Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,165)

Search Parameters:
Keywords = chimeric antigen receptor

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
4 pages, 164 KB  
Editorial
Targeting CAR T-Cell Therapy: Molecular Research and Its Future Implication
by Stella Bouziana
Int. J. Mol. Sci. 2025, 26(22), 10868; https://doi.org/10.3390/ijms262210868 - 9 Nov 2025
Viewed by 676
Abstract
The emergence of chimeric antigen receptor (CAR) T-cell therapy represents a significant milestone in the cellular treatment of cancer [...] Full article
13 pages, 610 KB  
Article
High Rate of Cytokine Release Syndrome-Related Coagulopathy with Low Incidence of Bleeding and Thrombosis in Patients Treated with B-Cell Maturation Antigen (BCMA)-Targeted Chimeric Antigen Receptor T-Cells (CAR-T)
by Ariela Arad, Maya Katz, Eyal Lebel, Yosef Kalish, Miri Assayag, Batia Avni, Shlomo Elias, Sigal Grisariu, Ela Shai, Shlomit Kfir-Erenfeld, Nathalie Asherie, Moshe E. Gatt, Polina Stepensky and Eran Zimran
Cancers 2025, 17(21), 3551; https://doi.org/10.3390/cancers17213551 - 2 Nov 2025
Viewed by 265
Abstract
Background: B-cell maturation antigen (BCMA)-targeted chimeric antigen receptor T-cell (CAR-T) therapy has demonstrated substantial efficacy in relapsed and/or refractory multiple myeloma. While toxicities such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) have been well characterized, the incidence [...] Read more.
Background: B-cell maturation antigen (BCMA)-targeted chimeric antigen receptor T-cell (CAR-T) therapy has demonstrated substantial efficacy in relapsed and/or refractory multiple myeloma. While toxicities such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) have been well characterized, the incidence and clinical consequences of the coagulopathy associated with CRS remain underexplored. Methods: We conducted a prospective analysis of 108 adult patients with multiple myeloma or light chain amyloidosis treated with the academic anti-BCMA CAR-T HBI0101 in a single-center trial (NCT04720313). Coagulopathy was evaluated via serial fibrinogen measurements, with hypofibrinogenemia defined as <200 mg/dL and severe coagulopathy as <100 mg/dL. Laboratory markers, tocilizumab and blood product use, and thrombotic and bleeding complications were recorded. Patients received a short (3-day) or extended course of enoxaparin thromboprophylaxis as well as fresh frozen plasma in cases of severe coagulopathy. Results: CRS grades 1–3 occurred in 100 patients (93%). Hypofibrinogenemia was observed in 79 patients (73%), including 20 (19%) with severe coagulopathy. Fibrinogen levels were significantly associated with CRS severity (p < 0.001), number of tocilizumab doses (p < 0.001), peak levels of the inflammation markers LDH (p = 0.001) and ferritin (p = 0.006), and neutropenia (p = 0.33). Five thrombotic events (4.6%) and three minor bleeding events (2.7%) occurred within 3 months post-CAR-T infusion and were not associated with degree of coagulopathy or CRS. No cases of major bleeding or fatal thrombosis occurred. Conclusions: CRS-related coagulopathy is common following BCMA-targeted CAR-T treatment and correlates closely with CRS severity. Despite the high rate of laboratory coagulopathy, thrombosis and bleeding events were infrequent, suggesting the benefit of the prophylactic strategies used. Full article
(This article belongs to the Special Issue CAR T-Cell Therapy and Multiple Myeloma)
Show Figures

Figure 1

16 pages, 1568 KB  
Review
CAR-T Cell Therapy in Autoimmune Diseases: Promise, Progress, and Pitfalls
by Alessandro Conforti, Carlos Cifuentes-González, Alarico Ariani, Alberto Lo Gullo and Rupesh Agrawal
Rheumato 2025, 5(4), 15; https://doi.org/10.3390/rheumato5040015 - 31 Oct 2025
Viewed by 285
Abstract
Background: Chimeric Antigen Receptor T-cell (CAR-T) cell therapy has revolutionized cancer treatment and is now being explored as a novel approach to treat refractory autoimmune diseases by targeting autoreactive immune components, especially B cells. Objective: Our aim was to provide a narrative review [...] Read more.
Background: Chimeric Antigen Receptor T-cell (CAR-T) cell therapy has revolutionized cancer treatment and is now being explored as a novel approach to treat refractory autoimmune diseases by targeting autoreactive immune components, especially B cells. Objective: Our aim was to provide a narrative review of the current evidence, mechanisms, efficacy, safety, and future directions of CAR-T cell therapy in autoimmune diseases. Methods: A structured literature search was conducted in MEDLINE via PubMed using keywords such as “CAR-T”, “chimeric antigen receptor T-cell”, “autoimmune diseases”, “lupus”, “rheumatoid arthritis”, “multiple sclerosis”, and “vasculitis”. Studies on CAR-T mechanisms, efficacy, safety, and clinical outcomes were included. Results: CAR-T cell therapies, especially CD19-directed constructs, demonstrated sustained drug-free remission in all patients across early SLE case series (n = 5–7), with normalization of serological markers and improved renal outcomes. Emerging preclinical and early clinical data in rheumatoid arthritis, multiple sclerosis, ANCA-associated vasculitis, juvenile autoimmune diseases, and idiopathic inflammatory myopathies also report clinical improvement and biomarker normalization. Reported adverse events in autoimmune cohorts were limited to mild cytokine release syndrome in a minority of cases, with no severe neurotoxicity or life-threatening infections, suggesting a more favorable safety profile compared to oncology settings. In parallel, next-generation innovations—including dual-target CARs, CAR-Tregs, and molecular safety switches—are advancing toward clinical translation. Conclusions: CAR-T cell therapy is emerging as a transformative strategy for autoimmune disease management, especially in refractory cases. Although initial outcomes are promising, long-term safety, cost-effectiveness, and broader accessibility remain key challenges. Future research should focus on optimizing cell targets, minimizing off-target effects, and improving affordability. Full article
Show Figures

Figure 1

37 pages, 1441 KB  
Review
Soil and Seed: Tumor Microenvironment Nurtures Immunotherapy Resistance and Renewal
by Yiya Li, Qiushi Feng, Yangyang Xia, Lingzi Liao and Shang Xie
Int. J. Mol. Sci. 2025, 26(21), 10547; https://doi.org/10.3390/ijms262110547 - 30 Oct 2025
Viewed by 423
Abstract
Cancer immunotherapy has become a powerful clinical strategy for cancer management, while its efficacy is frequently limited by primary and acquired resistance. The tumor microenvironment (TME) plays a pivotal role in mediating such resistance through multifaceted mechanisms involving cellular, metabolic, mechanical, and microbial [...] Read more.
Cancer immunotherapy has become a powerful clinical strategy for cancer management, while its efficacy is frequently limited by primary and acquired resistance. The tumor microenvironment (TME) plays a pivotal role in mediating such resistance through multifaceted mechanisms involving cellular, metabolic, mechanical, and microbial components. This review systematically examines how the TME contributes to immunotherapy failure. We compare resistance mechanisms common to both immune checkpoint inhibitors (ICIs) and chimeric antigen receptor T-cell (CAR-T) therapies, two cornerstone modalities in clinical practice. Furthermore, we discuss emerging strategies designed to overcome these barriers, including immune microenvironment, stromal normalization, metabolic modulation, and microbiota engineering. By integrating recent preclinical and clinical insights, this review aims to provide a comprehensive framework for understanding and targeting microenvironmental resistance, ultimately facilitating the translation of novel combination therapies into improved patient outcomes. Full article
Show Figures

Figure 1

31 pages, 1143 KB  
Review
Current Status of Molecularly Targeted Therapeutics in Blood Cancers
by Caitlin Kumala, Lynh Vu and Tamer E. Fandy
Int. J. Mol. Sci. 2025, 26(21), 10512; https://doi.org/10.3390/ijms262110512 - 29 Oct 2025
Viewed by 620
Abstract
Blood cancer is characterized by the uncontrolled growth of blood cells in the bone marrow or in the lymphatic system. Chemotherapy is still considered the first line of treatment in several types of blood cancer despite its adverse effects. Recent advances in understanding [...] Read more.
Blood cancer is characterized by the uncontrolled growth of blood cells in the bone marrow or in the lymphatic system. Chemotherapy is still considered the first line of treatment in several types of blood cancer despite its adverse effects. Recent advances in understanding the pathology and genomic changes in these cancers have led to the discovery of novel drug targets and the development of new therapeutic agents. In this review, we will discuss the mechanisms of action and clinical utility of several classes of targeted therapy used in blood cancers, including inhibitors of different types of tyrosine kinase enzymes (BCR-ABL, FLT3 and BTK), BCL-2 inhibitors, phosphoinositide 3-kinase inhibitors, nuclear export inhibitors, immune therapies (monoclonal antibodies, radioimmunoconjugates, chimeric antigen receptor T-cells, immune checkpoint inhibitors, and bispecific antibodies), and proteasome-dependent drugs (proteasome inhibitors and proteolysis targeting chimeras). Further advances in identifying distinct molecular subgroups in blood cancers will offer more opportunities for novel targeted therapies and more personalized medicine approaches. Full article
Show Figures

Figure 1

19 pages, 340 KB  
Review
Mechanisms of Resistance to Novel Immunotherapies in B-Cell Lymphomas: Focus on CAR T and Bispecific Antibodies
by Gloria Arena and Roberto Chiarle
Cancers 2025, 17(21), 3453; https://doi.org/10.3390/cancers17213453 - 28 Oct 2025
Viewed by 658
Abstract
Treatment paradigms for B-cell lymphomas have evolved significantly in the last decades. Nevertheless, the widespread clinical use of immunotherapy has demonstrated that it invariably leads to the development of resistance. This review outlines the underlying molecular mechanisms of resistance associated with emerging immunotherapeutic [...] Read more.
Treatment paradigms for B-cell lymphomas have evolved significantly in the last decades. Nevertheless, the widespread clinical use of immunotherapy has demonstrated that it invariably leads to the development of resistance. This review outlines the underlying molecular mechanisms of resistance associated with emerging immunotherapeutic strategies, including Chimeric Antigen Receptor (CAR) T cell therapy and bispecific antibodies (BsAbs). In high-grade B-cell lymphomas, nearly 50% of patients progress following CAR T treatment due to host-related factors affecting CAR T cell proliferation and persistence, as well as tumor-intrinsic factors, such as loss of CD19 epitope expression, trogocytosis, and other genomic alterations (e.g., CD19 mutations, chromothripsis, APOBEC mutational activity, and deletions of RHOA). Additional genomic and epigenetic events, including mutations, alternative splicing of CD19, and aberrant promoter methylation, further contribute to resistance. BsAbs, representing an off-the-shelf T-cell-redirecting strategy, have recently shown promising single-agent efficacy with a manageable toxicity profile, predominantly characterized by T cell overactivation syndromes. Similarly to CAR T cell therapy, BsAb resistance arises through diverse mechanisms, such as antigen loss, T cell dysfunction (exhaustion and regulatory T cell activation), tumor-intrinsic alterations (e.g., TP53 mutations and MYC amplifications), and immunosuppressive influences from the tumor microenvironment. These findings underscore the complexity of immune evasion in B-cell lymphomas and highlight the ongoing need to optimize immunotherapeutic strategies and develop combination approaches to overcome resistance. Full article
(This article belongs to the Special Issue Advances in B-Cell Lymphoma: From Diagnostics to Cure)
Show Figures

Graphical abstract

10 pages, 2295 KB  
Communication
CD5 Expression in CTCL and Its Implications for Anti-CD5 CAR T-Cell Therapy
by Leena Wardeh, Madeline Williams, Courtney Prestwood, Zachary Wolner and Neda Nikbakht
Int. J. Mol. Sci. 2025, 26(21), 10411; https://doi.org/10.3390/ijms262110411 - 27 Oct 2025
Viewed by 346
Abstract
Cutaneous T-Cell Lymphomas (CTCL) are a heterogenous group of T-cell malignancies in the skin and have poor treatment outcomes in advanced stages. CD5, a surface glycoprotein expressed on most mature T cells, has emerged as a promising target for chimeric antigen receptor (CAR) [...] Read more.
Cutaneous T-Cell Lymphomas (CTCL) are a heterogenous group of T-cell malignancies in the skin and have poor treatment outcomes in advanced stages. CD5, a surface glycoprotein expressed on most mature T cells, has emerged as a promising target for chimeric antigen receptor (CAR) T-cell therapy in systemic T-cell lymphomas. However, its expression profile in CTCL and relevance for targeted therapy remain unclear. Notably, in CTCL, the cell surface expression of receptors, such as CD7 and CD26, tends to become downregulated on the surfaces of malignant T cells In this study, we analyzed single-cell RNA sequencing (scRNA-seq) data from patients at two institutions with mycosis fungoides (MF), the most common subtype of CTCL with a predominantly CD4 phenotype. We utilized 5 patch/plaque MF skin biopsies (majority from early-stage patients), 8 MF tumor biopsies (all from advanced-stage patients), and 8 healthy control biopsies to evaluate lesion-specific CD5 gene expression on CD4 T cells. We found that CD5 was significantly increased in malignant MF CD4 T cells compared to healthy control CD4 T cells (21.1% of MF CD4 T cells expressed CD5 vs. 5.2% of healthy control CD4 T cells, respectively). In subgroup analysis, patch/plaque stage MF biopsies showed higher expression of CD5 in CD4 T cells than tumor stage MF biopsies. Notably, 94.3% of malignant CD4+ T cells in tumor stage MF lesions exhibited complete CD5 loss compared to only 76.6% in patch-plaque MF lesions, suggesting antigen escape in tumor stage disease. These findings demonstrate that CD5 expression in CTCL is dynamic and varies based on lesion type. Our work suggests CD5 may be a viable therapeutic target in MF with patch/plaque presentations but may not be as effective in advanced stages of MF with tumor presentations. This work informs CD5 gene expression in MF based on clinical lesion type and further information is needed to clarify clinical implications as a future therapeutic target. Full article
(This article belongs to the Special Issue Study on the Microenvironment in Lymphoma)
Show Figures

Figure 1

16 pages, 254 KB  
Review
Advanced Neuroimaging and Emerging Systemic Therapies in Glioblastoma: Current Evidence and Future Directions
by Ilona Bar-Letkiewicz, Anna Pieczyńska, Małgorzata Dudzic, Michał Szkudlarek, Krystyna Adamska and Katarzyna Hojan
Biomedicines 2025, 13(11), 2597; https://doi.org/10.3390/biomedicines13112597 - 23 Oct 2025
Viewed by 753
Abstract
Despite technological progress, glioblastoma (GBM) continues to confer dismal prognoses. Modern neuroimaging methods are assuming an ever greater role in diagnosing and monitoring brain tumors. This review shows current neuroimaging approaches and systemic therapeutic strategies for glioblastoma, with a focus on emerging and [...] Read more.
Despite technological progress, glioblastoma (GBM) continues to confer dismal prognoses. Modern neuroimaging methods are assuming an ever greater role in diagnosing and monitoring brain tumors. This review shows current neuroimaging approaches and systemic therapeutic strategies for glioblastoma, with a focus on emerging and innovative treatments. Advances in multiparametric magnetic resonance imaging—MRI (diffusion, perfusion, and spectroscopy) and novel positron emission tomography (PET) tracers, complemented by radiomics and artificial intelligence (AI), now refine tumor delineation, differentiate progression from treatment effects, and may help predict treatment responses. Maximal safe resection followed by chemoradiotherapy with temozolomide remains the standard, with the greatest benefit seen in O6-methylguanine DNA methyltransferase (MGMT) promoter-methylated tumors. Bevacizumab and other targeted modalities offer mainly progression-free, not overall survival, gains. Immune checkpoint inhibitors (e.g., nivolumab) have not improved survival in unselected GBM, while early multi-antigen CAR-T (chimeric antigen receptor T-cell) strategies show preliminary bioactivity without established durability. While actionable alterations (NTRK fusions and BRAF V600E) justify selective targeted therapy trials, their definitive benefit in classical GBM is unproven. Future priorities include harmonized imaging molecular integration, AI-driven prognostic modeling, novel PET tracers, and strategies to breach or transiently open the blood–brain barrier to enhance drug delivery. Convergence of these domains may convert diagnostic precision into improved patient outcomes. Full article
(This article belongs to the Special Issue Medical Imaging in Brain Tumor: Charting the Future)
30 pages, 1988 KB  
Review
Regulatory T Cell-Based Adoptive Cell Therapy in Autoimmunity
by Eduardo Gozálvez, Adrián Lario, Guillermo Muñoz-Sánchez and Francisco Lozano
Int. J. Mol. Sci. 2025, 26(21), 10340; https://doi.org/10.3390/ijms262110340 - 23 Oct 2025
Viewed by 1635
Abstract
Regulatory T cells (Tregs) are a distinctive subset of CD4+ T cells critical in self-tolerance maintenance to prevent the development of autoimmunity. The mechanisms by which these cells provide immune regulation are numerous and, consequently, deeply involved in the pathogenesis of many [...] Read more.
Regulatory T cells (Tregs) are a distinctive subset of CD4+ T cells critical in self-tolerance maintenance to prevent the development of autoimmunity. The mechanisms by which these cells provide immune regulation are numerous and, consequently, deeply involved in the pathogenesis of many autoimmune disorders. Treg-based adoptive cell transfer (ACT) therapy has generated interest as a novel, promising strategy to restore self-tolerance in autoimmunity. Polyclonal Treg-based ACT therapy was first implemented in clinical trials, presenting adequate safety profiles. Subsequent preclinical studies have shown antigen-specific Tregs to be safer and more effective than polyclonal approaches, so research has recently moved in this direction. Antigen-specificity can be conferred to Tregs by viral transduction of genes coding for engineered T cell receptors (eTCRs) or chimeric antigen receptors (CARs), with encouraging outcomes in different animal models of autoimmunity. This review focuses on the biology of Tregs, as well as on current preclinical and clinical data for Treg-based ACT in the field of autoimmunity. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

45 pages, 3535 KB  
Review
CAR-T Cell Therapy for Prostate Cancer: Current Advances and Future Perspectives
by Maria Luisa Calabrò, Roberta Ettari, Carla Di Chio, Fabiola De Luca, Santo Previti and Maria Zappalà
Biomedicines 2025, 13(10), 2545; https://doi.org/10.3390/biomedicines13102545 - 18 Oct 2025
Viewed by 1286
Abstract
Prostate cancer is the most frequently diagnosed solid-organ malignancy in men worldwide. Metastatic castration-resistant prostate cancer represents a rapidly fatal, end-stage form of the disease for which current therapies remain palliative rather than curative. The advent of chimeric antigen receptor (CAR) T-cell therapy [...] Read more.
Prostate cancer is the most frequently diagnosed solid-organ malignancy in men worldwide. Metastatic castration-resistant prostate cancer represents a rapidly fatal, end-stage form of the disease for which current therapies remain palliative rather than curative. The advent of chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment of refractory hematologic malignancies, and a growing number of studies are now exploring its potential in solid tumors. In this review, we first provide a concise overview of current immunotherapeutic strategies for prostate cancer, including checkpoint inhibitors, vaccine-based approaches, and bispecific antibodies. We then focus on the most recent and promising developments in CAR-T cell therapy for this malignancy. Specifically, we examine the key tumor-associated antigens targeted in prostate cancer-directed CAR-T cell therapy and summarize findings from preclinical research as well as ongoing and completed clinical trials. Finally, we discuss the main challenges that limit the efficacy of CAR-T therapy in prostate cancer, such as antigen heterogeneity, immunosuppressive tumor microenvironments, on-target/off-tumor toxicity, limited T-cell persistence, and inefficient trafficking to metastatic lesions, and outline potential strategies to overcome these barriers. Our aim is to define a translational roadmap for advancing CAR-T therapy toward clinical application in patients with metastatic castration-resistant prostate cancer. Full article
(This article belongs to the Special Issue Diagnostic and Therapeutic Challenges of CAR-T Cell Therapy)
Show Figures

Graphical abstract

29 pages, 1625 KB  
Review
Finding the Sweet Spot for the Treatment of B Cell Malignancies
by Valerie R. Wiersma
Cancers 2025, 17(20), 3366; https://doi.org/10.3390/cancers17203366 - 18 Oct 2025
Viewed by 578
Abstract
The glycan profile of cells comprises a high variety of sugar moieties that are attached to proteins (glycoproteins) and lipids (glycolipids) via a process called ‘glycosylation’. Cancer cells commonly carry aberrant glycans, which may be of interest for cancer diagnosis, prognosis, as well [...] Read more.
The glycan profile of cells comprises a high variety of sugar moieties that are attached to proteins (glycoproteins) and lipids (glycolipids) via a process called ‘glycosylation’. Cancer cells commonly carry aberrant glycans, which may be of interest for cancer diagnosis, prognosis, as well as the development of novel therapeutic strategies. This review focuses on the differential glycosylation patterns on malignant B cells, including both B cell lymphoma and leukemia. Well-known aberrant glycan profiles on malignant B cells include acquired high mannose N-glycans in the B cell receptor (BCR) of follicular lymphoma (FL), and increased expression of the glycosphingolipid Gb3/CD77 on Burkitt’s lymphoma (BL). These structures can be exploited for therapy by using lectins that specifically recognize these patterns with intrinsic cytotoxic activity or in a drug-conjugate format. Furthermore, immunotherapy can be improved by modulating glycans, especially sialic acids. Targeting glycans for immunotherapy is also of interest for chimeric antigen receptor (CAR) T cell therapy, a relatively novel therapy that has been quite effective in various B cell malignancies. Thus, the glycan profile of malignant B cells harbors many opportunities for therapeutic targeting. It is anticipated that further in-depth glycan profiling will open up many more opportunities for the treatment of B cell malignancies. Full article
(This article belongs to the Special Issue Oncology: State-of-the-Art Research in The Netherlands)
Show Figures

Figure 1

15 pages, 1293 KB  
Review
CAR-T Cell Therapy for Autoimmune Kidney Diseases: Where Do We Stand Now?
by Beata Kaleta
Int. J. Mol. Sci. 2025, 26(20), 10070; https://doi.org/10.3390/ijms262010070 - 16 Oct 2025
Viewed by 649
Abstract
Autoimmune kidney diseases (AIKDs) are a consequence of the dysregulation of immune response and the loss of tolerance to self-antigens, which led to glomerulonephritis and tissue damage. Autoantibody-producing B cells, as well as T cells, neutrophils and macrophages play a pivotal role in [...] Read more.
Autoimmune kidney diseases (AIKDs) are a consequence of the dysregulation of immune response and the loss of tolerance to self-antigens, which led to glomerulonephritis and tissue damage. Autoantibody-producing B cells, as well as T cells, neutrophils and macrophages play a pivotal role in the pathogenesis and progression of various AIKDs. In recent years, B cell-depleting/modulating therapies and molecules that modulate T cell differentiation pathways and cytokine production have become a new hope for patients with immune-mediated kidney diseases. However, these biologicals often do not bring satisfactory therapeutic benefits, which is most likely related to incomplete B cell depletion of tissue-resident B cells. A new hope is immunotherapy with chimeric antigen receptor (CAR) effector cells. In CAR therapy, immune cells (mostly T cells) are genetically modified to express a CAR, which enables the recognition of the specific antigen on a target cell. This interaction leads to the formation of immune synapse and cytotoxicity. CAR-based strategies are a potent form of cell therapy that offers a better chance for deep and durable response than other recently approved immune therapies. Moreover, CAR-T cells can be programmed for higher precision and safety. This review explores the current landscape of CAR-T cell therapy in AIKDs. Full article
(This article belongs to the Special Issue Chimeric Antigen Receptors Against Cancers and Autoimmune Diseases)
Show Figures

Figure 1

12 pages, 1461 KB  
Article
Clinical Impact of LAG3 Single-Nucleotide Polymorphism in DLBCL Treated with CAR-T Cell Therapy
by Katja Seipel, Sophia Maria Spahr, Inna Shaforostova, Ulrike Bacher, Henning Nilius and Thomas Pabst
Int. J. Mol. Sci. 2025, 26(20), 9905; https://doi.org/10.3390/ijms26209905 - 11 Oct 2025
Viewed by 485
Abstract
Lymphocyte-activation gene 3 (LAG3) is an immune checkpoint receptor and inhibitory regulator of T-cells. Here, we analyzed the prevalence of LAG3 rs870849 in B-cell lymphoma patients and the treatment outcomes according to the LAG3 genetic background and discovered that LAG3 germline [...] Read more.
Lymphocyte-activation gene 3 (LAG3) is an immune checkpoint receptor and inhibitory regulator of T-cells. Here, we analyzed the prevalence of LAG3 rs870849 in B-cell lymphoma patients and the treatment outcomes according to the LAG3 genetic background and discovered that LAG3 germline variants may affect the risk of developing lymphoma and also affect the treatment outcome of DLBCL patients in the current CD19 CAR-T cell therapies. The LAG3 rs870849 was prevalent at high frequency in DLBCL patients. Significant differences in treatment outcomes to CAR-T cell therapy emerged in LAG3 I455hom versus I455Thet and T455hom carriers. The overall and complete response rates to CAR-T cell therapy were lower in the I455hom genetic subgroup with median PFS in the I455hom of 2 versus 20 months in the T455hom and I455Thet subgroups (p = 0.025). Median OS was 6 months in the LAG3 I455hom versus 41 months in the T455hom and I455Thet subgroups (p = 0.007). LAG3 rs870849 may affect treatment outcome in CAR-T cell therapy, with favorable outcomes in T455 carriers. Specific combinations of CTLA4 and LAG3 germline variants may cooperate to affect the response to CAR-T cell therapy. Full article
(This article belongs to the Special Issue Immune Cell Therapy and Genome Engineering)
Show Figures

Graphical abstract

32 pages, 1122 KB  
Review
Bispecific Monoclonal Antibodies in Diffuse Large B-Cell Lymphoma: Dawn of a New Era in Targeted Therapy
by Mattia Schipani, Matteo Bellia, Carola Sella, Riccardo Dondolin, Mariangela Greco, Abdurraouf Mokhtar Mahmoud, Clara Deambrogi, Riccardo Moia, Gianluca Gaidano and Riccardo Bruna
Cancers 2025, 17(19), 3258; https://doi.org/10.3390/cancers17193258 - 8 Oct 2025
Viewed by 2327
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common aggressive non-Hodgkin lymphoma (NHL) worldwide. Currently, approximately sixty percent of patients are cured with R-CHOP as frontline treatment, while the remaining patients experience primary refractory or relapsed (R/R) disease. Recently, the introduction of Pola-R-CHP [...] Read more.
Diffuse large B-cell lymphoma (DLBCL) is the most common aggressive non-Hodgkin lymphoma (NHL) worldwide. Currently, approximately sixty percent of patients are cured with R-CHOP as frontline treatment, while the remaining patients experience primary refractory or relapsed (R/R) disease. Recently, the introduction of Pola-R-CHP as front-line therapy has represented a major advance in the management of DLBCL, resulting in improved outcomes. Prognosis of R/R DLBCL patients is poor, particularly for those eligible neither for chimeric antigen receptor (CAR) T-cell therapy nor autologous stem cell transplantation (ASCT), representing a significant unmet clinical need. The advent of bispecific monoclonal antibodies (BsAbs), such as bispecific T-cell engagers (BiTEs), dual affinity retargeting (DART) molecules and IgG-like bispecific antibodies, offers a novel promising therapeutic approach in the treatment of DLBCL, both as frontline treatment and in the R/R setting. BsAbs simultaneously engage two different antigens, a tumor-associated antigen and an immune cell antigen, redirecting T-cells against malignant cells and enhancing the immune response. Most BsAbs developed for the treatment of NHLs engage T-cells via CD3 and malignant B-cells via CD20, a surface antigen expressed on most lymphomatous cells. Engagement of malignant B-cells by BsAbs activates T-cells, leading to the release of multiple cytokines and potentially to two characteristic adverse events: cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). The most extensively studied BsAbs, in both the frontline and relapsed/refractory (R/R) settings, include epcoritamab, glofitamab, mosunetuzumab, and odronextamab. Epcoritamab and glofitamab have received FDA and EMA approval for R/R DLBCL after two or more systemic line of therapies. EMA has also approved glofitamab in combination with gemcitabine and oxaliplatin (GemOx) for patients with R/R DLBCL ineligible for ASCT, whereas this indication has not been approved by FDA. Odronextamab is approved by EMA for R/R DLBCL and FL in patients who have received at least two prior lines of therapy, but it has not been approved by FDA. Mosunetuzumab is approved by both agencies—but only for R/R follicular lymphoma (FL). BsAbs represent a breakthrough therapy in the treatment of DLBCL, especially in R/R diseases. The purpose of this article is to review the landscape of BsAbs in DLBCL. Full article
(This article belongs to the Special Issue Monoclonal Antibodies in Lymphoma)
Show Figures

Figure 1

13 pages, 243 KB  
Review
The Evolving Role of Hematopoietic Stem Cell Transplantation in Philadelphia-like Acute Lymphoblastic Leukemia: From High-Risk Standard to Precision Strategies
by Matteo Molica, Claudia Simio, Laura De Fazio, Caterina Alati, Marco Rossi and Massimo Martino
Cancers 2025, 17(19), 3237; https://doi.org/10.3390/cancers17193237 - 5 Oct 2025
Viewed by 811
Abstract
Background: Philadelphia-like acute lymphoblastic leukemia (Ph-like ALL) is a high-risk subtype of B-cell ALL characterized by a gene expression profile similar to BCR::ABL1-positive leukemia, but lacking the BCR::ABL1 fusion gene. It is frequently associated with kinase-activating alterations, such as CRLF2 rearrangements, JAK-STAT pathway [...] Read more.
Background: Philadelphia-like acute lymphoblastic leukemia (Ph-like ALL) is a high-risk subtype of B-cell ALL characterized by a gene expression profile similar to BCR::ABL1-positive leukemia, but lacking the BCR::ABL1 fusion gene. It is frequently associated with kinase-activating alterations, such as CRLF2 rearrangements, JAK-STAT pathway mutations, and ABL-class fusions. Patients with Ph-like ALL typically experience poor outcomes with conventional chemotherapy, underscoring the need for intensified and targeted therapeutic approaches. Methods: This review summarizes current evidence regarding the role of hematopoietic stem cell transplantation (HSCT) in patients with Ph-like ALL. We analyzed retrospective cohort studies, registry data, and ongoing clinical trials, focusing on transplant indications, molecular risk stratification, measurable residual disease (MRD) status, timing of transplant, and post-transplant strategies. Results: Retrospective data suggest that HSCT in first complete remission (CR1) may improve survival in patients with high-risk molecular lesions or MRD positivity at the end of induction. However, the lack of prospective data specific to Ph-like ALL limits definitive conclusions. Post-transplant relapse remains a challenge, and novel strategies, including the use of tyrosine kinase inhibitors or JAK inhibitors as post-HSCT maintenance therapy, are being explored. Emerging immunotherapies, such as chimeric antigen receptor (CAR) T cells, may reshape the therapeutic landscape and potentially alter the indications for transplantation. Conclusions: HSCT remains a crucial therapeutic option for selected patients with Ph-like ALL, particularly those with poor molecular risk features or persistent MRD. However, further prospective studies are needed to evaluate the indication for HSCT in CR1 and the potential integration of transplantation with targeted and immunotherapeutic strategies. Personalized treatment approaches based on genomic profiling and MRD assessment are essential to improve outcomes in this high-risk subset. Full article
(This article belongs to the Special Issue Hematopoietic Stem Cell Transplant in Hematological Malignancies)
Back to TopTop