bims-carter Biomed News
on CAR-T Therapies
Issue of 2025–11–02
28 papers selected by
Luca Bolliger, lxBio



  1. Clin Transl Oncol. 2025 Oct 30.
      Chimeric antigen receptor (CAR) T cell therapy has revolutionized oncology but is fraught with a potentially lethal toxicity: cytokine release syndrome (CRS). While much focus is placed on pharmaceutical interventions, the pivotal role of nursing in the frontline detection, management, and rescue of patients from CRS remains under-championed in the literature. This narrative review synthesizes the current evidence to formally position nursing at the vanguard of CRS rescue. We delineate the intricate immunobiology of CRS as a primer for advanced practice and deconstruct its clinical spectrum to empower precise assessment. A novel four-pillar framework is presented, outlining nursing's critical role in pre-infusion preparedness, vigilant predictive monitoring, graded algorithm-driven intervention, and multidisciplinary coordination. Furthermore, we pioneer the discussion on defining nursing-sensitive outcomes (NSOs) specific to CRS to quantify nursing's impact on patient safety and efficacy. Finally, we explore future directions, including point-of-care testing and specialized nursing roles, advocating for a paradigm shift where nurses are recognized as essential leaders in optimizing safety and unlocking the full potential of CAR-T cell therapy.
    Keywords:  CAR-T cell therapy; Cytokine release syndrome; Nursing management; Nursing-sensitive outcomes; Toxicity rescue
    DOI:  https://doi.org/10.1007/s12094-025-04087-6
  2. J Autoimmun. 2025 Oct 24. pii: S0896-8411(25)00140-4. [Epub ahead of print]157 103495
      Chimeric antigen receptor-T (CAR-T) cells are genetically engineered T cells that has demonstrated significant success in treating hematologic tumors. The therapeutic landscape of CAR-T cell therapy is undergoing paradigm-shifting expansion into autoimmune disease management. Emerging clinical evidence demonstrates that engineered CAR-T cells can achieve unprecedented disease remission in the treatment of autoimmune diseases by efficiently depleting autoreactive B cells, thereby resetting the immune system, while maintaining favorable safety profiles. These encouraging results have motivated in-depth exploration into expanding the application of CAR-T cells to a broader spectrum of autoimmune diseases, as well as the development of advanced cell products with better efficacy and safety. This review systematically explores fundamental research and clinical applications of CAR-T cell therapy in autoimmune diseases, critically examines existing limitations, and proposes potential future directions. Significantly, the study elucidates structural characteristics of disease-associated autoantigens in autoimmune diseases, offering novel perspectives and technical frameworks for developing next-generation chimeric autoantibody receptor-T (CAAR-T) therapeutics with enhanced precision.
    Keywords:  Autoimmune diseases; Autoreactive B cells; CAAR-T; CAR-T cell
    DOI:  https://doi.org/10.1016/j.jaut.2025.103495
  3. J Transl Med. 2025 Oct 28. 23(1): 1182
      Chimeric antigen receptor (CAR) therapy represents an innovative form of targeted treatment that employs genetically engineered effector cells to selectively target tumor cells. CAR-T immunotherapy, which uses T cells as effector cells, has demonstrated remarkable efficacy in hematological malignancies. However, its clinical application in solid tumors remains limited due to challenges such as poor tumor infiltration, cytokine release syndrome (CRS), neurotoxicity, off-target effects, and other adverse events. To address these limitations, CAR-engineered natural killer (CAR-NK) cells and macrophages (CAR-M) have been developed. Macrophages, as crucial components of innate and adaptive immunity, exhibit superior tumor microenvironment infiltration and long-term persistence, making them a promising tool for next-generation cancer immunotherapy. This review highlights the construction strategies and preclinical and clinical studies of CAR-M, comprehensively introduces the anti-tumor mechanisms, discusses their advantages and disadvantages compared with other CAR-engineered effector cells, and explores the challenges and prospects for CAR-M in treating solid tumors.
    Keywords:  CAR-M; Immunotherapy; Solid tumors; Targeted therapy; Tumor immunity
    DOI:  https://doi.org/10.1186/s12967-025-07115-9
  4. Biomolecules. 2025 Oct 02. pii: 1407. [Epub ahead of print]15(10):
      Therapy with chimeric antigen receptor (CAR)-T cells has revolutionized the treatment of hematological malignancies. However, their application in solid tumors remains a formidable challenge due to obstacles such as the immunosuppressive tumor microenvironment, tumor heterogeneity, and limited T cell persistence. Although second- and third-generation CAR-T cells have shown restricted efficacy in clinical trials, next-generation strategies-including cytokine-armored CAR-T cells (e.g., IL-15, IL-7/CCL19), logic-gated systems, and localized delivery approaches-demonstrate promising potential to overcome these limitations. This review examines the major barriers impeding CAR-T cell efficacy in solid tumors, evaluates clinical outcomes from conventional CAR constructs, and highlights innovative strategies being tested in recent clinical trials. Key advances discussed include the use of dominant-negative receptors (e.g., TGFβRII) to combat immunosuppression and the co-expression of bispecific T cell engagers (BiTEs) to address antigen escape.
    Keywords:  CAR-T; T-cells; cells; clinical; efficacy next; generation; immunotherapy; solid; tumor
    DOI:  https://doi.org/10.3390/biom15101407
  5. Int J Mol Sci. 2025 Oct 16. pii: 10070. [Epub ahead of print]26(20):
      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.
    Keywords:  autoimmune kidney disease; chimeric antigen receptor; immunotherapy
    DOI:  https://doi.org/10.3390/ijms262010070
  6. Biomol Ther (Seoul). 2025 Nov 01. 33(6): 907-923
      Chimeric Antigen Receptor (CAR)-based cell and gene therapies have become transformative treatments, offering targeted and durable responses, especially in hematologic malignancies. This review analyzes 1,744 CAR clinical trials registered on Clinical-Trials.gov as of 2024, focusing on platform types, indications, target antigens, therapeutic strategies, and late-phase development. CAR-T therapies predominate, followed by CAR-NK, CAR-NKT, CAR-M and CAR-DC platforms. Approximately 92% of trials target tumors, with hematologic malignancies accounting for 65% of indications; CD19 and BCMA are primary targets in Phase 3 studies. Solid tumor applications are expanding steadily, driven by unmet clinical needs and advances in CAR engineering. Although monospecific CARs dominate, dual, bispecific, and universal designs are gaining traction to overcome antigen heterogeneity and tumor escape. Combination therapies, such as CAR-T with chemotherapy or monoclonal antibodies, are increasingly used to improve efficacy. CAR-NK therapies, while in early development, show promise due to favorable safety profiles and off-the-shelf allogeneic potential. The United States and China lead global development, supported by robust research ecosystems and industrial investment. Overall, CAR-based therapeutics are evolving from hematologic specialization toward broader clinical application, addressing challenges and guiding future strategies.
    Keywords:  CAR-NK; CAR-T; Cell and gene therapy; Chimeric antigen receptor
    DOI:  https://doi.org/10.4062/biomolther.2025.153
  7. J Hematol Oncol. 2025 Oct 27. 18(1): 91
      Chimeric antigen receptor T-cell (CAR-T) therapy has revolutionized the treatment of hematologic malignancies, offering a highly personalized and potent immunotherapeutic approach. To date, the U.S. Food and Drug Administration has approved seven CAR-T therapies targeting CD19 and B-cell maturation antigen; each demonstrated remarkable clinical efficacy across various hematologic malignancies. Despite significant advancements in preclinical studies and clinical trials, no CAR-T therapy has been approved for solid tumors, which account for the majority of cancer cases worldwide. These key challenges include the lack of distinct and accessible target antigens, the immunosuppressive tumor microenvironment (TME) that impairs immune cell efficacy, the heterogeneity of solid tumors that complicates treatment uniformity, and the potential risks of off-tumor toxicity. These obstacles represent a complex array of biological and clinical obstacles, distinct from the more favorable immune environment of hematologic cancers that has been pivotal to the success of CAR-T therapy. Preclinical studies in multiple myeloma emphasize memory T-cell optimization and combinatorial strategies to enhance CAR-T efficacy in solid tumors. Our review emphasizes innovative strategies to address these key challenges in CAR-T therapy for solid tumors, including advanced multi-antigen targeting approaches, reprogramming of the TME, and the development of next-generation safety measures to mitigate toxicity risks. By addressing both scientific and clinical obstacles, this review envisions a future in which CAR-T therapy's full potential extends beyond hematologic malignancies, transforming the landscape of oncology and improving outcomes for patients with solid tumors.
    DOI:  https://doi.org/10.1186/s13045-025-01744-9
  8. Cureus. 2025 Sep;17(9): e93017
      Case reports and case studies increasingly demonstrate that chimeric antigen receptor (CAR) T cell therapy (CTCT) is beneficial not only in hematologic malignancies but also in immunologic diseases, including neuromuscular disorders. The aim of this review is to provide an overview of the current status of CTCT in immune-mediated neuromuscular disorders. This is a systematic review of relevant literature recruited using PubMed, Embase, Scopus, and Google Scholar search terms. Neuromuscular disorders for which CTCT has been used to date include myasthenia gravis (n = 4), Lambert-Eaton syndrome (n = 1), myasthenia/Lambert-Eaton overlap (n = 2), dermatomyositis (n = 2), immune-mediated necrotizing myositis (n = 2), idiopathic inflammatory myopathy (n = 1), anti-synthetase syndrome (n = 4), and chronic inflammatory demyelinating polyneuropathy (n = 2). In most cases, CTCT was directed against CD19-positive lymphocytes and in some cases against B cell maturation antigen. In all reported patients, there was a significant improvement in motor function and quality of life, with some even making a full recovery several months after the application of CTCT. In conclusion, CTCT appears to be a promising therapeutic option for patients with severe immune neuromuscular disorders in whom previous treatment with multiple immunomodulatory therapies has been ineffective. CTCT should be considered in patients with immune neuromyopathy who do not respond to immunomodulatory therapies.
    Keywords:  car t cell therapy; immunological; myasthenia; neuromuscular disorders; t-cells
    DOI:  https://doi.org/10.7759/cureus.93017
  9. Zhonghua Xue Ye Xue Za Zhi. 2025 Sep 14. 46(9): 875-880
      Autoimmune diseases (AID) are disorders in which the immune system mistakenly attacks the body's own tissues. However, current immunosuppressive therapies seldom achieve durable, drug-free remission, indicating the urgent need for therapeutic strategies that are both more precise and longer-lasting. Chimeric antigen receptor (CAR) -T cells are generated by genetically engineering T cells to specifically recognize and kill cells that express particular antigens, thereby providing a novel therapeutic approach for AID. This review summarizes the immunological mechanisms of CAR-T cells and -regulatory T cells (Treg) in treating AID, and systematically reviews the latest advances in applying these treatments to rheumatic AID, immune-mediated neurological AID, and refractory autoimmune hemolytic anemia, among others. Further, we discuss the safety-related limitations of CAR-T /CAR-Treg treatment for AID, and outline other CAR-based cellular therapies that can be used to treat AID beyond CAR-T and CAR-Treg cells.
    DOI:  https://doi.org/10.3760/cma.j.cn121090-20250524-00242
  10. Biomedicines. 2025 Oct 18. pii: 2545. [Epub ahead of print]13(10):
      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.
    Keywords:  CAR-T cells; chimeric antigen receptor; immunotherapy; prostate cancer; targeted prostate antigens
    DOI:  https://doi.org/10.3390/biomedicines13102545
  11. Int J Hematol. 2025 Oct 27.
      A 53-year-old woman experienced relapse of diffuse large B cell lymphoma (DLBCL) 16 years after achieving a first complete metabolic response (CMR). Despite initially being refractory to salvage chemotherapy, she achieved a second CMR and underwent umbilical cord blood transplantation (UCBT). On day 119 post-transplantation, she experienced a second relapse and received chimeric antigen receptor T (CAR T) cell therapy, achieving a third remission without cytokine release syndrome or graft-versus-host disease (GVHD). However, a third relapse occurred seven months after CAR T cell therapy. Epcoritamab treatment was initiated, resulting in CMR without severe complications. Subsequently, the patient underwent curative unrelated allogeneic peripheral blood hematopoietic stem cell transplantation (HSCT). She experienced no severe transplant-related complications, including serious GVHD or infections. She has remained in CMR for > 1 year after the second transplant. Epcoritamab appears to be an effective and safe treatment option for DLBCL relapse, even after both UCBT and CAR T cell therapy, suggesting that bridging therapy with epcoritamab followed by a second allogeneic HSCT may achieve long-term survival.
    Keywords:  Allogeneic hematopoietic stem cell transplantation; Bridging therapy; Chimeric antigen receptor T cell therapy failure; Epcoritamab; Relapsed or refractory diffuse large B cell lymphoma
    DOI:  https://doi.org/10.1007/s12185-025-04091-0
  12. Nat Rev Chem. 2025 Oct 29.
      Chimeric antigen receptor (CAR) T cell therapy is a 'living drug' in which the T cells of patients are genetically engineered with an artificial receptor that directs them to attack diseased cells. CAR T cell therapies have had remarkable impact, curing subsets of patients with previously untreatable, late-stage cancers. However, limitations persist, including severe toxicities, limited survival of engineered cells, and therapeutic resistance. Genetically encoded small-molecule control systems have been developed to address these limitations. They can halt toxicities by eliminating CAR T cells or switching off their function. Furthermore, they can enhance therapy by directly targeting antigens or broadening cell killing ability through cytotoxic pro-drug activation. Small-molecule controllers include protease inhibitors, protein dimerizers, protein degraders, bi-specific adaptors and conditionally activated chemotherapeutics. Here, we outline small-molecule-based control approaches, categorizing them by function and detailing their molecular mechanisms. We emphasize systems in the clinic and highlight emerging applications and unmet areas.
    DOI:  https://doi.org/10.1038/s41570-025-00768-6
  13. J Hematol Oncol. 2025 Oct 27. 18(1): 92
      In recent years, the development of chimeric antigen receptor (CAR) technology has greatly promoted the progress of cellular immunotherapy. Among them, CAR-T cell therapy has shown remarkable clinical effects in the treatment of hematological malignancies. However, this therapy still faces a series of challenges, including immunogenicity, toxic side effects, and insufficient maintenance of long-term efficacy. The latest research progress has extended CAR technology to mesenchymal stem cells (MSCs), and the resulting CAR-MSCs combine the precise targeting ability of CAR molecules with the inherent immunomodulatory, tissue homing, and regenerative repair properties of MSCs, providing a new therapeutic strategy for cancer and immune-related diseases. This review examines the engineering design, biological characteristics, and applications of CAR-MSCs in oncology and immune-related disorder therapy. Preclinical studies have shown their effectiveness against glioblastoma, Ewing sarcoma, acute myeloid leukemia, and lung cancer, as well as graft-versus-host disease, through TRAIL secretion, bispecific antibody production, and Treg induction. Despite promising results, significant hurdles persist in CAR-MSC manufacturing scalability, cell persistence, heterogeneous MSC tissue sourcing, and undefined application protocols, all of which are critical for clinical translation. We investigated corresponding strategies, including nonviral gene delivery, metabolic engineering, senescence-resistant MSC clones, and microenvironment-specific activation. Standardized production workflows incorporating rigorous quality control are essential for future applications. CAR-MSCs represent a paradigm shift in precision immunotherapy by providing dual therapeutic modalities for cancer and immune disorders. Fully unlocking their therapeutic potential will require interdisciplinary efforts to overcome biological and technical barriers while advancing combination therapies.
    Keywords:  CAR-MSCs; Cellular immunotherapy; Chimeric antigen receptor; Clinical translation; Genetic engineering; Immune diseases; Immunomodulation; Mesenchymal stem cells; Tumor microenvironment; Tumor targeting
    DOI:  https://doi.org/10.1186/s13045-025-01746-7
  14. ACS Cent Sci. 2025 Oct 22. 11(10): 1852-1861
      Chimeric antigen receptor (CAR) T cell therapy has revolutionized the treatment of hematologic malignancies; however, durable remissions remain limited due to antigen-negative cancer relapse, where tumor cells downregulate or lose the targeted antigen to evade immune recognition. To address this challenge, we developed cysteine-engineered CAR (CysCAR) T cells that redirect T cells to target cancer cells based on extracellular redox imbalances and the altered thiol/disulfide ratios, a marker we identified on B cell lymphomas. Here, we show that CysCAR-T cells, engineered with different cysteine-modified antibody fragments, exhibit a potent and specific cytotoxicity in vitro across various B cell lymphoma (BCL) subtypes, even in antigen escape models. Moreover, by integrating cysteine engineering with clinically used anti-CD19 CAR-T cells, we enabled simultaneous targeting of CD19 and altered redox states on BCL, potentially reducing the risk of antigen escape. In a pilot in vivo study, these bifunctional CD19-CysCAR-T cells suppressed tumor growth and prolonged survival of BCL-bearing mice without inducing systemic toxicity. Given that aberrant exofacial redox states are a hallmark of multiple cancers, our findings suggest a promising strategy to enhance the efficacy of anti-CD19 CAR-T cell therapy, overcome antigen escape, and reduce tumor relapse in BCL, with potential applicability to other malignancies.
    DOI:  https://doi.org/10.1021/acscentsci.5c00816
  15. Nat Rev Cancer. 2025 Oct 27.
      Current T cell-based immunotherapy strategies, including immune checkpoint blockade (ICB) and chimeric antigen receptor (CAR) T cells, have revolutionized cancer care. However, many patients with cancer who are treated with these approaches fail to respond or do not achieve durable protection against disease relapse, highlighting the need for further optimization of such strategies. The advent of cancer immunotherapy has ushered in an era of research centred on immune oncology with a specific focus on defining T cell-intrinsic mechanisms that delineate therapeutic responders and non-responders. Among the major barriers limiting immunotherapy efficacy, T cell exhaustion - which is characterized by repression of the effector functions and proliferative potential of T cells - has emerged as a common mechanism among various cancers. Here, we review transcriptional and epigenetic mechanisms that control T cell exhaustion. We discuss how T cell subset-specific gene regulatory programmes limit immunotherapy success and theorize on the development of next-generation strategies for increasing the clinical breadth, efficacy and durability of T cell immunotherapy.
    DOI:  https://doi.org/10.1038/s41568-025-00883-y
  16. J Neurooncol. 2025 Oct 31. 176(1): 60
      
    Keywords:  Chimeric antigen receptor T cell therapy (CAR T); Clinical trial; Diffuse midline glioma; Immunotherapy; Neurotoxicity; Pediatric
    DOI:  https://doi.org/10.1007/s11060-025-05251-2
  17. Clin Pharmacol Ther. 2025 Oct 27.
      This study undertook a comprehensive analysis to assess China's progress in the approval of cell and gene therapy products (CGTPs). It provided insights into clinical trials and regulatory advancements by examining the approval characteristics, pivotal clinical trials, expedited programs, orphan designations, development and review timelines, and drug lags through a comparative analysis across four regions: the United States, the European Union, Japan, and China. The findings indicated that China had achieved substantial advancements in the approval of CGTPs in recent years. This progress was evidenced by a year-over-year increase in the number of approvals, a flexible authorization approach utilizing surrogate endpoints and single-arm trials, support for product launches through various expedited programs, and the shortest development timelines among the four regions analyzed. Nevertheless, when compared to the United States, European Union, and Japan, China faced several challenges in the approvals for CGTPs. Notably, the therapeutic area is predominantly in oncology, with the market largely concentrated domestically. Two aspects to be highlighted were that the orphan designation was lacking as well as the implementation of multi-regional clinical trials (MRCTs) in pivotal clinical trials. Furthermore, the progression of post-marketing confirmatory trials was sluggish and lacked robust oversight mechanisms. The expedited programs supporting the CGTP approvals required optimization concerning categories and applications. Additionally, the review timelines had not demonstrated a competitive advantage, and the drug lags were the longest across the four regions. It is imperative for regulatory authorities to take proactive measures to mitigate any deficiencies resulting from the identified challenges.
    DOI:  https://doi.org/10.1002/cpt.70099
  18. Int Rev Cell Mol Biol. 2025 ;pii: S1937-6448(25)00118-2. [Epub ahead of print]398 1-51
      While the function of αβ T cells in host defense is quite well understood, insight into the role that γδ T cells play in health and disease has lagged behind, despite their discovery approximately forty years ago. Because γδ T cells represent a relatively small population in immune organs, their importance in host defense was previously questioned, which dampened research interest in γδ T cells, even among immunologists. Fortunately, there has been a relatively recent surge in interest in γδ T cells, which has revealed that they do in fact play a central role in human health and disease. They perform diverse functions that impact tissue homeostasis and integrity in addition to their role in pathogen resistance, which positions them at the interface between the innate and adaptive arms of the immune system. Here, we review the progress in understanding how γδ T cells can both promote health and cause disease, and how they are being exploited as a new and perhaps more effective weapon in the treatment of cancer.
    Keywords:  Autoimmunity; Cancer; Development; Function; Therapy; γδ T cells
    DOI:  https://doi.org/10.1016/bs.ircmb.2025.08.012
  19. Naunyn Schmiedebergs Arch Pharmacol. 2025 Oct 27.
      Exosomes have garnered significant interest in biomedical research due to their potential therapeutic applications. They are extracellular vesicles secreted by cells, distinguished by a lipid bilayer membrane that encases various biological substances, including nucleic acids and proteins, within their lumen or lipid bilayer. They offer several advantages, like superior compatibility and targeted delivery capability, enabling innovative therapeutic development and efficient drug transport across cellular barriers, including the BBB. They also provide long circulation times, low toxicity, and protection from degradation. They play critical roles in cell communication, tissue repair, facilitating immune response, modulating inflammation, homeostasis, transferring molecules between cells, and specifically homing to tumor sites. Techniques such as microfluidic-based isolation and surface modification are advancing the production of clinical-grade exosomes. Exosomes have shown potential in delivering drugs for a wide range of diseases, including cancer, neurological disorders, infectious diseases, and cardiovascular issues. Challenges such as low yield during isolation, difficulty in large-scale production, heterogeneity of exosome populations, and maintaining stability during storage hinder their practical use. Multidisciplinary research is needed to overcome these limitations and unlock their potential in early disease detection and therapy, with future applications expected in advanced drug delivery, diagnostic biomarkers, and disease prognosis. Regulatory considerations, including rigorous preclinical and clinical trials, are crucial for translating these innovations into approved therapies. This review highlights the emerging development of exosomes in therapeutic drug delivery systems. The loading technique is discussed in a detailed manner through which the drugs are attached to exosomes and delivered to the target site.
    Keywords:  Biogenesis; Drug loading; Engineered exosomes; Exosomes; Extracellular vesicles; Isolation
    DOI:  https://doi.org/10.1007/s00210-025-04615-9
  20. Transplant Cell Ther. 2025 Oct 23. pii: S2666-6367(25)01528-3. [Epub ahead of print]
       BACKGROUND: CD19-directed chimeric antigen receptor (CAR) T-cell therapies, including lisocabtagene maraleucel, axicabtagene ciloleucel, and tisagenlecleucel, have revolutionized the treatment landscape for patients with hematologic malignancies. However, identification and referral of patients who could benefit from treatment remains a significant challenge.
    OBJECTIVE: To report expert recommendations for CAR T-cell therapy referral.
    METHODS: Recommendations were gathered from 10 experts in oncology, hematology, cardiology, and infectious disease from a roundtable meeting and/or subsequent reviews from November 13, 2024, to June 9, 2025.
    RESULTS: The authors considered the following potential factors: age, performance status, disease status, cardiovascular function, pulmonary function, renal function, hepatic function, infections, and psychological health. Based on existing evidence, the authors agreed that none of the factors discussed should preclude patients from receiving referrals/further evaluation for CAR T-cell therapy, particularly with current advances in supportive care and integration of services from other specialties. Timely referral should be made by the patient's primary oncologist to specialists as early as the disease is deemed relapsed or refractory, and preferably before the subsequent line of therapy is started to allow better access to care and to improve treatment outcomes. Before CAR T-cell therapy, holding therapy (before leukapheresis) and/or bridging therapy (after leukapheresis) may be given to patients with high-volume disease, in consultation with CAR T-cell therapy specialists. Based on the safety profile of CAR T-cell therapies, experts recommended flexible monitoring and transfer of care back to primary/community oncology physicians, starting from 2 weeks after infusion to improve access to this potentially curative therapy. Adaptations to clinical practice based on the most recent regulations, policy requirements, and institutional guidelines should be made as needed.
    CONCLUSIONS: In summary, a panel of 10 experts provided recommendations for timely patient referral for CAR T-cell therapy upon occurrence of relapsed or refractory disease and before initiation of subsequent line of therapy to improve care access and treatment outcomes. Experts noted that with close collaboration between CAR T-cell therapy specialists and other medical disciplines, CAR T-cell therapy remains a feasible option for most patients despite their comorbidities.
    Keywords:  Axicabtagene ciloleucel; Brexucabtagene autoleucel; Cellular therapy; Chimeric antigen receptor T-cell therapy; Expert perspectives; Lisocabtagene maraleucel; Lymphoma; Patient identification; Patient selection; Referral; Tisagenlecleucel
    DOI:  https://doi.org/10.1016/j.jtct.2025.10.025
  21. Sci Rep. 2025 Oct 28. 15(1): 37638
      T cells are critical components of the human immune system. When a cell is infected by a virus, it presents viral peptides on its surface using human leukocyte antigen (HLA) proteins. These peptide-HLA complexes are recognized by T cells through interactions with T cell receptors (TCRs). A human blood sample can contain millions of unique TCRs, which is a sample from the individual's TCR repertoire. TCR repertoire-wide association studies (TReWAS) aim to evaluate the associations between individual TCRs and disease or exposure status. Previous studies have shown that TCRs associated with viral infections can be identified using TReWAS, and these TCRs can be used to predict current or past infection with high accuracy. Many TCRs are strongly associated with specific HLA alleles, suggesting that the incorporation of HLA information could improve the precision of TReWAS analyses and predictions based on TCRs. In this study, we evaluated TCR-based predictions while conditioning on individual HLA alleles or their k-nearest neighbors. We observed improved prediction accuracy for some HLA alleles. Furthermore, these HLA-specific predictions provide insight into the role of specific HLAs in coordinating immune response to immunogenic antigens, demonstrating the benefit of HLA-aware analysis of TCR data.
    DOI:  https://doi.org/10.1038/s41598-025-21453-0
  22. Int Rev Cell Mol Biol. 2025 ;pii: S1937-6448(25)00115-7. [Epub ahead of print]398 151-184
      gamma-delta (γδ) T cells reside in numerous tissues, recognize a vast array of antigens, and perform highly specialized functions. They have participated in antimicrobial and antitumor immunity, in modulation the immune responses and in tissue repair . γδ T cells are enriched in mucosa and skin; indeed, they are part of intraepithelial T lymphocytes, where they function as sentinels, promptly detecting infections, cellular stress, or tissue damage. It is well known that Escherichia coli (E. coli) is a common bacterium that can cause infections in humans. While many strains of E. coli are harmless and even beneficial to the host, some can cause gastroint estinal diseases or urinary tract infections, among other pathologies associated with the bacteria. The relationship between γδ T cells and E. coli remains relatively underexplored. However, gaining insight into this interaction is essential for understanding how γδ T cells recognize and respond to bacterial pathogens, a critical step in developing strategies for preventing and treating bacterial infections. This chapter explores the current knowledge about the role of human γδ T cells in infections caused by E. coli.
    Keywords:  Escherichia coli; Infection; γδ T cells
    DOI:  https://doi.org/10.1016/bs.ircmb.2025.08.009
  23. Glob Health Res Policy. 2025 Oct 27. 10(1): 52
       BACKGROUND: Access to novel medications matters quality-adjusted life years and the opportunity cost associated with productivity lost. Gaps in patient access to novel medicines exist due to insufficient public funding reimbursement in emerging countries. Evidence of time from regulatory approval to reimbursement decision by public funding, referred to as time to reimbursement (TTR), remained limited in emerging countries. This study compared and analyzed public funding reimbursement of novel medicines approved in five Eurasian countries that are global leaders in pharmaceutical innovation. All of them have a centralized mechanism for reimbursement decisions on novel medicines, allowing identification of a clear date of public funding reimbursement. By exploring the facilitators of rapid application of pharmaceutical innovations, we expected to inform the public funding reimbursement decision-making in emerging countries, so as to improve patient access and contribute to addressing the global health challenge in achieving universal health coverage.
    METHODS: This is a retrospective study which investigated the public funding reimbursement and TTR of novel medicines that obtained marketing authorization between 2018 and 2023 in China, Japan, France, the United Kingdom (UK) and Switzerland. We firstly conducted descriptive analyses of TTR across countries, followed by the pairwise comparisons using Kruskal-Wallis H tests with Bonferroni corrections. We then performed the survival analysis of time-to-event data using the multiple Cox proportional hazards regression by inclusion of country and year dummy variables. Other covariates associated with the characteristics of novel medicines and manufacturers, as well as the review and approval pathways were included in the regression. We estimated the differences of hazard ratios (HR) of novel medicines being reimbursed by public funding across countries. Subgroup analyses were conducted to assess the specific factors associated with the public funding reimbursement in different countries. Since China began to systematically publicly fund novel medicines in 2019, sensitivity analyses were conducted by removing the 2018 data and repeating the same analyses.
    RESULTS: As of July 1st, 2024, Japan had the highest proportion and fastest rate of public funding reimbursement of novel medicines, which were approved between 2018 and 2023 (HR = 11.29, [95% CI 8.63, 14.77], P < 0.001). In contrast, the TTR of novel medicines approved in China was generally longer than those in the other four countries. Factors associated with a higher likelihood of being reimbursed by public funding included priority review procedure in China and the UK, medicines for rare diseases approved in Japan and France, anti-cancer medicines approved in the UK, locally developed novel medicines approved in China and Switzerland, and medicines launched by large multinational pharmaceutical companies in France and Switzerland. China was the only country in which novel medicines approved through conditional market authorization were less likely to be publicly funded (HR = 0.42, 95% CI [0.27, 0.68], P < 0.001).
    CONCLUSIONS: Compared to other global pharmaceutical innovation leaders, China still needs to make further efforts in strengthening public funding reimbursement of novel medicines. A forward-thinking strategy for health technology assessment that provides advanced technical support in conjunction with the regulatory authority to pharmaceutical innovation companies at the early research and development stage is critical for reducing TTR of novel medicines and accelerating patient access. To balance timely patient access and risk control, strategies such as risk-sharing mechanisms for novel medicines with clinical and cost uncertainties, and temporary reimbursement with alternative sources of funding to support real-world evidence collection could be considered.
    Keywords:  Cox proportional hazards regression; Novel medicines; Public funding; Survival analysis; Time to reimbursement
    DOI:  https://doi.org/10.1186/s41256-025-00457-3
  24. Curr Opin Immunol. 2025 Oct 30. pii: S0952-7915(25)00161-X. [Epub ahead of print]98 102685
      Regulatory T cells (Tregs) are a rare subset of T cells that are potent regulators of the immune system. As a result, the use of Tregs to prevent undesired immune activation and to re-establish immune balance is an attractive cellular therapy approach. In hematopoietic cell transplantation (HCT), there has been significant interest in using Tregs to prevent graft-versus-host disease (GVHD), which occurs when donor alloreactive T cells recognize recipient HLA- or other minor transplantation antigens as non-self. There is a critical unmet need for novel strategies to prevent GVHD since as many as 50% of patients will develop clinically significant GVHD with current therapies. Evidence from preclinical murine models and early-phase human studies suggests that an additional benefit to Treg cellular therapy in HCT is the apparent maintenance of pathogenic immunity and graft-versus-tumor effects, which are required to prevent leukemia relapse. While the major limiting factor for Treg-based therapies has been the rarity of these cells, novel methods to improve Treg isolation and expansion have demonstrated feasibility for implementation of these strategies in the clinic. Moreover, genetic engineering of Tregs has been shown to be a promising strategy to improve their specificity, longevity, and function. Clinical trials have established that such approaches are feasible and can be effective in both human leukocyte antigen (HLA)-matched and haplo-matched settings. In this review, we describe proposed mechanisms for Treg control of alloreactivity, modern methods for Treg isolation and expansion, the history of Treg clinical trials for GVHD prevention, and the horizon for Treg cellular therapy.
    DOI:  https://doi.org/10.1016/j.coi.2025.102685
  25. Biomedicines. 2025 Oct 14. pii: 2497. [Epub ahead of print]13(10):
      Dendritic cells (DCs) are the most highlighted cell population for cancer immunotherapy development. Currently, DC-derived exosomes show promising anti-cancer activity. Exosomes are a subpopulation of extracellular vesicles (EVs) and originate from endosomes. It transports dynamic molecular cargos such as DNA, RNA, protein, and lipid. This cellular cargo exchange reprograms the recipient cell naturally. In cancer research, DC-derived exosomes (DEXs) are used as a therapeutic tool. There are some approaches followed in the application of DEX in cancer as a therapeutic tool. DEX-based drug delivery, tumor antigen-loaded DEX, and modified DEX are applicable approaches in cancer therapy. DEXs are biocompatible, nontoxic, and have ability-specific targeting. On the other hand, this method faces some challenges, such as large-scale production, isolation, and heterogeneity. A multidisciplinary approach (advanced nanotechnology, multi-omics, and single-exosome profiling) comes up with a solution to this issue. This review provides a comprehensive overview of the DEX approach, tracing its developmental journey and therapeutic application in cancer immunotherapy. It examines key findings from clinical trials and outlines the challenges and future research directions in this field, ultimately underscoring the potential of DC-derived exosomes as a research-backed, cell-free solution for the next generation of cancer immunotherapies.
    Keywords:  DC exosomes; cancer; cancer immunotherapy; metastasis
    DOI:  https://doi.org/10.3390/biomedicines13102497
  26. Brief Bioinform. 2025 Aug 31. pii: bbaf554. [Epub ahead of print]26(5):
      Accurately determining the binding affinity of a ligand with a protein is important for drug design, development, and screening. With the advent of accessible protein structure prediction methods such as AlphaFold, predicted protein 3D structures are readily available; however, scalable methods for predicting binding affinity currently do not take full advantage of 3D protein information. Here, we present CASTER-DTA (Cross-Attention with Structural Target Equivariant Representations for Drug-Target Affinity), which uses an equivariant graph neural network (GNN) to learn more robust protein representations alongside a standard GNN to learn molecular representations to predict DTA. We augment these representations by incorporating an attention-based mechanism between protein residues and drug atoms to improve interpretability. We show that CASTER-DTA represents a state-of-the-art improvement on multiple benchmarks for predicting DTA, and that it generates novel insights for several related tasks. We then apply CASTER-DTA to create a large resource of the binding affinities of every drug approved by the U.S. Food and Drug Administration (FDA) against every protein in the human proteome and make these predictions freely available for download. We also make available a web server for researchers to apply a pretrained CASTER-DTA model for predicting binding affinities between arbitrary proteins and drugs.
    Keywords:  deep learning; graph neural networks; protein representation learning; structural biology
    DOI:  https://doi.org/10.1093/bib/bbaf554
  27. J Chemother. 2025 Nov 01. 1-22
      Polymer-based nanoparticles (PNPs) are emerging as a cornerstone in cancer therapy due to their ability to enhance drug solubility, improve pharmacokinetics and achieve precise tumor targeting. Nanoparticles, encompassing liposomes, dendrimers, and metallic particles, include typical characteristics such as improved surface area, regulated release and the capacity to encapsulate diverse therapeutic compounds augmenting the pharmacokinetics and bioavailability of anticancer drugs. Recent studies demonstrate drug loading efficiencies of 80-90%, circulation half-life extensions of 2-5 fold, and tumor accumulation improvements of 3-10 times compared to free drugs. FDA-approved formulations such as Abraxane® (albumin-bound paclitaxel) and clinical candidates like Genexol-PM® (polymeric micelles) highlight the translational relevance of PNPs. This review consolidates advancements in polymeric nanocarriers, including nanospheres, nano-capsules and hybrid composites, while addressing limitations in regulatory approval and personalized oncology integration. This study shows that nanoparticle-based cancer therapeutics hold immense potential to improve treatment efficacy and patient outcomes in clinical oncology.
    Keywords:  Nanoparticles; biocompatibility; oncology; targeted drug delivery; therapeutic agents
    DOI:  https://doi.org/10.1080/1120009X.2025.2573516
  28. Biomedicines. 2025 Oct 13. pii: 2494. [Epub ahead of print]13(10):
      Background/Objectives: T cell dysfunction represents a fundamental barrier to effective cancer immunotherapy. Although immune checkpoint blockades and adoptive cell transfer have achieved clinical success, therapeutic resistance remains prevalent across cancer types. Thymopentin (TP5), a synthetic immunomodulatory pentapeptide (Arg-Lys-Asp-Val-Tyr), has demonstrated immunostimulatory properties, yet its anticancer potential remains unexplored. The aim of this study was to investigate TP5's antitumor efficacy and underlying immunological mechanisms. Methods: We evaluated TP5's therapeutic effects in multiple murine tumor models, including B16-F10 melanoma, MC38 colorectal carcinoma, Hepa 1-6, and LM3 hepatocellular carcinoma. Immune cell populations and functional states were characterized using flow cytometry, ELISAs, and immunofluorescence analyses. The potential of TP5 as an adjuvant for T cell-based therapies was also systematically assessed. Results: The TP5 treatment markedly suppressed tumor growth across caner models through strictly T cell-dependent mechanisms. Critically, TP5 promoted thymic rejuvenation under immunocompromised conditions, restoring the thymus-tumor immunological balance and revitalizing peripheral T cell immunity. TP5 functionally reprogrammed T cell states, preserving effector function while ameliorating exhaustion. Furthermore, TP5 demonstrated synergistic efficacy when combined with adoptive T cell therapies, enhancing both proliferation and effector functions. Conclusions: TP5 represents a promising immunomodulator that addresses fundamental limitations of current T cell therapies by simultaneously enhancing T cell function and reversing thymic involution under immunocompromised conditions. Our findings provide compelling evidence for TP5's clinical translation in cancer treatment.
    Keywords:  T cell exhaustion; cancer; cancer immunotherapy; thymopentin
    DOI:  https://doi.org/10.3390/biomedicines13102494