bims-carter Biomed News
on CAR-T Therapies
Issue of 2025–11–23
34 papers selected by
Luca Bolliger, lxBio



  1. Clin Rev Allergy Immunol. 2025 Nov 19. 68(1): 100
      Chimeric Antigen Receptor (CAR) T cell therapy has revolutionised the treatment of relapsed/refractory B cell leukaemia, lymphoma and multiple myeloma through targeting of CD19 and BCMA antigens on the surface of these cells. A growing body of evidence has recently demonstrated that these cell-based therapies can also target autoimmune diseases including systemic lupus erythematosus, systemic sclerosis, neuromyelitis optica spectrum disorder, myasthenia gravis, idiopathic inflammatory myositis, multiple sclerosis and rheumatoid arthritis. To date, ten patients with rheumatoid arthritis have been treated with CAR T cells targeting CD19/CD20 or BCMA antigens on B cells. Nine patients with seropositive disease have shown remarkable responses, including depletion of circulating B cells, ablation of autoantibody levels and drug-free remission. A tenth patient with seronegative disease initially responded to CAR T cell therapy but later relapsed. This review provides in-depth analysis of these single case studies and highlights emerging in-vitro and animal model studies where T cell subsets have been engineered with CARs to fine-tune their immune responses for the treatment of rheumatoid arthritis, including targeting of autoreactive B cells, autoreactive T cells or fibroblasts. CAR T cell therapy holds enormous promise for the treatment of difficult-to-treat rheumatoid arthritis, but more research and large clinical trials are needed to confirm its efficacy and safety.
    Keywords:  BCMA; CAR T cells; CD19; Inflammation; Rheumatoid arthritis
    DOI:  https://doi.org/10.1007/s12016-025-09113-7
  2. Front Immunol. 2025 ;16 1675114
      Regulatory T cell (Treg) therapies are emerging as powerful tools for treating autoimmune and inflammatory diseases, preventing graft-versus-host disease (GvHD), and promoting organ transplant tolerance. Building on the identification of chimeric antigen receptor (CAR)-expressing Tregs as a correlate of poor patient outcomes in CD19-CAR T cell therapy, this review examines strategies for learning from clinical samples and data to improve Treg therapies. We highlight current and next-generation Treg modalities, including polyclonal, antigen-specific, converted, TCR-engineered, and CAR-engineered Tregs, provide a comprehensive overview of Treg clinical trials, and evaluate the evolving toolkit for in vivo Treg monitoring. Emphasis is placed on advanced immunomonitoring technologies, such as single-cell multi-omic profiling, epigenetic analysis, and spatial transcriptomics, which enable precise characterization of Treg persistence, function, and lineage stability. By integrating insights from adoptive T cell therapies and cutting-edge multi-omic platforms, this review outlines how Treg therapies can be optimized as "living drugs" capable of establishing immune tolerance across diverse clinical contexts.
    Keywords:  GvHD; T cell therapy; Treg; autoimmunity; immune tolerance; immunomonitoring; regulatory T cell; transplantation
    DOI:  https://doi.org/10.3389/fimmu.2025.1675114
  3. Neurol Neuroimmunol Neuroinflamm. 2026 Jan;13(1): e200511
      Chimeric antigen receptor (CAR) T cells are genetically modified T cells expressing CARs, initially developed to recognize tumor antigens and kill cancer cells that evade T-cell recognition. Because of their impressive success in hemato-oncologic malignancies, CAR T cells are being repurposed with redesigned constructs for safety and sustained efficacy to target refractory systemic autoimmune or neurologic diseases. The CD19 CAR T cells-targeting those CD19-positive, antibody-secreting, long-lived plasma cells, and plasmablasts-are now extensively explored in refractory neuroautoimmunities with promising benefits based on case series in patients with myasthenia gravis (MG), stiff person syndrome (SPS), neuromyelitis, myositis, and multiple sclerosis; some patients with MG and SPS with steadily progressive and disabling disease refractory to all available therapies, including rituximab and new biologics, exhibit impressive clinical improvements with long-lasting benefits. The review, triggered by these early results and ongoing trials, addresses what these cells are and why they show effectiveness not only in antibody-mediated B-cell neurologic diseases unresponsive to available anti-B-cell agents but also in patients with nonpathogenic antibodies, implying effects even beyond B cells; points out that CARs are "living cells" penetrating physiologic barriers, such as the blood-brain barrier, expanding within tissues to memory cells ensuring sustained effects; describes the process and challenges of preparing and administering CAR T cells and their safety profile stressing the differences in toxicities when treating autoimmunites vs malignancies; and highlights that CD19 CAR T cells can successfully target even 2 different autoimmune diseases in the same patient, such as SPS and MG, offering promising prospects of changing the therapeutic algorithm in all neuroautoimmunities with potential for achieving even an immune reset shifting immunity to a healthy state.
    DOI:  https://doi.org/10.1212/NXI.0000000000200511
  4. Front Immunol. 2025 ;16 1608277
      Chimeric antigen receptors (CARs) are genetically engineered fusion proteins composed of extracellular antigen-recognition domains and multiple intracellular signaling domains. Although CAR T-cell immunotherapy has achieved significant advancements in treating hematologic malignancies, its application against solid tumors remains less successful. Key challenges-including production complexities, the scarcity of tumor-specific antigens, and limitations in cell trafficking and tumor infiltration-continue to impede therapeutic efficacy. Natural killer (NK) cells, essential innate immune lymphocytes, play a critical role in targeting malignant cells. Their unique antigen-recognition mechanisms, potent cytotoxicity, and favorable clinical safety profile position CAR NK cells as a promising alternative for targeted cancer therapy, especially for solid tumors. However, the transient persistence of NK cells in vivo and the technical challenges associated with their preparation currently limit the broader clinical adoption of this approach. This review examines the advantages of CAR NK cells immunotherapy and synthesizes current domestic and international research to advance the understanding of CAR NK cells therapeutics.
    Keywords:  CAR NK cells; NK; chimeric antigen receptors; immunotherapy; natural killer cells
    DOI:  https://doi.org/10.3389/fimmu.2025.1608277
  5. Nat Commun. 2025 Nov 21. 16(1): 10306
      Chimeric antigen receptor (CAR) T cells have become an established immunotherapy with promising results for the treatment of hematological malignancies. However, modulation of the targeted antigen's surface level in cancer cells affects the quality and safety of CAR-T cell therapy. Here we present an engineered yeast-based antigen system for simulation of cancer cells with precise regulation of surface-antigen densities, providing a tool for controlled activation of CAR T cells and systematic assessment of antigen density effects. This Synthetic Cellular Advanced Signal Adapter (SCASA) system uses G protein-coupled receptor signaling to control cancer antigen densities on the yeast surface and provides a customizable platform allowing selectable signal inputs and modular pathway engineering for precise output fine-tuning. In relation to CD19+ cancers, we demonstrate synthetic cellular communication between CD19-displaying yeast and human CAR T cells as well as applications in high-throughput characterization of different CAR designs. We show that yeast is an alternative to conventional technologies (e.g. microbeads) and can provide higher activation control of clinically derived CAR T cells in vitro, relative to cancer cells. In summary, we present a customizable yeast-based platform for high-throughput characterization of CAR-T cell functionality and show potential applications within therapeutic T cells in clinical settings.
    DOI:  https://doi.org/10.1038/s41467-025-65236-7
  6. Lupus Sci Med. 2025 Nov 19. pii: e001742. [Epub ahead of print]12(2):
       OBJECTIVE: To evaluate the safety and efficacy of CD19 chimeric antigen receptor (CAR) T cell therapy in a patient with refractory lupus nephritis who experienced severe disease flare during immunosuppressive washout, and to assess whether pulse corticosteroid intervention affects CAR T cell therapeutic outcomes.
    METHODS: We report a single case of a 22-year-old woman with SLE and lupus podocytopathy refractory to multiple therapies including rituximab, belimumab and obinutuzumab. The patient was treated under single-patient IND (#30146) with autologous CD19 CAR T cells (KYV-101). During the preinfusion washout period, she developed severe lupus flare requiring pulse intravenous methylprednisolone. Clinical outcomes, CAR T cell expansion, B cell depletion and laboratory parameters were monitored before and after therapy.
    RESULTS: Despite experiencing severe lupus flare (fever, rash, arthritis, anti-dsDNA elevation, hypocomplementaemia) during washout, pulse methylprednisolone (250 mg intravenous, rapidly tapered) successfully controlled symptoms without compromising subsequent CAR T cell expansion (peak 15.5 cells/µL on day 7). The patient achieved sustained clinical remission with SLE Disease Activity Index Score decreasing from 17 prior to leukapheresis to 4 by week 17. At 12 months postinfusion, she remained in drug-free remission with stable kidney function and had returned to full-time work.
    CONCLUSION: This case report illustrates that targeted pulse corticosteroids during CAR T cell therapy washout can effectively manage severe lupus flares without impairing therapeutic efficacy.
    Keywords:  Glucocorticoids; Lupus Erythematosus, Systemic; Lupus Nephritis
    DOI:  https://doi.org/10.1136/lupus-2025-001742
  7. J Transl Med. 2025 Nov 19. 23(1): 1316
      Chimeric antigen receptor (CAR)-T cell therapy has achieved significant progress in the treatment of hematologic cancers but continues to face major obstacles in solid tumors, including antigen heterogeneity, limited infiltration, and an immunosuppressive tumor microenvironment (TME). Oncolytic viruses (OVs) have emerged as promising tools to reshape the TME and improve CAR-T cell activity, yet many OVs encounter translational hurdles due to human seroprevalence and safety concerns. Newcastle disease virus (NDV), a naturally tumor-selective avian paramyxovirus, offers unique advantages as a non-integrating, non-pathogenic platform with a longstanding veterinary safety record and minimal pre-existing immunity in humans. NDV mediates direct oncolysis and immunogenic cell death, while simultaneously activating dendritic cells, repolarizing macrophages, and enhancing immune cell recruitment, thereby creating a TME that is more permissive to CAR-T cell therapy. Recent advances have enabled NDV to deliver immunostimulatory payloads locally within tumors, offering synergistic combinations to address CAR-T cell exhaustion and persistence. Looking ahead, further engineering efforts may expand the potential of this combined approach. This review summarizes the biological rationale, preclinical evidence, and translational prospects for integrating NDV with CAR-T cell therapy to improve outcomes in solid tumors.
    Keywords:  CAR-T cell therapy; Immunomodulation; Immunotherapy; Newcastle disease virus; Oncolytic virus; Tumor microenvironment; Virotherapy
    DOI:  https://doi.org/10.1186/s12967-025-07342-0
  8. J Hematol Oncol. 2025 Nov 20. 18(1): 105
      Chimeric antigen receptor T cell (CAR-T) therapy has emerged as a highly efficacious treatment for refractory and relapsed hematological malignancies in recent years. However, the complex manufacturing procedures, stringent logistical requirements, and protracted production timelines associated with autologous ex vivo CAR-T cells render them costly and inaccessible to many patients. In contrast, in vivo CAR-T therapy directly delivers CAR-encoding transgenes to endogenous T cells, reprogramming them in situ. This approach obviates the need for apheresis, ex vivo cell manufacturing, and lymphodepleting chemotherapy inherent in conventional CAR-T therapy. Consequently, in vivo CAR-T represents a more efficient and economical paradigm, transforming CAR-T from individualized cellular products towards truly "ready-to-use" therapeutics. This review summarizes the latest research progress in in vivo CAR cell therapies, spanning from bench to bedside, to provide insights for advancing their clinical translation.
    Keywords:  Adeno-associated virus; Gene delivery; In vivo CAR cell therapy; Lentiviral vectors; Lipid nanoparticles; Nucleic acid payloads; Polymer nanoparticles
    DOI:  https://doi.org/10.1186/s13045-025-01759-2
  9. Int Immunopharmacol. 2025 Nov 13. pii: S1567-5769(25)01826-0. [Epub ahead of print]168(Pt 1): 115838
      Chimeric Antigen Receptor T-cell (CAR-T) therapy has revolutionized cancer immunotherapy, achieving remarkable success in hematological malignancies. However, its clinical application to solid tumors is hindered by significant challenges, including tumor antigen heterogeneity, the immunosuppressive tumor microenvironment (TME), and physical barriers that limit effective T-cell infiltration. This review examines the intricate mechanisms of CAR-T therapy, with a focus on T-cell engineering, activation, and tumor targeting, highlighting the interplay between therapeutic design and tumor-specific complexities. The barriers unique to solid tumors, such as immune evasion mediated by suppressive cytokines and regulatory cells, tumor antigen escape, and the resilience of the extracellular matrix, are critically analyzed. Innovative strategies, including multi-antigen targeting constructs, logic-gated CARs for tumor-selective activation, and armored CAR-T cells equipped to counteract immunosuppressive signals, are evaluated for their potential to enhance therapeutic efficacy. Furthermore, the incorporation of matrix-degrading enzymes and immune checkpoint inhibitors is discussed as a means to overcome physical and immune-mediated resistance. Emerging targets such as B7-H3, Claudin 18.2, and MUC1, along with advancements in companion diagnostics, are reshaping the landscape of CAR-T therapy by enabling more precise patient selection and real-time therapeutic monitoring. This review synthesizes recent progress and persisting challenges, aiming to provide a comprehensive framework for advancing CAR-T therapy into a transformative modality for the treatment of solid tumors.
    Keywords:  CAR-T cells; Immune checkpoint inhibitors; Solid tumors; Tumor microenvironment
    DOI:  https://doi.org/10.1016/j.intimp.2025.115838
  10. Front Immunol. 2025 ;16 1699894
      Chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment of relapsed or refractory hematologic malignancies. While its clinical efficacy is well established, CAR T-cell therapy is frequently associated with severe immune-mediated toxicities, including cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), coagulopathy, and hemophagocytic lymphohistiocytosis-like syndrome (IEC-HS). Increasing evidence suggests that endothelial dysfunction, hemostatic imbalance, and complement activation are key contributors to the pathogenesis of these complications. Substantial research efforts have focused on identifying circulating biomarkers capable of predicting toxicity onset and severity, as well as stratifying patients at risk for early non-relapse mortality. In this review, we summarize the current understanding of the pathophysiological mechanisms underlying early CAR T cell-related toxicities, with particular emphasis on biomarkers of endotheliopathy and related pathways involved in their development. We focus on highlighting translational biomarkers with potential diagnostic, prognostic, and monitoring value that could be implemented in clinical practice to improve patient risk stratification, differential diagnosis, and therapeutic follow-up.
    Keywords:  CAR T toxicities; CRS; ICANS; IEC-HS; biomarker panels; biomarkers; coagulopathy; prediction
    DOI:  https://doi.org/10.3389/fimmu.2025.1699894
  11. Front Immunol. 2025 ;16 1724641
      
    Keywords:  CAR-NK; CAR-T cell therapy; chimeric antigen receptor; clinical application; immunotherapy
    DOI:  https://doi.org/10.3389/fimmu.2025.1724641
  12. Discov Oncol. 2025 Nov 13.
       BACKGROUND: Lymphoma continues to pose a major global health challenge. While the application of nanoparticles (NPs) in lymphoma research has shown considerable promise, a comprehensive evaluation of research trends and the underlying knowledge structure remains lacking. This study aims to systematically evaluate the current progress and future trends in NP applications for lymphoma.
    METHODS: We performed a bibliometric analysis using CiteSpace, VOSviewer, and R software on 1040 publications retrieved from the Web of Science Core Collection (WoSCC) between 2004 and 2024. Data on publications, countries, institutions, journals, authors, references, and keywords were analyzed.
    RESULTS: Research output demonstrated a consistent upward trend, with China and the United States emerging as the leading contributors. The Chinese Academy of Sciences had the highest output, while the University of North Carolina led in citation impact. The International Journal of Nanomedicine published the most studies, whereas Nature Reviews Drug Discovery ranked first in citations. Partha Partim Manna was the most prolific author, and Jindrich Kopecek's work had the strongest influence. Our analysis revealed crucial research areas, including enhanced drug delivery, nanovaccines, nanodiagnostics and monitoring, green synthesis, and the optimization of chimeric antigen receptor (CAR)-T cell therapy.
    CONCLUSION: This bibliometric analysis suggests that several directions in NP applications for lymphoma may hold promise for near-term progress. NP-mediated chemotherapy and targeted therapy could improve drug delivery and help overcome resistance, while NP-based imaging agents may enhance sensitivity and enable real-time monitoring. The integration of NPs with CAR-T therapy may also offer strategies to improve efficacy and safety. Additional avenues, such as nanovaccines and green synthesis, highlight opportunities for immune modulation and biocompatibility. Despite notable translational challenges, multifunctional NP platforms may gradually contribute to advances in lymphoma management.
    Keywords:  Bibliometrics; Drug delivery; Lymphoma; Nanoparticles; Therapy
    DOI:  https://doi.org/10.1007/s12672-025-04064-z
  13. Life Sci. 2025 Nov 14. pii: S0024-3205(25)00723-4. [Epub ahead of print]384 124087
      CRISPR is a programmable genome editing method that has transformed genome engineering and advanced cancer research. Various engineering iterations of CRISPR, including base and prime editing, facilitate more accurate and adaptable genomic manipulations, opening new frontiers in understanding and combating cancer. This review emphasizes CRISPR's significant influence in cancer biology, from its ability to accurately recreate somatic mutations in cellular and animal models to its application in developing precise diagnostic tools. We also examine advancements in cancer therapies, particularly through the ex vivo engineering of robust chimeric antigen receptor (CAR) T cells that perform effectively in restrictive tumor microenvironments. Lastly, we consider the existing limitations of CRISPR-Cas systems and outline future directions that could further enhance the application of this powerful technology against cancer.
    Keywords:  CAR-T cell therapy; CRISPR/Cas9; Cancer; Genome editing; Oncoviruses
    DOI:  https://doi.org/10.1016/j.lfs.2025.124087
  14. Methods Mol Biol. 2026 ;2990 19-38
      T cells carry out their functions by specifically recognizing antigens through their T cell receptors (TCRs), making the identification of TCR specificities crucial for understanding health and disease. Over the past decade, several antigen discovery methods have been developed to deconvolute TCRs. Our laboratory has established a cell-based antigen discovery platform based on chimeric receptors termed "SABRs" (Signaling and Antigen-presenting Bifunctional Receptors). SABRs are extracellular peptide-MHC complexes fused to intracellular signaling domains. SABRs present a covalently linked epitope to T cells and induce a readable signal upon their recognition. Moreover, large epitope libraries in SABRs can be constructed to screen the cognate epitopes of a given TCR. The modularity and continued refinement of SABRs allow antigen discovery for CD8+ and CD4+ T cells. In this chapter, we provide an overview of the SABR platform, followed by a detailed description of the steps involved in the generation of SABR libraries and performing SABR screens to deconvolute TCR specificities.
    Keywords:  Antigen discovery; Epitope libraries; SABR libraries; T cell receptor deconvolution
    DOI:  https://doi.org/10.1007/978-1-0716-4997-8_2
  15. Biomed Pharmacother. 2025 Nov 20. pii: S0753-3322(25)01000-5. [Epub ahead of print]193 118806
      Cholangiocarcinoma (CCA) is an aggressive epithelial malignancy characterized by poor prognosis, limited treatment options, and high recurrence rates even after surgery. Chimeric antigen receptor (CAR) T cell therapy has achieved notable success in hematologic malignancies, but its efficacy in solid tumors such as CCA is hindered by the immunosuppressive tumor microenvironment, particularly through PD-1/PD-L1 axis. To address these barriers, we developed a sixth-generation CAR T cell, A20 CAR6, incorporating the A20 peptide, a high-affinity ligand for integrin αvβ6-a tumor-associated antigen frequently overexpressed in CCA. Beyond antigen targeting, A20 CAR6 T cells are engineered to secrete a bispecific protein engager (BiPE) that binds PD-L1 on tumor cells and CD3 on T cells. This dual-function design aims to neutralize PD-L1-mediated immune suppression and recruit both CAR and bystander T cells to enhance tumor killing. Compared with conventional fourth-generation A20 CAR4 T cells lacking BiPE secretion, A20 CAR6 T cells exhibited superior cytotoxicity, cytokine production, and proliferation against integrin αvβ6+/PD-L1+ CCA cells. Notably, the secreted αPD-L1/αCD3 BiPE augmented CAR T cell activity and redirected non-engineered T cells to target tumor cells, amplifying the overall anti-tumor response. These findings suggest that A20 CAR6 T cells represent a promising next-generation immunotherapy with the potential to overcome key resistance mechanisms in CCA and improve treatment outcomes.
    Keywords:  Bi-specific protein engager; CAR T cell therapy; Cholangiocarcinoma; Integrin αvβ6; Programmed death-ligand 1
    DOI:  https://doi.org/10.1016/j.biopha.2025.118806
  16. J Hematol Oncol. 2025 Nov 19. 18(1): 104
      Chimeric antigen receptor T-cell (CAR-T) therapy has demonstrated a transformative impact in hematologic malignancies and offers a promising strategy to offer new hope for patients with solid tumors who have failed multiple lines of treatment. The clinical application of CAR-T therapy in solid tumors, however, still has challenges, including tumor heterogeneity, an immunosuppressive tumor microenvironment, and safety concerns. These hurdles have mean that CAR-T therapy has become both a focal point and a pivotal trend in contemporary clinical research. The American Society of Clinical Oncology (ASCO) Annual Meeting serves as a premier venue for unveiling groundbreaking clinical data. In this review, we highlight the main phase I clinical trial advances in CAR-T therapy for solid tumors presented at the 2025 ASCO Meeting.
    Keywords:  CAR-T; Clinical trials; Solid tumors
    DOI:  https://doi.org/10.1186/s13045-025-01760-9
  17. Front Immunol. 2025 ;16 1712344
      Immunoglobulin M (IgM) antibodies are gaining renewed attention as next-generation platforms for cancer immunotherapy. Compared with IgG, IgM exhibits distinct biological advantages, including higher avidity from multivalent binding, potent complement activation, and enhanced recognition of heterogeneous tumor antigens within immunosuppressive microenvironments. These attributes position IgM as a promising candidate for solid tumor therapy, despite the absence of currently approved IgM-based therapeutics. Recent advances in genetic engineering, antibody design, and protein manufacturing have enabled the generation of diverse IgM formats-ranging from monoclonal and bispecific constructs to engineered IgM derivatives-demonstrating substantial antitumor potential in preclinical and early translational studies. Nonetheless, clinical development faces persistent challenges, including short serum half-life, restricted tumor penetration, structural and biophysical complexity, and scalability of production. In this review, we discuss the structure and biology of IgM, highlight progress in developing novel IgM-based antibody formats for solid tumors, and critically examine the key translational barriers and future opportunities. Together, these insights underscore the therapeutic promise of IgM and chart a path toward its integration into the next generation of antibody-based cancer immunotherapies.
    Keywords:  IgM; antibody therapy; clinical translations; immunotherapy; solid tumor
    DOI:  https://doi.org/10.3389/fimmu.2025.1712344
  18. Liver Int. 2025 Dec;45(12): e70450
       BACKGROUND & AIMS: The prognosis for patients with hepatocellular carcinoma (HCC) remains suboptimal, despite the rapid advancement of anti-cancer immunotherapy. Chimeric antigen receptor (CAR) T cell therapy targeting glypican-3 (GPC3) has been developed for HCC; however, clinical trials have demonstrated heterogeneous responses among patients and limited CAR-T cell infiltration. Locoregional administration has emerged as a promising strategy for CAR-T therapy against solid tumours, yet its potential for HCC treatment has not been thoroughly explored.
    METHODS: In this study, we constructed anti-GPC3 CAR-T cells and examined their therapeutic efficacy through locoregional and systemic administration using multiple HCC xenograft mouse models.
    RESULTS: Comparison of CAR-T cell injections via portal vein and tail vein in mice with orthotopic HepG2 tumours demonstrated significantly enhanced tumour growth inhibition with locoregional CAR-T therapy. Consistently, tumour infiltration of CAR-T cells was significantly enhanced by portal vein injection and correlated with increased cytotoxicity, enhanced chemotaxis and reduced exhaustion of the tumour-infiltrating CAR-T cells compared to the tail vein injection group. Treatment with escalating CAR-T cell dosages resulted in further improved functionality of CAR-T cells and treatment efficacy, alongside improved liver function. Furthermore, portal vein injection exhibited superior tumour inhibition compared to tail vein injection in a metastatic model concurrently bearing orthotopic and extrahepatic tumour lesions.
    CONCLUSION: Collectively, our study demonstrates that locoregional CAR-T therapy through the portal vein is associated with increased CAR-T cell infiltration and improved therapeutic efficacy, offering promise for the treatment of both early- and late-stage patients.
    Keywords:  CAR‐T therapy; GPC3; hepatocellular carcinoma (HCC); locoregional therapy
    DOI:  https://doi.org/10.1111/liv.70450
  19. Adv Ther. 2025 Nov 21.
    on behalf of the Rare Disease Clinical Outcome Assessment Consortium
      Gene therapies are emerging as a promising strategy for the treatment of rare genetic diseases, for which treatment options are often limited and do not address the underlying disease mechanisms. However, there are significant challenges for gene therapy programs, including defining a suitable first-in-human cohort and selecting endpoints with appropriate variability, sensitivity, reliability, and clinical meaningfulness; a systematic framework for the assessment and approval of these treatments is lacking. In this review, we share insights from 12 clinical development programs that culminated in recent approvals of gene therapies for rare genetic diseases (2016-2023). These approvals highlight useful strategies for navigating the unique challenges of gene therapy trials, including early and frequent engagement with regulatory bodies, incorporating the patient voice, selecting meaningful clinical outcome assessments and suitable controls, and leveraging well-matched real-world data to understand long-term efficacy, durability, and safety. By systematically documenting and analyzing detailed examples in this review, it becomes possible to derive data-driven solutions that can inform the design of future studies. Such solutions may diverge from prior assumptions or preconceptions but can provide a more evidence-based foundation for improving trial efficiency, and ultimately accelerate the development of urgently needed therapies for patients with rare genetic diseases.
    Keywords:  Clinical development; Clinical outcome assessments; Clinical trials; Endpoints; Gene therapy; Genetic disorders; Marketing authorization; Rare diseases; Regulatory approval; Study design
    DOI:  https://doi.org/10.1007/s12325-025-03385-3
  20. Curr Opin HIV AIDS. 2025 Nov 21.
       PURPOSE OF REVIEW: HIV infects a broad array of tissues throughout the body. Consequently, any successful HIV cure strategy will need to target tissue HIV reservoirs, in addition to peripheral blood. Here we review recent immunotherapy approaches for HIV cure, with a focus on their ability to target viral tissue reservoirs, including immune privileged sites like the central nervous system (CNS).
    RECENT FINDINGS: Recent clinical trials of immunotherapy for HIV cure have demonstrated viral control in a subset of participants. T cell therapies, especially chimeric antigen receptor (CAR) T cells that can be targeted to lymphoid tissue, are highly promising, as are monoclonal antibody therapies such as broadly neutralizing antibodies to suppress HIV viremia and immune checkpoint inhibitors to enhance immune function. Despite this success, the penetration of many of these agents into the CNS is limited, and this remains a barrier to more widespread success of these therapies.
    SUMMARY: Immunotherapies represent a promising path toward an HIV cure, however their ability to target viral reservoirs within tissues represents a major challenge. Combination approaches leveraging multiple immunotherapy strategies, and other agents to reduce the HIV reservoir will likely be required to achieve viral control in the absence of antiretroviral therapy.
    Keywords:  HIV cure; antibodies; central nervous system; chimeric antigen receptor T cells; immunotherapy; latency
    DOI:  https://doi.org/10.1097/COH.0000000000000996
  21. Cell Rep Med. 2025 Nov 18. pii: S2666-3791(25)00410-0. [Epub ahead of print]6(11): 102337
      In vivo CAR engineering is an emerging therapeutic platform for cancer and autoimmune diseases. Enabled by advanced delivery systems such as immune-evasive lentiviral vectors and targeted lipid nanoparticles, this strategy has progressed to early clinical testing, showing initial safety and efficacy. Here, key opportunities and translational hurdles are critically discussed.
    DOI:  https://doi.org/10.1016/j.xcrm.2025.102337
  22. J Immunother Cancer. 2025 Nov 18. pii: e013123. [Epub ahead of print]13(11):
       BACKGROUND: The translational study of chimeric antigen receptor (CAR) T-cell function, persistence, immunophenotype, and spatial localization after infusion is crucial for understanding factors that influence clinical outcomes. However, research has been limited by a lack of optimized tools to reliably detect CAR-engineered cells. To address this, we developed a novel platform to generate monoclonal antibodies (mAbs) targeting a linker peptide incorporated in single-chain variable fragments (scFvs) of most CAR constructs.
    METHODS: Using recombinant proteins and scFv linker peptides as immunogens, we generated murine mAbs against the Whitlow linker peptide, capable of binding cells expressing Whitlow linker-containing CARs in both fresh and formalin-fixed paraffin-embedded (FFPE) tissues. We evaluated these antibodies in multiple in vitro translational applications relevant to CAR T-cell research and manufacturing.
    RESULTS: We identified five unique mAbs reactive against the Whitlow linker and characterized their binding properties and three-dimensional structural conformation. One clone was evaluated in depth, demonstrating comparable capacity to identify CAR T cells in peripheral blood relative to other methods using anti-idiotype antibodies or recombinant CAR-target proteins. In contrast to these reagents, the anti-Whitlow mAb detects cells expressing Whitlow linker-containing CARs with different antigen specificities, including those harboring the widely employed anti-CD19 FMC63-derived scFv as well as other scFvs, such as those targeting B-cell maturation antigen (BCMA) or CD33. Importantly, the anti-Whitlow mAb identified CAR T cells in situ in archival FFPE tissues, and a DNA-barcoded format enabled their spatial characterization and immunophenotyping in highly multiplexed immunohistochemistry. We also assessed the functional consequences of antibody binding on CAR T cells in vitro and demonstrated the feasibility of anti-Whitlow mAb-mediated selective enrichment of CAR-expressing T cells for potential utility in manufacturing workflows.
    CONCLUSIONS: Anti-Whitlow mAb clones exhibited distinct structural and functional properties that can be leveraged for multiple applications, providing versatile tools for detection, selection and manipulation of a broad range of clinical and preclinical CAR T-cell products.
    Keywords:  Antibody; Chimeric antigen receptor - CAR; Histology; Immunotherapy; T cell
    DOI:  https://doi.org/10.1136/jitc-2025-013123
  23. Front Immunol. 2025 ;16 1676112
      Cancer immunotherapy represents a major breakthrough in oncology, particularly with immune checkpoint inhibitors (ICIs) and CAR-T cell therapies. Despite improved outcomes, challenges such as immune-related adverse events (irAEs) and treatment resistance limit clinical use. Artificial intelligence (AI) offers new opportunities to address these barriers, including target identification, efficacy prediction, toxicity monitoring, and personalized treatment design. This review highlights recent advances in AI applications for biomarker discovery, safety evaluation, gene editing, nanotechnology, and microbiome modulation, integrating evidence from clinical and preclinical studies. We also discuss future directions and challenges in applying AI to cancer immunotherapy, aiming to support further research and clinical translation.
    Keywords:  CAR-T cell therapy; artificial intelligence; biomarkers; cancer immunotherapy; immune checkpoint inhibitors; machine learning; predictive modeling; safety assessment
    DOI:  https://doi.org/10.3389/fimmu.2025.1676112
  24. Front Immunol. 2025 ;16 1675251
      Anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitis (AAV) is a rare autoimmune disease characterized by the inflammation of small vessels. It is most commonly caused by ANCA targeting proteinase 3 (PR3) and myeloperoxidase (MPO) which are found in neutrophil lysosomes. The most common affected organs are respiratory tracts and kidneys, though other organs can be involved too. Although the cause of disease between PR3-AAV and MPO-AAV is similar, they vary in pathogenesis. Epigenetic and genetic factors may play a role in the disease development as certain population such as Chinese with HLA-DRB1*04:05 are more prevalent in MPO-AAV patient population. The prognosis for them is usually poor, often resulting in end-stage renal failure even with existing treatment. Current treatment for AAV relies heavily on corticosteroids which are toxic for long-term usage. Hence, there is a strong need to develop new, less toxic and targeted therapy for this disease. Regulatory T cell (Treg) therapy is a new type of therapy with the potential to specifically re-establish tolerance to the target autoantigen (MPO or PR3). This review will delve into the pathogeneses of AAV and discuss the potential of using genetically engineered Tregs to treat the cause of disease.
    Keywords:  ANCA; HLA; MPO-AAV; TCR-T and CAR-T therapy; preclinical model
    DOI:  https://doi.org/10.3389/fimmu.2025.1675251
  25. J Immunother Cancer. 2025 Nov 18. pii: e013120. [Epub ahead of print]13(11):
       BACKGROUND: Chimeric antigen receptor (CAR)-T cell therapy has achieved significant success in hematologic malignancies; however, its efficacy in solid tumors remains limited. A major limitation is the difficulty in identifying suitable target antigens that are abundantly expressed on the surface of tumor cells while sparing life-sustaining normal tissues.
    METHODS: We identified MUC17, a membrane-tethered mucin-type glycoprotein with minimal expression in normal tissues and frequent upregulation in gastric cancers, as a potential target for CAR-T therapy. We developed and validated MUC17-specific CAR-T cells incorporating a 4-1BB/CD3ζ signaling domain. In vitro assays assessed cytotoxicity, cytokine secretion, and T cell phenotypes across multiple gastric cancer cell lines, including CRISPR-mediated MUC17 knockout controls. In vivo efficacy was evaluated using NSG xenograft models.
    RESULTS: MUC17 CAR-T cells exhibited potent, antigen-specific cytotoxicity, robust cytokine release, and sustained effector functions characterized by enrichment of central memory phenotypes. In vivo, MUC17 CAR-T cells significantly suppressed tumor growth without signs of toxicity in GSU and ASPC-1 models.
    CONCLUSIONS: These findings support MUC17 as a promising immunotherapeutic target for gastric cancer and demonstrate how targeting glycocalyx-associated antigens can expand the range of surface proteins amenable to CAR-T cell-based therapies in solid tumors.
    Keywords:  Chimeric antigen receptor - CAR; Gastric Cancer; Immunotherapy
    DOI:  https://doi.org/10.1136/jitc-2025-013120
  26. Transplant Cell Ther. 2025 Nov 19. pii: S2666-6367(25)01601-X. [Epub ahead of print]
       BACKGROUND: Chimeric antigen receptor T-cell (CAR T) therapy has been shown to improve survival for patients with diffuse large B-cell lymphoma (DLBCL), but it comes at a high cost. Few studies have assessed the impact of CAR T on the health utilities of patients outside clinical trials. This information is important for economic evaluations aimed at determining the value for money of CAR T therapy and for guiding patient care decisions.
    OBJECTIVE: This objective of this study was to evaluate the impact of CAR T-cell therapy on the health utilities of patients diagnosed with relapsed/refractory (r/r) DLBCL.
    STUDY DESIGN: A prospective, longitudinal study was conducted at Princess Margaret Cancer Centre (Toronto) from April 2022-March 2023. Patients were assessed at baseline, 2 weeks, 3 months, and 6 months post-treatment. Patients completed the EQ-5D-5L and EQ-5D-5L VAS, the EORTC QLQ-C30, and a clinical data form. Using a Canadian valuation algorithm and population weights, the EQ-5D-5L and EORTC QLQ-C30 were converted to utility values. The EORTC QLQ C-30 was converted to the QLU-C10D. Descriptive analysis was conducted for each assessment, the mean utility scores and mean change from baseline were calculated. We used weighted generalized estimating equations to examine the predictors of health utility values.
    RESULTS: A total of 55 patients were treated with CAR T (mean age 58 years, 55% male). The baseline scores were 0.82 ± 0.12 (EQ-5D-5L), 0.67 ± 0.23 (QLU-C10D), and 74 ± 17 (VAS). The mean change from baseline and 2 weeks post-treatment was -0.044, -0.163, and -8.54, respectively. From baseline to 6 months post-treatment, the change was -0.053, -0.002, and 3.3, respectively. Progression status was a significant predictor of utility scores across all instruments.
    Keywords:  Chimeric antigen receptor T-cell therapy; patient reported outcomes; quality of life
    DOI:  https://doi.org/10.1016/j.jtct.2025.11.026
  27. Immunooncol Technol. 2025 Dec;28 101076
      The T-cell receptor (TCR) repertoire, representing the vast diversity of T cells, is a cornerstone of adaptive immunity and a powerful tool in oncology. Advances in high-throughput sequencing have enabled deep profiling of TCR diversity and clonality, highlighting the repertoire as a promising biomarker for cancer diagnosis, prognosis and therapeutic monitoring. This review synthesizes the current understanding of TCR repertoire analysis in cancer care. Distinct TCR features in tumors and peripheral blood can differentiate cancer patients from healthy individuals and help stage disease. Prognostically, a focused, clonal intratumoral repertoire is often associated with improved survival, whereas high diversity in peripheral blood typically reflects robust immune competence and better outcomes. In cancer immunotherapy, TCR profiling offers predictive insights; high baseline tumor clonality frequently correlates with response to anti-programmed cell death protein 1/programmed death-ligand 1 inhibitors, while greater peripheral diversity may predict benefit from anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4) therapy. Dynamic monitoring often shows an increase in clonality in patients responding to treatment. Furthermore, TCR analysis is integral to optimizing and tracking adoptive cell therapies and cancer vaccines. Despite this potential, significant challenges, including a lack of methodological standardization, currently limit widespread clinical application. Integrating TCR analysis with multi-omic and single-cell technologies is essential to overcoming these hurdles and advancing personalized immunotherapy.
    Keywords:  T-cell receptors; biomarker; cancer immunotherapy; immune profiling; immune repertoire; repertoire clonality; repertoire diversity
    DOI:  https://doi.org/10.1016/j.iotech.2025.101076
  28. Sci Transl Med. 2025 Nov 19. 17(825): eads4214
      Glypican-2 (GPC2) and the disialoganglioside GD2 are validated CAR T cell targets in neuroblastoma, but durable clinical responses remain limited. This modest chimeric antigen receptor T cell (CAR T cell) efficacy is in part due to suboptimal T cell persistence, antigen down-regulation, and a hostile tumor microenvironment, which includes immune cell-modulating extracellular vesicles (EVs). Neuroblastoma-derived EVs may contain CAR targets or other immunoregulatory elements that can modulate CAR T cell antitumor activity. Thus, we first profiled the surfaceome of neuroblastoma EVs and assessed their impact on both GPC2 and GD2 CAR T cell function. Neuroblastoma EVs displayed GPC2 and GD2, with minimal expression of programmed death-ligand 1 (PD-L1), and were detected in blood from tumor-bearing mice and patients. These EVs directly activated paired CAR T cells, suggesting a role for a peripheral source of CAR antigen. To exploit this therapeutically, we engineered nontumor-derived GPC2+ synthetic EVs (SyntEVs) as CAR T cell enhancers and armored them with either albumin-binding domains or GD2-binding domains. In mice harboring human neuroblastoma cell line-derived or patient-derived xenografts, serial infusion of armored SyntEVs after GPC2 CAR T cells enhanced tumor control by boosting peripheral CAR T cell persistence. Moreover, GD2-targeting SyntEVs decorated low-antigen tumor cells with GPC2, circumventing antigen down-regulation. This SyntEV platform offers a versatile system to address the therapeutic limitations of CAR T cells in solid tumors.
    DOI:  https://doi.org/10.1126/scitranslmed.ads4214
  29. Rheum Dis Clin North Am. 2026 Feb;pii: S0889-857X(25)00082-1. [Epub ahead of print]52(1): 203-216
      Integrative rheumatology is an emerging field that combines evidence-based conventional and complementary therapies to manage rheumatic diseases while addressing systemic, psychological, and environmental factors. Despite challenges such as skepticism, regulatory gaps, funding gaps, limited access, and lack of provider training, it has the potential to offer a patient-centered, cost-effective model that improves health outcomes and promotes planetary health. This approach emphasizes rigorous evidence-based practice, holistic care, and advocacy for equitable access to sustainable therapies, envisioning a health care paradigm that is both patient-centered and environmentally conscious.
    Keywords:  Complementary medicine; Human health; Integrative rheumatology; Planetary health
    DOI:  https://doi.org/10.1016/j.rdc.2025.09.005
  30. Lancet. 2025 Nov 12. pii: S0140-6736(25)01671-X. [Epub ahead of print]
       BACKGROUND: Lately, autologous CD19-targeting chimeric antigen receptor (CAR) T cells have shown excellent efficacy in treatment of autoimmune diseases, but with great safety concerns, such as infections. In this study, we aimed to evaluate the safety, tolerability, and efficacy of allogeneic CD19 CAR natural killer (NK)-cell therapy in patients with relapsed or refractory systemic lupus erythematosus (SLE).
    METHODS: In this open-label, single-arm, prospective, first-in-human case series, we evaluated allogeneic CD19 CAR NK-cell therapy in adult patients (aged 18-65 years) with relapsed or refractory SLE at one site in China. Patients who had received at least two previous standard systemic therapies and continued to exhibit moderate-to-severe disease activity were eligible for inclusion. This study consisted of schedule escalation and dose escalation, with schedule escalation from 7 days and dose escalation commencing at 0·75 × 109 CAR NK cells on day 0. All patients received a lymphodepleting conditioning regimen with fludarabine (25 mg/m2 per day) and cyclophosphamide (300 mg/m2 per day) administered daily from days -5 to -3, followed by three CAR NK-cell infusions within a single treatment cycle at identical dose levels and inter-infusion intervals. Dose-limiting adverse events were monitored in patients for 28 days. The primary endpoints of this study were safety and tolerability, including the incidence of dose-limiting toxicities and adverse events according to National Cancer Institute Common Terminology Criteria for Adverse Events version 5.0. This study was registered with ClinicalTrials.gov (NCT06010472) and follow-up is ongoing.
    FINDINGS: 18 patients with relapsed or refractory SLE with moderate-to-severe disease activity were enrolled between Aug 21, 2023, and June 16, 2024. Of the 18 patients, 17 (94%) were female; the median age was 37·5 years (IQR 32·0-39·8), and the median disease duration was 10·5 years (IQR 4·5-14·8). Patients had received at least two standard systemic therapies, including biological agents (belimumab and telitacicept) in 14 (78%) of 18 patients, and plasmapheresis in one patient. Cytokine release syndrome was reported in one (6%) of 18 patients (grade 1). Neurotoxicity and other CAR NK-cell therapy-related severe adverse events were not observed, and there were no dose-limiting toxicities. Of the nine patients with more than 12 months' follow-up, six (67%) attained DORIS remission and lupus low disease activity state.
    INTERPRETATION: This study suggests that allogeneic CAR NK-cell therapy is a potent option for treatment of autoimmune diseases and indicates that such a therapy might address limitations of current autologous CAR T-cell therapy, including manufacturing scale and time, access, safety, and cost.
    FUNDING: Shanghai Municipal Health Commission, Changhai Hospital Affiliated to Naval Medical University, and National Natural Science Foundation of China.
    TRANSLATION: For the Chinese translation of the abstract see Supplementary Materials section.
    DOI:  https://doi.org/10.1016/S0140-6736(25)01671-X
  31. Acad Radiol. 2025 Nov 19. pii: S1076-6332(25)01049-9. [Epub ahead of print]
      Foundation Models (FMs) mark a significant evolution in medical AI, enabling multimodal and multitask performance across text and imaging. Radiology, with its structured data formats and early adoption of AI, is uniquely positioned to benefit from FM capabilities. However, despite promising technical advances, questions remain about their clinical readiness, safety, and regulatory oversight. This narrative review explores the development, utility, and implementation challenges of FMs in U.S. radiology. Literature from PubMed, Scopus, arXiv, and IEEE Xplore (January 2022 to May 2025) was synthesized to highlight architectural trends, clinical applications, evaluation methods, and regulatory developments. U.S.-based models like CheXzero, BioMedCLIP, and Med-PaLM demonstrate strong diagnostic and reporting performance but face key limitations-including lack of FDA clearance, limited external validation, and integration barriers with PACS/RIS systems. Safety issues such as hallucination, automation bias, and underperformance in edge cases persist. While human-in-the-loop frameworks, federated learning, and emerging reporting standards show promise, institutional readiness and regulatory clarity remain fragmented. We propose a roadmap that includes continuous monitoring, equity-focused design, and a national FM registry to guide responsible deployment. Radiology's digital maturity makes it a critical testbed for foundational AI integration-offering lessons for broader clinical adoption.
    Keywords:  Bias and Health Equity; Clinical Validation; Diagnostic Imaging AI; FDA Regulation; Federated Learning; Foundation models; Human-in-the-Loop AI; Multimodal Learning; Radiology Artificial Intelligence; Vision-Language Models
    DOI:  https://doi.org/10.1016/j.acra.2025.10.063
  32. Front Immunol. 2025 ;16 1696914
      Sepsis remains a leading cause of critical illness worldwide. Despite advances in supportive care, durable benefit from immune-directed therapies is limited, reflecting heterogeneity with immune low-response states ('immunoparalysis') across innate and adaptive compartments. In this review we summarize advances from single-cell RNA and ATAC profiling, immune-repertoire assays and 3D spatial transcriptomics that resolve monocyte, dendritic-cell (cDC1, cDC2 and pDC), lymphocyte and NK-cell programs, and appraise translational opportunities spanning endotype-guided risk stratification, pharmacodynamic monitoring and spatial biomarkers. We also discuss enduring challenges-including assay standardization, harmonized thresholds for monocyte HLA-DR and whole-blood stimulation, and limited availability of clinically compatible spatial platforms-that temper implementation. By integrating bedside function (HLA-DR trajectories, LPS-induced cytokine capacity) with single-cell endotypes (MS1/HLA-DR^low S100A^high monocytes, dendritic-cell attrition, checkpoint-biased T cells) and host-pathogen topology from FFPE-ready spatial assays, emerging strategies aim to restore antigen presentation, reconstitute priming, disrupt inhibitory myeloid-lymphoid circuits and prevent secondary infection. Our synthesis provides an appraisal of the evolving landscape of immunoparalysis-informed precision medicine in sepsis and outlines pragmatic standards for composite biomarkers, patient selection and on-therapy decision rules. We hope these insights will assist investigators and clinicians as they endeavor to convert descriptive immune low-response states into tractable, reversible clinical entities.
    Keywords:  endotoxin tolerance; immune low-response states; immunoparalysis; monocyte HLA-DR; sepsis; single-cell RNA sequencing
    DOI:  https://doi.org/10.3389/fimmu.2025.1696914
  33. ACR Open Rheumatol. 2025 Nov;7(11): e70130
      Systemic autoimmune diseases (sAIDs) affect 1 in 10 people and represent a leading cause of death in women under the age of 65 years. Despite the emergence of targeted-biologic therapies, mortality rates for sAIDs have not improved. Emerging cellular therapies represent a promising therapeutic strategy for patients living with sAIDs given their potential to induce sustained remission and limit long-term exposure to glucocorticoids. Encouraging outcomes from a case series of patients with systemic lupus erythematosus, idiopathic inflammatory myopathy, and systemic sclerosis (SSc) have ignited clinical development in this therapeutic area. However, numerous questions remain regarding cellular therapy clinical trial design, ranging from the optimal time to intervene with cellular therapies during a sAID to the appropriate duration of the washout period, conditioning regimen, and management of serious treatment-related adverse events in these patients. Building the infrastructure and multidisciplinary teams necessary to conduct cellular therapy trials for sAIDs also represents a major challenge. To address these critical issues, the National Cancer Institute's Center for Cancer Research and the National Institute of Arthritis and Musculoskeletal and Skin Diseases convened a workshop in which stakeholders from diverse areas addressed strategies to bridge the gap between preclinical and clinical research, foster collaboration between rheumatologists and hematologists-oncologists, and develop optimal treatment protocols grounded in solid science. This manuscript summarizes the main concepts put forward and makes practical recommendations for advancing the development of cellular therapy for sAIDs.
    DOI:  https://doi.org/10.1002/acr2.70130
  34. Nano Lett. 2025 Nov 19.
      DNA origami has been widely employed in biomedical applications, including bioimaging, biosensing, and drug delivery. However, the gene-encoding capability of the DNA scaffold in DNA origami has rarely been developed. Herein, we introduce a universal strategy to construct genetically encoded DNA origami for CAR-T-based immunotherapy. Two complementary sense and antisense strands encoding the chimeric antigen receptor (CAR) can be easily included as DNA scaffolds in folding DNA origami. After hybridization, genetically encoded DNA origami can function as the template for lipid growth. With decoration of the CD3 antibody targeting T cells, the lipid-coated and genetically encoded DNA origami can efficiently recognize and penetrate the cell membrane of T cells. After targeted cellular uptake, a successful CAR expression can be achieved to capture related tumor cells for CAR-T-based immunotherapy. This rationally developed DNA-origami-based delivery system provides a new avenue for the development of CAR-T therapy.
    Keywords:  CAR-T; DNA nanotechnology; drug delivery; self-assembly; tumor immunotherapy
    DOI:  https://doi.org/10.1021/acs.nanolett.5c04782