its application in solid tumors has encountered several challenges. These challenges include limited infiltration of CAR-T cells into the tumor microenvironment, compromised recognition of cancer cells due to antigen heterogeneity, and the presence of immunosuppressive factors within the tumor [53]. The tumor microenvironment can create physical barriers that hinder the persistence and functionality of CAR-T cells, leading to suboptimal therapeutic outcomes [54].
To enhance the efficacy of CAR-T therapy, researchers are increasingly exploring combination strategies. The integration of CAR-T cells with targeted inhibitors, chemotherapy, and immune checkpoint inhibitors has shown potential in improving the overall therapeutic efficacy. For instance, combining CAR-T therapy with immune checkpoint inhibitors like PD-1 blockers can alleviate T-cell exhaustion and enhance tumor inhibition, leading to improved outcomes in both preclinical and clinical settings [55].
Moreover, small-molecule agents, such as Bruton's tyrosine kinase (BTK) inhibitors and hypomethylating agents, can augment the persistence and proliferative capacity of CAR-T cells, facilitating a more robust anti-tumor response [23]. The rationale behind these combination therapies lies in their ability to synergistically target malignant cells, thereby overcoming resistance mechanisms and minimizing the risks of disease relapse [54].
In the context of solid tumors, researchers have identified various strategies to improve CAR-T cell functionality. These include modifying the CAR structure to enhance intracellular signaling, utilizing oncolytic viruses to increase CAR-T cell infiltration, and employing nanoparticles for targeted delivery [56]. Additionally, rational design approaches aim to optimize CAR-T cells and manipulate the tumor microenvironment to facilitate better outcomes [56].
In summary, CAR-T therapy operates by genetically engineering T cells to target and destroy cancer cells, with ongoing research focused on overcoming its limitations, particularly in solid tumors. The future of CAR-T therapy appears promising, with a strong emphasis on combination therapies that leverage the strengths of various treatment modalities to enhance efficacy, reduce toxicity, and improve patient outcomes across a broader range of malignancies [9].
7 Conclusion
CAR-T therapy has emerged as a groundbreaking advancement in cancer treatment, particularly for hematological malignancies, by leveraging the immune system's ability to target and eliminate cancer cells. The main findings highlight the effectiveness of CAR-T therapy in inducing remission in previously treatment-resistant cancers, with significant success observed in acute lymphoblastic leukemia and certain types of non-Hodgkin lymphoma. However, the therapy faces challenges, particularly in solid tumors, where factors such as antigen heterogeneity and an immunosuppressive tumor microenvironment limit its efficacy. Ongoing research is addressing these challenges through enhancements in CAR design, combination therapies, and innovative engineering approaches. Future directions include optimizing CAR-T cell functionality, exploring novel combinations with other therapeutic modalities, and expanding applications to solid tumors. The integration of these strategies holds the potential to revolutionize cancer treatment further, providing new hope for patients with diverse malignancies and improving long-term outcomes.
References
- [1] Shigui Zhu. CAR-T in cancer therapeutics and updates.. Journal of the National Cancer Center(IF=9.4). 2024. PMID:39281717. DOI: 10.1016/j.jncc.2024.01.001.
- [2] Mi-Gyeong Kim;Dongyoon Kim;Soo-Kyung Suh;Zewon Park;Min Joung Choi;Yu-Kyoung Oh. Current status and regulatory perspective of chimeric antigen receptor-modified T cell therapeutics.. Archives of pharmacal research(IF=7.5). 2016. PMID:26895243. DOI: 10.1007/s12272-016-0719-7.
- [3] John Oluwafemi Teibo;Virginia Picanço-Castro;Lucas Eduardo Botelho de Souza;Vitor Marcel Faça. A Proteomics Outlook on the Molecular Effectors of CAR-T Cell Therapy in Cancer Management.. Journal of proteome research(IF=3.6). 2025. PMID:40047620. DOI: 10.1021/acs.jproteome.4c00930.
- [4] Mehmet Kanbay;Berk Mizrak;Ezgi N Alper;Sidar Copur;Alberto Ortiz. Acute kidney injury following CAR-T cell therapy: a nephrologist's perspective.. Clinical kidney journal(IF=4.6). 2025. PMID:39781479. DOI: 10.1093/ckj/sfae359.
- [5] Mackenzie M Honikel;Scott H Olejniczak. Co-Stimulatory Receptor Signaling in CAR-T Cells.. Biomolecules(IF=4.8). 2022. PMID:36139142. DOI: 10.3390/biom12091303.
- [6] Jiayi Dong;Jiexiong Wu;Ye Jin;Zhu Zheng;Ting Su;Lijuan Shao;Jiaxin Bei;Size Chen. In-depth analysis of the safety of CAR-T cell therapy for solid tumors.. Frontiers in immunology(IF=5.9). 2025. PMID:40066440. DOI: 10.3389/fimmu.2025.1548979.
- [7] Mégane Jassin;Alix Block;Laury Désiront;Louise Vrancken;Céline Grégoire;Frédéric Baron;Grégory Ehx;Thi Tham Nguyen;Jo Caers. From spheroids to organoids: next-generation models for CAR-T cell therapy research in solid tumors.. Frontiers in immunology(IF=5.9). 2025. PMID:40718488. DOI: 10.3389/fimmu.2025.1626369.
- [8] Alina Ershova;Alexandra Goldaeva;Alena Staliarova;Emil Bulatov;Alexey Petukhov;Nikolai Barlev. Future perspectives on novel CAR-T therapeutics beyond CD19 and BCMA in onco-hematology.. Frontiers in immunology(IF=5.9). 2025. PMID:40726984. DOI: 10.3389/fimmu.2025.1592377.
- [9] Ye Kang;Da-Sheng Dang;Xue Sun;Xiao Zhang. Secondary Malignancies of Chimeric Antigen Receptor T-cell Therapy: A Multidimensional Analysis of Mechanisms, Risk Factors, and Treatment Strategies.. Anti-cancer agents in medicinal chemistry(IF=3.0). 2025. PMID:40588994. DOI: 10.2174/0118715206378956250618182616.
- [10] Pouya Safarzadeh Kozani;Abdolhossein Naseri;Seyed Mohamad Javad Mirarefin;Faeze Salem;Mojtaba Nikbakht;Sahar Evazi Bakhshi;Pooria Safarzadeh Kozani. Nanobody-based CAR-T cells for cancer immunotherapy.. Biomarker research(IF=11.5). 2022. PMID:35468841. DOI: 10.1186/s40364-022-00371-7.
- [11] Young-Ho Lee;Chan Hyuk Kim. Evolution of chimeric antigen receptor (CAR) T cell therapy: current status and future perspectives.. Archives of pharmacal research(IF=7.5). 2019. PMID:30830661. DOI: 10.1007/s12272-019-01136-x.
- [12] Hongjing Liu;Xueping Liu;Xuyan Zhou;Siliang Duan;Xin Huang;Hongxin Fei;Yali Kou. Application of nanobody‑based CAR‑T in tumor immunotherapy (Review).. International journal of molecular medicine(IF=5.8). 2025. PMID:40910255. DOI: .
- [13] Duško Lainšček;Anja Golob-Urbanc;Veronika Mikolič;Jelica Pantović-Žalig;Špela Malenšek;Roman Jerala. Regulation of CD19 CAR-T cell activation based on an engineered downstream transcription factor.. Molecular therapy oncolytics(IF=5.3). 2023. PMID:37223115. DOI: 10.1016/j.omto.2023.04.005.
- [14] Valeria Ukrainskaya;Yuri Rubtsov;Dmitry Pershin;Nadezhda Podoplelova;Stanislav Terekhov;Igor Yaroshevich;Anstasiia Sokolova;Dmitry Bagrov;Elena Kulakovskaya;Victoria Shipunova;Sergey Deyev;Rustam Ziganshin;Aleksandr Chernov;Georgii Telegin;Eugene Maksimov;Oleg Markov;Anastasiya Oshchepkova;Marina Zenkova;Jia Xie;Hongkai Zhang;Alexander Gabibov;Michael Maschan;Alexey Stepanov;Richard Lerner. Antigen-Specific Stimulation and Expansion of CAR-T Cells Using Membrane Vesicles as Target Cell Surrogates.. Small (Weinheim an der Bergstrasse, Germany)(IF=12.1). 2021. PMID:34605165. DOI: 10.1002/smll.202102643.
- [15] Somayeh Vandghanooni;Morteza Eskandani;Zohreh Sanaat;Yadollah Omidi. Recent advances in the production, reprogramming, and application of CAR-T cells for treating hematological malignancies.. Life sciences(IF=5.1). 2022. PMID:36179813. DOI: 10.1016/j.lfs.2022.121016.
- [16] Kisha K Patel;Mito Tariveranmoshabad;Siddhant Kadu;Nour Shobaki;Carl June. From concept to cure: The evolution of CAR-T cell therapy.. Molecular therapy : the journal of the American Society of Gene Therapy(IF=12.0). 2025. PMID:40070120. DOI: 10.1016/j.ymthe.2025.03.005.
- [17] Rafaela Abrantes;Henrique O Duarte;Catarina Gomes;Sébastien Wälchli;Celso A Reis. CAR-Ts: new perspectives in cancer therapy.. FEBS letters(IF=3.0). 2022. PMID:34978080. DOI: 10.1002/1873-3468.14270.
- [18] Paolo Strati;Tara Gregory;Navneet S Majhail;Nitin Jain. Chimeric Antigen Receptor T-Cell Therapy for Hematologic Malignancies: A Practical Review.. JCO oncology practice(IF=4.6). 2023. PMID:37406255. DOI: 10.1200/OP.22.00819.
- [19] Samer Al Hadidi;Helen E Heslop;Malcolm K Brenner;Masataka Suzuki. Bispecific antibodies and autologous chimeric antigen receptor T cell therapies for treatment of hematological malignancies.. Molecular therapy : the journal of the American Society of Gene Therapy(IF=12.0). 2024. PMID:38822527. DOI: 10.1016/j.ymthe.2024.05.039.
- [20] Zijun Zhao;Yu Chen;Ngiambudulu M Francisco;Yuanqing Zhang;Minhao Wu. The application of CAR-T cell therapy in hematological malignancies: advantages and challenges.. Acta pharmaceutica Sinica. B(IF=14.6). 2018. PMID:30109179. DOI: 10.1016/j.apsb.2018.03.001.
- [21] Samane Abbasi;Milad Asghari Totmaj;Masoumeh Abbasi;Saba Hajazimian;Pouya Goleij;Javad Behroozi;Behrouz Shademan;Alireza Isazadeh;Behzad Baradaran. Chimeric antigen receptor T (CAR-T) cells: Novel cell therapy for hematological malignancies.. Cancer medicine(IF=3.1). 2023. PMID:36583504. DOI: 10.1002/cam4.5551.
- [22] Jingxian Li;Huiguang Chen;Chaoping Xu;Mengci Hu;Jiangping Li;Wei Chang. Systemic toxicity of CAR-T therapy and potential monitoring indicators for toxicity prevention.. Frontiers in immunology(IF=5.9). 2024. PMID:39253080. DOI: 10.3389/fimmu.2024.1422591.
- [23] Baiyan Yang;Yang Su;Chunling Wang;Liang Yu. Advances in the combination of CAR-T therapy with small-molecule reagents for hematologic malignancies.. Frontiers in immunology(IF=5.9). 2025. PMID:41169368. DOI: 10.3389/fimmu.2025.1663522.
- [24] Yanyu Lin;Shuqi Luo;Jianhui Wei;Shujin Lin;Dawei Wang;Xiangqian Zhao;Zexin Feng;Yangkun Shen;Qi Chen. Limitations of CAR-T-Cell Therapy in Hematologic Malignancies: Focusing on Antigen Escape and T-Cell Dysfunction.. International journal of molecular sciences(IF=4.9). 2025. PMID:41096934. DOI: 10.3390/ijms26199669.
- [25] Swati Arora;Pranshul Sethi;Adrita Banerjee;Md Khokon Miah Akanda;Mohamed S Abd El Hafeez;Sanzia Mehjabin;Keshav S Moharir;Swati Mohanty;Sumit Sheoran. Chimeric antigen receptor T-cell therapy for solid tumors: A review of the intricate mechanisms and potential strategies.. International immunopharmacology(IF=4.7). 2025. PMID:41240717. DOI: 10.1016/j.intimp.2025.115838.
- [26] Jing Guo;Chunhe Zhou;Hongmei Zhao;Huiyan Li. Challenges and breakthroughs: current landscape and future prospects of CAR-T cell therapy clinical trials for solid tumors.. Frontiers in oncology(IF=3.3). 2025. PMID:40969260. DOI: 10.3389/fonc.2025.1652329.
- [27] Yang Chen;Ran Ren;Lirong Yan;Yu Zhou;Ruokai Sun;Huicong Song;Hongfei Yan;Yongsheng Li. From Bench to Bedside: Emerging Paradigms in CAR-T Cell Therapy for Solid Malignancies.. Advanced science (Weinheim, Baden-Wurttemberg, Germany)(IF=14.1). 2025. PMID:40855662. DOI: 10.1002/advs.202505822.
- [28] Tong Chen;Mingzhao Wang;Yanchao Chen;Yutao Liu. Current challenges and therapeutic advances of CAR-T cell therapy for solid tumors.. Cancer cell international(IF=6.0). 2024. PMID:38622705. DOI: 10.1186/s12935-024-03315-3.
- [29] Zheng-Zheng Zhang;Tian Wang;Xiao-Feng Wang;Yu-Qing Zhang;Shu-Xia Song;Cui-Qing Ma. Improving the ability of CAR-T cells to hit solid tumors: Challenges and strategies.. Pharmacological research(IF=10.5). 2022. PMID:34920118. DOI: 10.1016/j.phrs.2021.106036.
- [30] Natalia Picheta;Julia Piekarz;Karolina Daniłowska;Katarzyna Szklener;Sławomir Mańdziuk. CAR-T in the Treatment of Solid Tumors-A Review of Current Research and Future Perspectives.. International journal of molecular sciences(IF=4.9). 2025. PMID:41096753. DOI: 10.3390/ijms26199486.
- [31] Tang-Her Jaing;Yi-Wen Hsiao;Yi-Lun Wang. Chimeric Antigen Receptor Cell Therapy: Empowering Treatment Strategies for Solid Tumors.. Current issues in molecular biology(IF=3.0). 2025. PMID:39996811. DOI: 10.3390/cimb47020090.
- [32] Siyuan Qian;Pedro Villarejo-Campos;Ismael Guijo;Sergio Hernández-Villafranca;Damián García-Olmo;Sara González-Soares;Héctor Guadalajara;Santos Jiménez-Galanes;Cheng Qian. Update for Advance CAR-T Therapy in Solid Tumors, Clinical Application in Peritoneal Carcinomatosis From Colorectal Cancer and Future Prospects.. Frontiers in immunology(IF=5.9). 2022. PMID:35401510. DOI: 10.3389/fimmu.2022.841425.
- [33] Noelle Frey;David Porter. Cytokine Release Syndrome with Chimeric Antigen Receptor T Cell Therapy.. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation(IF=4.3). 2019. PMID:30586620. DOI: 10.1016/j.bbmt.2018.12.756.
- [34] Lele Miao;Zhengchao Zhang;Zhijian Ren;Yumin Li. Reactions Related to CAR-T Cell Therapy.. Frontiers in immunology(IF=5.9). 2021. PMID:33995389. DOI: 10.3389/fimmu.2021.663201.
- [35] Huihui Zhang;Xiaofei Lv;Qunfang Kong;Yi Tan. IL-6/IFN-γ double knockdown CAR-T cells reduce the release of multiple cytokines from PBMCs in vitro.. Human vaccines & immunotherapeutics(IF=3.5). 2022. PMID:35049413. DOI: 10.1080/21645515.2021.2016005.
- [36] Zhenyu Wei;Qi Cheng;Nan Xu;Chengkui Zhao;Jiayu Xu;Liqing Kang;Xiaoyan Lou;Lei Yu;Weixing Feng. Investigation of CRS-associated cytokines in CAR-T therapy with meta-GNN and pathway crosstalk.. BMC bioinformatics(IF=3.3). 2022. PMID:36100873. DOI: 10.1186/s12859-022-04917-2.
- [37] Alexandre V Hirayama;Cameron J Turtle. Toxicities of CD19 CAR-T cell immunotherapy.. American journal of hematology(IF=9.9). 2019. PMID:30784102. DOI: 10.1002/ajh.25445.
- [38] Sarju Ganatra;Joseph R Carver;Salim S Hayek;Bonnie Ky;Monika J Leja;Daniel J Lenihan;Carrie Lenneman;Negaresh Mousavi;Jae H Park;Miguel Angel Perales;Thomas D Ryan;Marielle Scherrer-Crosbie;Richard M Steingart;Eric H Yang;Vlad Zaha;Ana Barac;Jennifer E Liu. Chimeric Antigen Receptor T-Cell Therapy for Cancer and Heart: JACC Council Perspectives.. Journal of the American College of Cardiology(IF=22.3). 2019. PMID:31856973. DOI: 10.1016/j.jacc.2019.10.049.
- [39] Alexander Shimabukuro-Vornhagen;Philipp Gödel;Marion Subklewe;Hans Joachim Stemmler;Hans Anton Schlößer;Max Schlaak;Matthias Kochanek;Boris Böll;Michael S von Bergwelt-Baildon. Cytokine release syndrome.. Journal for immunotherapy of cancer(IF=10.6). 2018. PMID:29907163. DOI: 10.1186/s40425-018-0343-9.
- [40] Gabrielle Leclercq;Nathalie Steinhoff;Hélène Haegel;Donata De Marco;Marina Bacac;Christian Klein. Novel strategies for the mitigation of cytokine release syndrome induced by T cell engaging therapies with a focus on the use of kinase inhibitors.. Oncoimmunology(IF=6.3). 2022. PMID:35694193. DOI: 10.1080/2162402X.2022.2083479.
- [41] Chieh Yang;John Nguyen;Yun Yen. Complete spectrum of adverse events associated with chimeric antigen receptor (CAR)-T cell therapies.. Journal of biomedical science(IF=12.1). 2023. PMID:37864230. DOI: 10.1186/s12929-023-00982-8.
- [42] Weronika Ploch;Karol Sadowski;Wioletta Olejarz;Grzegorz W Basak. Advancement and Challenges in Monitoring of CAR-T Cell Therapy: A Comprehensive Review of Parameters and Markers in Hematological Malignancies.. Cancers(IF=4.4). 2024. PMID:39409959. DOI: 10.3390/cancers16193339.
- [43] Catherine Belin;Perrine Devic;Xavier Ayrignac;Amélie Dos Santos;Adrien Paix;Lila Sirven-Villaros;Claire Simard;Sylvain Lamure;Thomas Gastinne;Renata Ursu;Colette Berger;Laura Platon;Benoît Tessoulin;Elie Azoulay;Florent Wallet;Catherine Thieblemont;Emmanuel Bachy;Guillaume Cartron;David A Laplaud;Antoine F Carpentier. Description of neurotoxicity in a series of patients treated with CAR T-cell therapy.. Scientific reports(IF=3.9). 2020. PMID:33149178. DOI: 10.1038/s41598-020-76055-9.
- [44] Bradley D Hunter;Caron A Jacobson. CAR T-Cell Associated Neurotoxicity: Mechanisms, Clinicopathologic Correlates, and Future Directions.. Journal of the National Cancer Institute(IF=7.2). 2019. PMID:30753567. DOI: 10.1093/jnci/djz017.
- [45] Tian Deng;Chao Tang;Guizhong Zhang;Xiaochun Wan. DAMPs released by pyroptotic cells as major contributors and therapeutic targets for CAR-T-related toxicities.. Cell death & disease(IF=9.6). 2021. PMID:33504767. DOI: 10.1038/s41419-021-03428-x.
- [46] Lidia Gatto;Ilaria Ricciotti;Alicia Tosoni;Vincenzo Di Nunno;Stefania Bartolini;Lucia Ranieri;Enrico Franceschi. CAR-T cells neurotoxicity from consolidated practice in hematological malignancies to fledgling experience in CNS tumors: fill the gap.. Frontiers in oncology(IF=3.3). 2023. PMID:37397356. DOI: 10.3389/fonc.2023.1206983.
- [47] Philipp Karschnia;Jörg Dietrich. Neurological complications of CAR T cell therapy for cancers.. Nature reviews. Neurology(IF=33.1). 2025. PMID:40562951. DOI: 10.1038/s41582-025-01112-8.
- [48] Anna Fleischer;Sophia Kurth;Johannes Duell;Max Topp;Patrick-Pascal Strunz;Julia Mersi;Leo Rasche;Carmen Sanges;Michael Hudecek;Hermann Einsele;Imad Maatouk. Neuropsychiatric manifestations following chimeric antigen receptor T cell therapy for cancer: a systematic review of clinical outcomes and management strategies.. Journal for immunotherapy of cancer(IF=10.6). 2024. PMID:39794934. DOI: 10.1136/jitc-2024-009174.
- [49] Karol Sadowski;Weronika Ploch;Alicja Downar;Wiktoria Giza;Dorota Szcześ;Wioletta Olejarz;Wiesław W Jędrzejczak;Jolanta Małyszko;Grzegorz Basak. Nephrotoxicity in CAR-T Cell Therapy.. Transplantation and cellular therapy(IF=4.4). 2025. PMID:40107382. DOI: 10.1016/j.jtct.2025.03.007.
- [50] Ahdab A Alsaieedi;Kawther A Zaher. Tracing the development of CAR-T cell design: from concept to next-generation platforms.. Frontiers in immunology(IF=5.9). 2025. PMID:40771804. DOI: 10.3389/fimmu.2025.1615212.
- [51] Yizhao Chen;Qianling Xin;Jiaqi Qiu;Mengjuan Zhu;Zixuan Li;Ji Qiu;Jiajie Tu;Ruilin Li. In vivo CAR-T cell engineering: concept, research progress, potential challenges and enhancement strategies.. Experimental hematology & oncology(IF=13.5). 2025. PMID:41250215. DOI: 10.1186/s40164-025-00725-5.
- [52] Luyao Wang;Ruixue Yao;Lifa Zhang;Chuanbo Fan;Leina Ma;Jia Liu. Chimeric antigen receptor T cell therapy and other therapeutics for malignancies: Combination and opportunity.. International immunopharmacology(IF=4.7). 2019. PMID:30875561. DOI: 10.1016/j.intimp.2019.01.010.
- [53] Jiahao Liu;Xiaofei Jiao;Ding Ma;Yong Fang;Qinglei Gao. CAR-T therapy and targeted treatments: Emerging combination strategies in solid tumors.. Med (New York, N.Y.)(IF=11.8). 2024. PMID:38547867. DOI: 10.1016/j.medj.2024.03.001.
- [54] Kyohei Misawa;Hina Bhat;Prasad S Adusumilli;Zhaohua Hou. Combinational CAR T-cell therapy for solid tumors: Requisites, rationales, and trials.. Pharmacology & therapeutics(IF=12.5). 2025. PMID:39617146. DOI: 10.1016/j.pharmthera.2024.108763.
- [55] Bibhu Prasad Satapathy;Pooja Sheoran;Rohit Yadav;Dewan Chettri;Dhruba Sonowal;Chinmayee Priyadarsini Dash;Prachi Dhaka;Vivek Uttam;Ritu Yadav;Manju Jain;Aklank Jain. The synergistic immunotherapeutic impact of engineered CAR-T cells with PD-1 blockade in lymphomas and solid tumors: a systematic review.. Frontiers in immunology(IF=5.9). 2024. PMID:38799440. DOI: 10.3389/fimmu.2024.1389971.
- [56] Chaojie Zhu;Qing Wu;Tao Sheng;Jiaqi Shi;Xinyuan Shen;Jicheng Yu;Yang Du;Jie Sun;Tingxizi Liang;Kaixin He;Yuan Ding;Hongjun Li;Zhen Gu;Weilin Wang. Rationally designed approaches to augment CAR-T therapy for solid tumor treatment.. Bioactive materials(IF=20.3). 2024. PMID:38059121. DOI: 10.1016/j.bioactmat.2023.11.002.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
Chimeric Antigen Receptor T cells · Immunotherapy · Hematological Malignancies · Cell Engineering · Adverse Effects
© 2025 MaltSci
