Appearance
This report is written by MaltSci based on the latest literature and research findings
How does CAR-T therapy treat hematologic malignancies?
Abstract
The advent of Chimeric Antigen Receptor T-cell (CAR-T) therapy has revolutionized the treatment of hematologic malignancies, particularly for patients with relapsed or refractory forms of acute lymphoblastic leukemia (ALL), diffuse large B-cell lymphoma (DLBCL), and multiple myeloma (MM). CAR-T therapy harnesses the patient's immune system by genetically modifying T cells to express receptors that specifically target tumor-associated antigens, resulting in remarkable response rates and durable remissions. This review comprehensively examines the mechanisms of CAR-T therapy, including the design and engineering of CARs, the activation and expansion of CAR-T cells, and the specific targeting of tumor antigens. It highlights the clinical successes achieved with CAR-T therapy in hematologic malignancies, emphasizing its potential as a curative option for patients who have exhausted conventional treatments. However, the implementation of CAR-T therapy is not without challenges, including severe adverse effects such as cytokine release syndrome and neurotoxicity, along with manufacturing complexities and issues related to patient accessibility. Furthermore, the risk of antigen escape and relapse poses significant hurdles that need to be addressed. Future directions in CAR-T therapy involve optimizing CAR designs, exploring combination therapies, and expanding applications to solid tumors, which hold promise for improving patient outcomes. This review aims to elucidate the transformative role of CAR-T therapy in hematologic malignancies while identifying critical areas for future research and development.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Mechanisms of CAR-T Therapy
- 2.1 Design and Engineering of CARs
- 2.2 Activation and Expansion of CAR-T Cells
- 2.3 Targeting Mechanisms and Tumor Antigens
- 3 Clinical Applications of CAR-T Therapy
- 3.1 Success in Acute Lymphoblastic Leukemia
- 3.2 Impact on Diffuse Large B-Cell Lymphoma
- 3.3 Efficacy in Multiple Myeloma
- 4 Challenges and Limitations
- 4.1 Adverse Effects: Cytokine Release Syndrome and Neurotoxicity
- 4.2 Manufacturing and Accessibility Issues
- 4.3 Antigen Escape and Relapse
- 5 Future Directions in CAR-T Therapy
- 5.1 Enhancements in CAR Design
- 5.2 Combination Therapies
- 5.3 Expanding Applications to Solid Tumors
- 6 Conclusion
1 Introduction
The treatment landscape for hematologic malignancies has undergone a significant transformation in recent years, particularly with the advent of Chimeric Antigen Receptor T-cell (CAR-T) therapy. Initially approved in 2017 for relapsed or refractory acute lymphoblastic leukemia (ALL), CAR-T therapy represents a paradigm shift in cancer treatment by utilizing the patient's own immune system to target and eliminate malignant cells. This innovative approach involves the genetic modification of T-cells to express CARs that specifically recognize tumor-associated antigens, leading to remarkable response rates and long-term remission in various hematologic cancers, including diffuse large B-cell lymphoma (DLBCL) and multiple myeloma [1][2].
The significance of CAR-T therapy lies not only in its therapeutic efficacy but also in its potential to provide a curative option for patients who have exhausted all conventional treatment avenues. Traditional therapies, such as chemotherapy and radiation, often come with substantial limitations, including toxicity and the development of resistance [3]. In contrast, CAR-T therapy has demonstrated the ability to induce durable remissions, particularly in B-cell malignancies, highlighting its role as a cornerstone in modern oncology [4].
Despite its promise, the implementation of CAR-T therapy is fraught with challenges. The risk of severe adverse effects, such as cytokine release syndrome (CRS) and neurotoxicity, poses significant hurdles in clinical practice [5]. Furthermore, issues related to the manufacturing process, patient selection, and the emergence of antigen-negative relapses complicate the widespread adoption of this treatment modality [6]. As the field continues to evolve, understanding the mechanisms underlying CAR-T therapy, along with its clinical applications and limitations, is essential for optimizing patient outcomes.
This review aims to provide a comprehensive overview of CAR-T therapy in the context of hematologic malignancies. The discussion will be organized into several key sections. First, we will delve into the mechanisms of CAR-T therapy, covering the design and engineering of CARs, the activation and expansion of CAR-T cells, and the targeting mechanisms involved in tumor recognition. Next, we will explore the clinical applications of CAR-T therapy, highlighting its success in treating ALL, DLBCL, and multiple myeloma. Following this, we will address the challenges and limitations associated with CAR-T therapy, including adverse effects, manufacturing and accessibility issues, and the phenomenon of antigen escape and relapse. Finally, we will outline future directions in CAR-T therapy, focusing on enhancements in CAR design, potential combination therapies, and the expansion of CAR-T applications to solid tumors.
By synthesizing current literature and clinical data, this report will elucidate the transformative role of CAR-T therapy in treating hematologic malignancies and identify critical areas for future research and development. The ongoing evolution of CAR-T therapy not only holds promise for improved patient outcomes but also represents a significant advancement in the field of cancer immunotherapy.
2 Mechanisms of CAR-T Therapy
2.1 Design and Engineering of CARs
Chimeric antigen receptor T-cell (CAR-T) therapy has emerged as a transformative approach in the treatment of hematologic malignancies, demonstrating significant clinical efficacy, particularly in B-cell malignancies such as acute lymphoblastic leukemia (ALL), diffuse large B-cell lymphoma (DLBCL), and multiple myeloma (MM). The fundamental mechanism of CAR-T therapy involves the genetic modification of a patient’s T cells to express a CAR that specifically recognizes tumor-associated antigens on malignant cells. This enables the engineered T cells to identify and eradicate tumor cells, thus enhancing the patient’s prognosis (Mi et al. 2021; Abbasi et al. 2023).
The design and engineering of CARs are critical to their function and efficacy. CARs typically consist of an extracellular antigen recognition domain, often derived from an antibody, a transmembrane domain, and an intracellular signaling domain that activates T cells upon antigen binding. The most common targets for CAR-T therapy include CD19, which is predominantly expressed on B-cell malignancies, and B-cell maturation antigen (BCMA), targeted in multiple myeloma. Recent advancements have also introduced multi-targeted CARs, which aim to address challenges such as antigen escape and T-cell dysfunction, thus enhancing the durability of responses in patients (Lin et al. 2025; Zhou et al. 2024).
Moreover, the engineering of CARs has evolved to improve T-cell persistence and function. This includes the incorporation of costimulatory domains (e.g., CD28, 4-1BB) that enhance T-cell activation and survival, and the use of advanced techniques such as CRISPR for precise genome editing to improve the specificity and safety of CAR-T cells (Głowacki & Rieske 2022; Yan et al. 2025). The optimization of CAR design is pivotal for overcoming the limitations of current therapies, such as treatment resistance and adverse effects like cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), which are significant challenges in clinical settings (Strati et al. 2023; English et al. 2024).
The integration of CAR-T therapy with other therapeutic modalities, such as small-molecule inhibitors and bispecific antibodies, is also being explored to enhance the overall antitumor efficacy and address the limitations of monotherapy. These combination strategies aim to create a synergistic effect, allowing for more effective elimination of malignant cells and improvement of patient outcomes (Yang et al. 2025; Al Hadidi et al. 2024).
In conclusion, CAR-T therapy represents a novel and powerful strategy for treating hematologic malignancies, with ongoing research focused on optimizing CAR design and engineering to improve efficacy, safety, and clinical outcomes. The evolution of this therapy continues to provide hope for patients with refractory and relapsed hematologic cancers, making it a cornerstone of modern oncological treatment paradigms.
2.2 Activation and Expansion of CAR-T Cells
Chimeric antigen receptor T-cell (CAR-T) therapy has revolutionized the treatment of hematologic malignancies by employing a sophisticated mechanism that enhances the body's immune response against cancer cells. The core principle of CAR-T therapy involves the genetic modification of a patient's T cells to express a CAR, which specifically recognizes antigens predominantly present on the surface of tumor cells. This allows the engineered T cells to effectively identify and eradicate malignant cells, thereby significantly improving patient prognosis.
The activation of CAR-T cells begins with the recognition of tumor-associated antigens. Upon encountering these antigens, the CAR-T cells undergo a series of activation steps. This activation is crucial as it leads to the proliferation and expansion of CAR-T cells. Once activated, the CAR-T cells proliferate rapidly, producing a large number of effector T cells that are capable of mounting a robust immune response against the tumor. This expansion is essential for ensuring that sufficient numbers of CAR-T cells are available to effectively target and eliminate tumor cells in the body.
Moreover, CAR-T therapy enhances the persistence of these engineered T cells within the patient's system. By improving the proliferative capacity and longevity of CAR-T cells, the therapy aims to sustain an effective immune response over time, potentially reducing the risk of disease relapse. The persistence of CAR-T cells is critical, as it allows for continued surveillance against residual malignant cells, which may not have been eliminated during the initial treatment phase.
Combination strategies further augment the efficacy of CAR-T therapy. Small molecule compounds, such as Bruton's tyrosine kinase (BTK) inhibitors and immunomodulatory drugs, have been shown to enhance the antitumor efficacy of CAR-T cells. These agents can improve the persistence and proliferative capacity of CAR-T cells, enabling a more effective and sustained attack on malignant cells. By employing a multi-faceted approach, CAR-T therapy not only targets the tumor directly but also modulates the tumor microenvironment to facilitate a more effective immune response [6].
In summary, CAR-T therapy treats hematologic malignancies through a well-orchestrated mechanism involving the activation, expansion, and persistence of engineered T cells. By enhancing the immune system's ability to recognize and eliminate cancer cells, CAR-T therapy has emerged as a cornerstone in the treatment of various hematologic malignancies, providing new hope for patients facing these challenging conditions.
2.3 Targeting Mechanisms and Tumor Antigens
Chimeric antigen receptor T-cell (CAR-T) therapy represents a groundbreaking advancement in the treatment of hematologic malignancies, particularly in B-cell cancers such as acute lymphoblastic leukemia (ALL), diffuse large B-cell lymphoma (DLBCL), chronic lymphocytic leukemia (CLL), and multiple myeloma (MM). The underlying mechanism of CAR-T therapy involves the genetic modification of a patient's T cells to express chimeric antigen receptors that specifically recognize and bind to tumor-associated antigens on malignant cells.
The targeting mechanism of CAR-T therapy is primarily based on the identification of tumor-specific antigens. For instance, the CAR-T cells are engineered to target CD19, a protein commonly expressed on the surface of B cells, including malignant B cells. This targeted approach allows CAR-T cells to selectively recognize and eliminate cancer cells while sparing normal cells that do not express the target antigen. The binding of CAR-T cells to tumor antigens initiates a cytotoxic response, leading to the destruction of the tumor cells through various mechanisms, including the release of cytotoxic granules, induction of apoptosis, and recruitment of other immune cells to the tumor site [2][6].
Moreover, CAR-T therapy enhances the persistence and proliferation of T cells within the tumor microenvironment. By employing costimulatory domains within the CAR construct, such as CD28 or 4-1BB, the engineered T cells receive additional signals that promote their survival and functional activity. This augmentation is crucial for overcoming the immunosuppressive milieu often present in hematologic malignancies, thereby improving the overall efficacy of the treatment [7].
Despite its success, CAR-T therapy is not without challenges. One significant limitation is the phenomenon of antigen escape, where tumor cells may downregulate or lose the expression of the targeted antigen, leading to treatment resistance. This has prompted research into multi-targeted CAR-T strategies, where T cells are engineered to recognize multiple antigens simultaneously, thereby reducing the likelihood of tumor escape [7].
In summary, CAR-T therapy utilizes a precise targeting mechanism that exploits tumor-specific antigens to direct T-cell-mediated cytotoxicity against hematologic malignancies. By enhancing T-cell persistence and addressing potential resistance mechanisms, CAR-T therapy continues to evolve as a potent treatment modality for patients with relapsed or refractory hematologic cancers. Future research aims to refine these strategies further, improving efficacy and minimizing adverse effects [8][9].
3 Clinical Applications of CAR-T Therapy
3.1 Success in Acute Lymphoblastic Leukemia
Chimeric antigen receptor T-cell (CAR-T) therapy has emerged as a groundbreaking treatment modality for hematologic malignancies, particularly acute lymphoblastic leukemia (ALL). The success of CAR-T therapy in ALL can be attributed to its ability to genetically modify a patient's T cells to express a CAR that specifically targets tumor-associated antigens, such as CD19, which is predominantly expressed on B cells.
Since the first CAR-T therapy was approved for relapsed/refractory ALL in 2017, the clinical application of this therapy has shown remarkable efficacy. In a pivotal study, CAR-T therapy demonstrated high response rates, with many patients achieving complete remission. This therapeutic approach allows for the precise targeting of malignant cells, leading to significant tumor reduction and improved patient outcomes. For instance, CAR-T therapies targeting CD19 have led to durable remissions in patients who previously had limited treatment options, thereby revolutionizing the treatment landscape for ALL[10][11].
The clinical application of CAR-T therapy in ALL involves several steps: T cells are collected from the patient, genetically modified to express the CAR, and then infused back into the patient. Once reintroduced, these CAR-T cells can recognize and bind to CD19 on leukemia cells, triggering an immune response that results in the destruction of the cancerous cells[1].
Despite the promising results, challenges remain. Patients may experience side effects such as cytokine release syndrome (CRS) and neurotoxicity, which require careful management. Nevertheless, the therapeutic benefits of CAR-T therapy in ALL have led to ongoing research aimed at expanding its use to other hematologic malignancies and solid tumors, thus underscoring its potential as a cornerstone of cancer immunotherapy[2][3].
Overall, CAR-T therapy represents a significant advancement in the treatment of ALL, offering hope for long-term remission and potential cures for patients with this aggressive malignancy. Its success has not only improved clinical outcomes but has also paved the way for further innovations in the field of immunotherapy[4][12].
3.2 Impact on Diffuse Large B-Cell Lymphoma
Chimeric Antigen Receptor T-cell (CAR-T) therapy has emerged as a groundbreaking treatment modality for hematologic malignancies, particularly in cases of diffuse large B-cell lymphoma (DLBCL). This innovative therapeutic approach involves the genetic modification of a patient's T cells to express chimeric antigen receptors that specifically target tumor antigens, enabling the CAR-T cells to identify and eliminate malignant cells.
The efficacy of CAR-T therapy has been particularly notable in the treatment of DLBCL, where it has shown significant clinical success. Studies have demonstrated that CAR-T cell therapy can lead to durable complete remissions in heavily pretreated patients suffering from aggressive forms of B-cell non-Hodgkin lymphoma, including DLBCL. For instance, clinical trials have reported that patients treated with CD19-targeted CAR-T cells have achieved improved survival rates compared to traditional treatment options, effectively altering the treatment landscape for this malignancy (Castaneda Puglianini & Chavez, 2024) [13].
Despite the remarkable advancements, challenges remain in the application of CAR-T therapy for DLBCL. One major concern is the occurrence of relapses following treatment, which can arise due to several factors, including genetic mutations, antigen escape, and CAR-T cell exhaustion (Wang, 2022) [14]. The durability of response is critical, as research indicates that the pooled prevalence of relapse within the first 12 months after CAR-T infusion can be as high as 61% [15]. Therefore, understanding the mechanisms behind these relapses is essential for optimizing treatment strategies and improving patient outcomes.
The safety profile of CAR-T therapy is another important consideration. While it has revolutionized treatment for DLBCL, it is associated with specific toxicities such as cytokine release syndrome (CRS) and neurotoxicity, which can complicate patient management (Strati et al., 2023) [2]. Furthermore, the treatment's cost remains a barrier to access for many patients, limiting its widespread adoption despite its potential benefits.
Ongoing research efforts are directed towards enhancing the safety and efficacy of CAR-T therapies. These include the development of next-generation CAR constructs aimed at improving persistence and reducing adverse effects, as well as exploring combination therapies with other novel agents to maximize therapeutic benefits while minimizing risks (Hijazi et al., 2025) [16]. By refining these approaches, the field aims to broaden the applicability of CAR-T therapy to a wider range of hematologic malignancies, thereby improving treatment outcomes for patients with DLBCL and beyond.
In summary, CAR-T therapy represents a significant advancement in the treatment of hematologic malignancies, particularly DLBCL, with its ability to induce durable remissions. However, addressing the challenges of relapse and toxicity is crucial for the continued evolution and success of this promising therapeutic modality.
3.3 Efficacy in Multiple Myeloma
Chimeric antigen receptor T-cell (CAR-T) therapy has emerged as a significant advancement in the treatment of hematologic malignancies, particularly multiple myeloma (MM). This innovative immunotherapeutic approach involves the genetic modification of a patient’s T cells to express CARs, which are engineered receptors that enable T cells to recognize and attack cancer cells. The efficacy of CAR-T therapy in multiple myeloma has been supported by various clinical studies, highlighting its potential to revolutionize treatment strategies for this challenging malignancy.
Multiple myeloma is characterized by the abnormal proliferation of plasma cells in the bone marrow, leading to various complications, including bone disease and hematologic issues. The treatment landscape for MM has traditionally included chemotherapy, immunomodulatory agents, and proteasome inhibitors, but these therapies often yield limited long-term success, particularly in relapsed or refractory cases [17]. The introduction of CAR-T therapy has provided new hope, especially for patients who have exhausted other treatment options.
CAR-T therapies targeting B-cell maturation antigen (BCMA) have shown particularly promising results in clinical trials. BCMA is highly expressed on malignant plasma cells, making it an ideal target for CAR-T cells. Studies have reported significant antimyeloma activity from CAR-T cells directed against BCMA, leading to high response rates in patients with relapsed/refractory MM [18].
Clinical applications of CAR-T therapy in MM include its use as a standalone treatment or in combination with other modalities, such as autologous stem cell transplantation (ASCT). The integration of CAR-T therapy post-ASCT may enhance the eradication of residual disease and improve long-term outcomes by re-establishing the immune system [19]. Moreover, the therapy is designed to target specific antigens on myeloma cells while minimizing damage to normal cells, thus reducing off-target effects.
Despite its efficacy, CAR-T therapy is not without challenges. The treatment can lead to severe side effects, including cytokine release syndrome (CRS) and neurotoxicity, which necessitate careful monitoring and management [20]. Additionally, the genetic and phenotypic heterogeneity of multiple myeloma suggests that targeting multiple antigens may be necessary to achieve optimal treatment outcomes [17].
The ongoing research aims to refine CAR-T therapies further, focusing on enhancing their efficacy, safety, and accessibility. Efforts are being made to develop next-generation CAR constructs that can persist longer in the body and induce more robust immune responses against myeloma [2]. Additionally, combination therapies that integrate CAR-T cells with other therapeutic agents are being explored to overcome resistance mechanisms and improve patient outcomes [21].
In summary, CAR-T therapy represents a promising and transformative approach for treating multiple myeloma, with significant clinical efficacy observed in relapsed and refractory cases. The potential for CAR-T cells to target specific tumor antigens while sparing normal cells, coupled with ongoing advancements in the field, positions this therapy as a key component of future treatment strategies for hematologic malignancies.
4 Challenges and Limitations
4.1 Adverse Effects: Cytokine Release Syndrome and Neurotoxicity
Chimeric antigen receptor T (CAR-T) cell therapy has emerged as a revolutionary treatment for hematologic malignancies, particularly in cases of relapsed or refractory (R/R) B-cell malignancies, including acute lymphoblastic leukemia, non-Hodgkin lymphoma, and multiple myeloma. The therapy involves engineering a patient's T cells to express CARs that specifically target tumor-associated antigens, such as CD19 and BCMA. This targeted approach allows for the selective destruction of malignant cells, leading to significant therapeutic success in achieving remission for patients who have exhausted other treatment options [2][22].
Despite its efficacy, CAR-T therapy is not without challenges and limitations. One of the primary concerns is the occurrence of adverse effects, which can range from mild to life-threatening. The most notable adverse events associated with CAR-T therapy include cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). CRS is characterized by a hyper-inflammatory response due to the rapid proliferation and activation of CAR-T cells, leading to elevated levels of cytokines. Symptoms can vary widely, including fever, fatigue, hypotension, and multi-organ dysfunction, with severe cases requiring intensive medical intervention [23][24].
ICANS, on the other hand, presents as neurological symptoms, which can include confusion, agitation, seizures, and in severe cases, encephalopathy. The pathophysiology of ICANS is believed to be closely related to the severity of CRS, as elevated cytokine levels can affect the central nervous system [2][25]. Both CRS and ICANS pose significant challenges in the clinical management of patients undergoing CAR-T therapy, necessitating careful monitoring and supportive care strategies [26].
In addition to CRS and ICANS, other adverse effects associated with CAR-T therapy include hematologic toxicities such as cytopenias, particularly thrombocytopenia, which can persist for an extended duration post-treatment [27]. The risk of infections is also heightened due to the immunocompromised state of patients following T cell infusion. These toxicities can complicate the clinical course and may limit the applicability of CAR-T therapy in certain patient populations [2][28].
The management of these adverse effects is an area of active research, with ongoing studies aimed at identifying biomarkers for early detection, optimizing treatment protocols, and developing strategies to mitigate the risks associated with CAR-T therapy [24][29]. Future advancements may involve the use of biomaterials and novel engineering approaches to enhance the safety profile of CAR-T therapies while maintaining their efficacy [29].
In conclusion, while CAR-T therapy represents a significant advancement in the treatment of hematologic malignancies, its associated adverse effects, particularly CRS and neurotoxicity, present substantial challenges that require ongoing research and clinical attention to improve patient outcomes and expand the therapy's applicability.
4.2 Manufacturing and Accessibility Issues
Chimeric antigen receptor T-cell (CAR-T) therapy has emerged as a transformative treatment for hematologic malignancies, offering significant clinical benefits, particularly for patients with relapsed or refractory diseases. However, despite its success, the therapy faces numerous challenges and limitations, particularly concerning manufacturing and accessibility.
The manufacturing process of CAR-T cells is inherently complex and time-consuming. It involves the extraction of T cells from the patient, genetic modification to express the CAR, and subsequent expansion of these engineered cells in a controlled environment. This intricate procedure requires specialized facilities, skilled personnel, and a consistent supply of reagents, which can lead to delays and variability in the product quality. As noted by Mikhael et al. (2022), the manufacturing of CAR-T cells is dependent on timely transportation and the availability of a skilled workforce, which can pose significant logistical challenges[30].
Moreover, the high cost associated with CAR-T therapy is a major barrier to accessibility. The production of CAR-T cells involves not only the expenses related to the manufacturing process but also the costs of hospitalization and monitoring during treatment. As a result, many health insurance plans in the United States restrict coverage for these therapies, further limiting patient access. This issue is compounded by the COVID-19 pandemic, which has exacerbated existing barriers, including the availability of clinical trial slots and the complexity of treatment delivery, requiring patients to be near treatment centers and often to have caregivers available post-infusion[5][30].
Additionally, the therapy's reliance on autologous T cells presents another layer of complexity. Autologous CAR-T therapies are derived from the patient's own cells, which can be problematic in cases where patients have severely compromised health or where the collection of sufficient viable T cells is not feasible. The potential for allogeneic CAR-T cells—derived from healthy donors—could address some of these issues, yet the development and regulatory approval of such products are still in progress[2][31].
In summary, while CAR-T therapy has significantly advanced the treatment landscape for hematologic malignancies, challenges related to manufacturing complexity, high costs, and accessibility continue to hinder its widespread application. Addressing these barriers is crucial for expanding the reach of CAR-T therapies to a broader patient population, necessitating collaborative efforts among stakeholders, including healthcare providers, regulatory bodies, and the industry[5][30].
4.3 Antigen Escape and Relapse
Chimeric antigen receptor T (CAR-T) cell therapy has emerged as a transformative treatment for hematologic malignancies, particularly B cell malignancies such as acute lymphoblastic leukemia (ALL) and diffuse large B-cell lymphoma (DLBCL). This therapy involves engineering a patient's T cells to express CARs that specifically target tumor-associated antigens, leading to targeted destruction of malignant cells. Despite its remarkable success, CAR-T therapy faces significant challenges, notably antigen escape and relapse, which hinder its long-term efficacy.
Antigen escape occurs when tumor cells lose or alter the expression of the target antigens recognized by CAR-T cells. This phenomenon can arise through several mechanisms, including the enrichment of pre-existing target-negative tumor clones, mutations in the antigen gene, or alternative splicing events that result in altered antigen presentation. Other mechanisms contributing to antigen escape include deficits in antigen processing, redistribution of antigens, lineage switching of tumor cells, epitope masking, and trogocytosis-mediated antigen loss, where tumor cells acquire antigens from CAR-T cells, effectively camouflaging themselves from immune detection [32].
The implications of antigen escape are profound, as it leads to treatment resistance and subsequent relapse in patients. Approximately 50% of patients treated with CAR-T products experience relapse or refractory disease, primarily due to antigen shutdown and CAR-T cell dysfunctionality. These challenges are exacerbated in solid tumors, where antigen heterogeneity and a hostile tumor microenvironment further complicate therapeutic outcomes [33].
To combat these limitations, various strategies have been proposed. For instance, the development of multi-targeted CAR-T cells aims to address antigen heterogeneity by targeting multiple antigens simultaneously, thereby reducing the likelihood of tumor escape [7]. Furthermore, enhancing CAR-T cell persistence and functionality through advanced engineering techniques, such as incorporating costimulatory domains or utilizing bispecific antibodies, is being explored to improve therapeutic efficacy [1].
In summary, while CAR-T cell therapy represents a groundbreaking advancement in the treatment of hematologic malignancies, the challenges posed by antigen escape and relapse necessitate ongoing research and innovation. Understanding the underlying mechanisms of these phenomena is crucial for developing effective strategies to enhance the durability of responses and improve patient outcomes in this promising therapeutic landscape [34][35][36].
5 Future Directions in CAR-T Therapy
5.1 Enhancements in CAR Design
Chimeric antigen receptor T-cell (CAR-T) therapy has emerged as a groundbreaking approach in the treatment of hematologic malignancies, particularly for conditions such as acute lymphoblastic leukemia (ALL), B-cell lymphomas, and multiple myeloma. This innovative therapy involves the genetic modification of a patient’s T cells to express a CAR that specifically targets antigens present on malignant cells, thereby enabling the immune system to recognize and eliminate these cancer cells.
The treatment process begins with the collection of T cells from the patient, which are then engineered to express CARs that recognize specific tumor-associated antigens. Upon reinfusion into the patient, these CAR-T cells proliferate and exert their cytotoxic effects on the targeted malignancies. The therapy has shown remarkable efficacy, with high response rates observed in clinical settings. For instance, CAR-T therapy has been associated with complete remission rates (CRR) of 72%-100% in relapsed/refractory B-cell lymphomas and multiple myeloma, as highlighted in recent studies [5][37].
Despite its success, the application of CAR-T therapy in hematologic malignancies is not without challenges. The therapy is often accompanied by significant side effects, including cytokine release syndrome (CRS) and neurotoxicity, which necessitate careful management [2][38]. Furthermore, there are concerns regarding the durability of response and the potential for relapse, primarily due to factors such as tumor heterogeneity and the presence of immunosuppressive microenvironments [1][9].
Future directions in CAR-T therapy focus on enhancing the efficacy and safety of this treatment modality. One promising area of development involves optimizing CAR designs. Advances in CAR engineering, such as the incorporation of co-stimulatory domains and the use of dual-targeting CARs, aim to improve T-cell activation, persistence, and overall antitumor activity [38][39]. Additionally, the exploration of combination therapies, where CAR-T cells are used alongside other treatments like checkpoint inhibitors or bispecific antibodies, has shown potential in overcoming resistance and enhancing therapeutic outcomes [1][9].
Moreover, ongoing research is investigating the use of allogeneic CAR-T cells, which are derived from healthy donors, as a means to reduce the time and cost associated with autologous CAR-T therapy. This approach could expand patient access to CAR-T therapy while potentially mitigating some of the manufacturing challenges currently faced [31][37].
In summary, CAR-T therapy represents a significant advancement in the treatment of hematologic malignancies, offering hope to patients with refractory diseases. Continued innovation in CAR design and therapeutic strategies is essential to address existing challenges and improve patient outcomes in this rapidly evolving field.
5.2 Combination Therapies
Chimeric antigen receptor T-cell (CAR-T) therapy has emerged as a transformative approach in the treatment of hematologic malignancies, demonstrating significant clinical efficacy in various conditions such as acute lymphoblastic leukemia (ALL), multiple myeloma (MM), and non-Hodgkin lymphoma (NHL). This innovative therapy involves the genetic modification of a patient’s T cells to express CARs that specifically recognize and bind to tumor-associated antigens (TAAs), predominantly CD19 for B-cell malignancies. This targeted action enables CAR-T cells to effectively identify and eliminate malignant cells, leading to high response rates and prolonged disease control in a subset of patients [1].
However, while CAR-T therapy has revolutionized treatment paradigms, it is not without challenges. Monotherapy can lead to issues such as therapy resistance, disease relapse, and minimal residual disease (MRD) persistence post-remission [6]. The persistence of CAR-T cells can vary, and adverse effects such as cytokine release syndrome (CRS) and neurotoxicity are common [2]. These factors underscore the necessity for optimizing CAR-T therapy and exploring combination strategies to enhance its effectiveness.
Future directions in CAR-T therapy focus on improving patient outcomes through combination therapies. Combining CAR-T therapy with small-molecule agents has shown promising results in augmenting antitumor efficacy. For instance, the integration of Bruton's tyrosine kinase (BTK) inhibitors, hypomethylating agents, and immunomodulatory drugs can enhance the persistence and proliferative capacity of CAR-T cells, thereby improving overall therapeutic outcomes [6]. Furthermore, combining CAR-T with immune checkpoint inhibitors aims to overcome the immunosuppressive tumor microenvironment, which can limit the efficacy of CAR-T cells [40].
Clinical trials have indicated that combination therapies not only enhance the antitumor effects of CAR-T cells but also improve patient response and survival rates across various hematologic malignancies [9]. For example, combining CAR-T therapy with autologous stem cell transplantation (ASCT) has demonstrated synergistic effects, with complete remission rates reported between 72% and 100% and two-year progression-free survival rates ranging from 59% to 83% in patients with relapsed/refractory B-cell lymphomas and multiple myeloma [37].
In summary, CAR-T therapy represents a critical advancement in the treatment of hematologic malignancies, with ongoing research emphasizing the importance of combination therapies to address existing challenges. The integration of CAR-T with other therapeutic modalities is expected to enhance its efficacy, reduce relapse rates, and ultimately improve patient outcomes in this patient population. Continued exploration of these combination strategies will be vital in optimizing CAR-T therapy and expanding its applicability across a broader range of hematologic and potentially solid tumors.
5.3 Expanding Applications to Solid Tumors
Chimeric antigen receptor T-cell (CAR-T) therapy has emerged as a transformative treatment modality for hematologic malignancies, particularly in cases of relapsed or refractory diseases. The therapy involves the genetic modification of a patient's T cells to express CARs that specifically recognize and target tumor-associated antigens, such as CD19 on B cells. This mechanism enables the engineered T cells to selectively attack malignant cells, leading to significant clinical responses in conditions like acute lymphoblastic leukemia (ALL) and various forms of lymphoma [4].
The clinical application of CAR-T therapy has demonstrated remarkable efficacy, with complete remission rates ranging from 72% to 100% in trials involving relapsed/refractory B-cell lymphomas and multiple myeloma [37]. However, challenges remain, including treatment resistance, relapse, and adverse effects such as cytokine release syndrome (CRS) and neurotoxicity [5]. The success of CAR-T therapy has prompted ongoing research to enhance its efficacy and safety, focusing on optimizing CAR designs, improving T cell persistence, and developing combination therapies with small molecules and immune checkpoint inhibitors [6].
Future directions for CAR-T therapy include expanding its applications beyond hematologic malignancies to solid tumors. While CAR-T therapy has shown impressive results in hematologic cancers, its efficacy in solid tumors has been limited due to challenges such as tumor heterogeneity, an immunosuppressive microenvironment, and off-tumor toxicities [5]. Researchers are exploring innovative strategies to overcome these barriers, including advanced CAR designs that can target multiple antigens, combination therapies that integrate CAR-T with other treatment modalities, and novel approaches to modify the tumor microenvironment [39].
Moreover, the incorporation of immune checkpoint blockade into CAR-T therapy represents a promising avenue for enhancing its effectiveness against solid tumors. This strategy aims to modulate the immunosuppressive environment and improve T cell activation and persistence [40].
Overall, while CAR-T therapy has revolutionized the treatment landscape for hematologic malignancies, its expansion into solid tumors requires further investigation to address the unique challenges posed by these cancers. Continued research and clinical trials will be crucial in unlocking the full potential of CAR-T therapy across a broader spectrum of malignancies, paving the way for more effective and accessible cancer treatments in the future [9].
6 Conclusion
CAR-T therapy has significantly transformed the treatment landscape for hematologic malignancies, particularly in cases of acute lymphoblastic leukemia, diffuse large B-cell lymphoma, and multiple myeloma. The therapy leverages the patient's own immune system, employing genetically modified T cells to target specific tumor-associated antigens, resulting in impressive clinical outcomes and durable remissions. However, the implementation of CAR-T therapy is accompanied by notable challenges, including severe adverse effects such as cytokine release syndrome and neurotoxicity, as well as manufacturing complexities and accessibility issues that limit its widespread adoption. Furthermore, the phenomenon of antigen escape poses a significant barrier to long-term efficacy, necessitating ongoing research into multi-targeted CAR strategies and combination therapies to enhance treatment outcomes. Looking ahead, the future of CAR-T therapy lies in optimizing CAR designs, expanding its applications to solid tumors, and developing innovative combination approaches that can improve patient outcomes while mitigating risks. The continuous evolution of CAR-T therapy represents a beacon of hope for patients with refractory hematologic cancers and sets the stage for advancements in cancer immunotherapy.
References
- [1] 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.
- [2] 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.
- [3] 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.
- [4] Theresa Haslauer;Richard Greil;Nadja Zaborsky;Roland Geisberger. CAR T-Cell Therapy in Hematological Malignancies.. International journal of molecular sciences(IF=4.9). 2021. PMID:34445701. DOI: 10.3390/ijms22168996.
- [5] Gengtian Zhang;Mengyao Bai;Hanzhi Du;Yue Yuan;Yidan Wang;Weijing Fan;Huachao Zhu;Di Wu;Pengcheng He;Busheng Xue. Current Advances and Challenges in CAR-T Therapy for Hematological and Solid Tumors.. ImmunoTargets and therapy(IF=4.4). 2025. PMID:40599347. DOI: 10.2147/ITT.S519616.
- [6] 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.
- [7] 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.
- [8] 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.
- [9] Delian Zhou;Xiaojian Zhu;Yi Xiao. CAR-T cell combination therapies in hematologic malignancies.. Experimental hematology & oncology(IF=13.5). 2024. PMID:39026380. DOI: 10.1186/s40164-024-00536-0.
- [10] Yasser Mostafa Kamel. CAR-T Therapy, the End of a Chapter or the Beginning of a New One?. Cancers(IF=4.4). 2021. PMID:33670515. DOI: 10.3390/cancers13040853.
- [11] Aamir Ahmad. CAR-T Cell Therapy.. International journal of molecular sciences(IF=4.9). 2020. PMID:32560285. DOI: 10.3390/ijms21124303.
- [12] Craig W Freyer;David L Porter. Advances in CAR T Therapy for Hematologic Malignancies.. Pharmacotherapy(IF=3.4). 2020. PMID:32383222. DOI: 10.1002/phar.2414.
- [13] Omar Castaneda Puglianini;Julio C Chavez. CARs Moving Forward: The Development of CAR T-Cell Therapy in the Earlier Treatment Course of Hematologic Malignancies.. Seminars in hematology(IF=4.1). 2024. PMID:39306480. DOI: 10.1053/j.seminhematol.2024.08.005.
- [14] Le Wang. Clinical determinants of relapse following CAR-T therapy for hematologic malignancies: Coupling active strategies to overcome therapeutic limitations.. Current research in translational medicine(IF=3.0). 2022. PMID:34768218. DOI: 10.1016/j.retram.2021.103320.
- [15] Alessia Zinzi;Mario Gaio;Valerio Liguori;Cecilia Cagnotta;Donatella Paolino;Giuseppe Paolisso;Giuseppe Castaldo;Giovanni Francesco Nicoletti;Francesco Rossi;Annalisa Capuano;Concetta Rafaniello. Late relapse after CAR-T cell therapy for adult patients with hematologic malignancies: A definite evidence from systematic review and meta-analysis on individual data.. Pharmacological research(IF=10.5). 2023. PMID:36963592. DOI: 10.1016/j.phrs.2023.106742.
- [16] Assia Hijazi;Frederick L Locke;Nathalie Scholler;Mike Mattie;Simone Filosto;Davide Bedognetti;Jérôme Galon. Facts and Hopes: CAR T-Cell Therapy and Immune Contexture in Non-Hodgkin Lymphoma.. Clinical cancer research : an official journal of the American Association for Cancer Research(IF=10.2). 2025. PMID:40622821. DOI: 10.1158/1078-0432.CCR-24-2267.
- [17] Binod Dhakal;Parameswaran N Hari;Saad Z Usmani;Mehdi Hamadani. Chimeric antigen receptor T cell therapy in multiple myeloma: promise and challenges.. Bone marrow transplantation(IF=5.2). 2021. PMID:32770147. DOI: 10.1038/s41409-020-01023-w.
- [18] Lekha Mikkilineni;James N Kochenderfer. Chimeric antigen receptor T-cell therapies for multiple myeloma.. Blood(IF=23.1). 2017. PMID:28928126. DOI: 10.1182/blood-2017-06-793869.
- [19] Shihui Yuan;Ying Chen;Huasheng Liu. Progress and prospect of ASCT combined with CAR-T therapy in the treatment of multiple myeloma.. Therapeutic advances in hematology(IF=3.1). 2024. PMID:38481949. DOI: 10.1177/20406207241237594.
- [20] Mariam Markouli;Fauzia Ullah;Serhan Unlu;Najiullah Omar;Nerea Lopetegui-Lia;Marissa Duco;Faiz Anwer;Shahzad Raza;Danai Dima. Toxicity Profile of Chimeric Antigen Receptor T-Cell and Bispecific Antibody Therapies in Multiple Myeloma: Pathogenesis, Prevention and Management.. Current oncology (Toronto, Ont.)(IF=3.4). 2023. PMID:37504327. DOI: 10.3390/curroncol30070467.
- [21] Faroogh Marofi;Safa Tahmasebi;Heshu Sulaiman Rahman;Denis Kaigorodov;Alexander Markov;Alexei Valerievich Yumashev;Navid Shomali;Max Stanley Chartrand;Yashwant Pathak;Rebar N Mohammed;Mostafa Jarahian;Roza Motavalli;Farhad Motavalli Khiavi. Any closer to successful therapy of multiple myeloma? CAR-T cell is a good reason for optimism.. Stem cell research & therapy(IF=7.3). 2021. PMID:33781320. DOI: 10.1186/s13287-021-02283-z.
- [22] Xiaomin Zhang;Lingling Zhu;Hui Zhang;Shanshan Chen;Yang Xiao. CAR-T Cell Therapy in Hematological Malignancies: Current Opportunities and Challenges.. Frontiers in immunology(IF=5.9). 2022. PMID:35757715. DOI: 10.3389/fimmu.2022.927153.
- [23] 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.
- [24] Yanping Li;Yue Ming;Ruoqiu Fu;Chen Li;Yuanlin Wu;Tingting Jiang;Ziwei Li;Rui Ni;Li Li;Hui Su;Yao Liu. The pathogenesis, diagnosis, prevention, and treatment of CAR-T cell therapy-related adverse reactions.. Frontiers in pharmacology(IF=4.8). 2022. PMID:36313336. DOI: 10.3389/fphar.2022.950923.
- [25] Kiril Dimitrov;Florian Merkle;Marketa Dimitrov;Svatava Merkle;Alex Hoover;Veronika Bachanova. Major Adverse Events with Chimeric Antigen Receptor T-Cell Therapy: Presentation, Diagnosis, and Resuscitation.. Annals of emergency medicine(IF=5.0). 2025. PMID:40817894. DOI: 10.1016/j.annemergmed.2025.07.005.
- [26] 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.
- [27] Mengyi Du;Linlin Huang;Haiming Kou;Chenggong Li;Yu Hu;Heng Mei. Case Report: ITP Treatment After CAR-T Cell Therapy in Patients With Multiple Myeloma.. Frontiers in immunology(IF=5.9). 2022. PMID:35784357. DOI: 10.3389/fimmu.2022.898341.
- [28] Tony Joseph;Jimmy Sanchez;Ahmed Abbasi;Lili Zhang;R Alejandro Sica;Tim Q Duong. Cardiotoxic Effects Following CAR-T Cell Therapy: A Literature Review.. Current oncology reports(IF=5.0). 2025. PMID:39836349. DOI: 10.1007/s11912-024-01634-2.
- [29] Zhaozhao Chen;Yu Hu;Heng Mei. Harnessing Biomaterials for Safeguarding Chimeric Antigen Receptor T Cell Therapy: An Artful Expedition in Mitigating Adverse Effects.. Pharmaceuticals (Basel, Switzerland)(IF=4.8). 2024. PMID:38276012. DOI: 10.3390/ph17010139.
- [30] Joseph Mikhael;Jessica Fowler;Nina Shah. Chimeric Antigen Receptor T-Cell Therapies: Barriers and Solutions to Access.. JCO oncology practice(IF=4.6). 2022. PMID:36130152. DOI: 10.1200/OP.22.00315.
- [31] Massimo Martino;Virginia Naso;Barbara Loteta;Filippo Antonio Canale;Marta Pugliese;Caterina Alati;Gerardo Musuraca;Davide Nappi;Anna Gaimari;Fabio Nicolini;Massimiliano Mazza;Sara Bravaccini;Daniele Derudas;Giovanni Martinelli;Claudio Cerchione. Chimeric Antigen Receptor T-Cell Therapy: What We Expect Soon.. International journal of molecular sciences(IF=4.9). 2022. PMID:36362130. DOI: 10.3390/ijms232113332.
- [32] Haolong Lin;Xiuxiu Yang;Shanwei Ye;Liang Huang;Wei Mu. Antigen escape in CAR-T cell therapy: Mechanisms and overcoming strategies.. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie(IF=7.5). 2024. PMID:39098176. DOI: 10.1016/j.biopha.2024.117252.
- [33] Dennis Christoph Harrer;Jan Dörrie;Niels Schaft. CARs and Drugs: Pharmacological Ways of Boosting CAR-T-Cell Therapy.. International journal of molecular sciences(IF=4.9). 2023. PMID:36768665. DOI: 10.3390/ijms24032342.
- [34] Wanying Zeng;Pumin Zhang. Resistance and recurrence of malignancies after CAR-T cell therapy.. Experimental cell research(IF=3.5). 2022. PMID:34906583. DOI: 10.1016/j.yexcr.2021.112971.
- [35] Yufei Lu;Fu Zhao. Strategies to overcome tumour relapse caused by antigen escape after CAR T therapy.. Molecular cancer(IF=33.9). 2025. PMID:40289115. DOI: 10.1186/s12943-025-02334-6.
- [36] Ikram Salih;Meryem Fakhkhari;Hicham Berrougui;Abdelouahed Khalil;Karim Benabdellah;Erden Atilla;Khalid Sadki. HLA molecules: Another challenge for CAR T cell therapy.. Current research in translational medicine(IF=3.0). 2025. PMID:41086595. DOI: 10.1016/j.retram.2025.103547.
- [37] Yixin Yan;Zigang Dai;Dengju Li;Xia Mao;Liang Huang. Bridging transplantation and immunotherapy: Clinical promise of autologous stem cell transplantation with chimeric antigen receptor T-cell therapy.. Chinese journal of cancer research = Chung-kuo yen cheng yen chiu(IF=6.3). 2025. PMID:40979669. DOI: 10.21147/j.issn.1000-9604.2025.04.03.
- [38] Yajing Zhang;Yang Xu;Xiuyong Dang;Zeyu Zhu;Wenbin Qian;Aibin Liang;Weidong Han. Challenges and optimal strategies of CAR T therapy for hematological malignancies.. Chinese medical journal(IF=7.3). 2023. PMID:36848181. DOI: 10.1097/CM9.0000000000002476.
- [39] Alejandrina Hernández-López;Alberto Olaya-Vargas;Juan Carlos Bustamante-Ogando;Angélica Meneses-Acosta. Expanding the Horizons of CAR-T Cell Therapy: A Review of Therapeutic Targets Across Diverse Diseases.. Pharmaceuticals (Basel, Switzerland)(IF=4.8). 2025. PMID:40005970. DOI: 10.3390/ph18020156.
- [40] Dok Hyun Yoon;Mark J Osborn;Jakub Tolar;Chong Jai Kim. Incorporation of Immune Checkpoint Blockade into Chimeric Antigen Receptor T Cells (CAR-Ts): Combination or Built-In CAR-T.. International journal of molecular sciences(IF=4.9). 2018. PMID:29364163. DOI: 10.3390/ijms19020340.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
CAR-T therapy · hematologic malignancies · tumor antigens
© 2025 MaltSci
