Pub Date : 2026-01-14DOI: 10.1016/j.ymthe.2026.01.003
Hongzhi Zeng, Aidi Liu, Tyler C. Daniel, Devin A. Golla, Zhenyu Lu, Rebecca Serodio, Brigid A. Millette, Ananya Lingineni, Peretz Gilberd, Kelly Chee, Komal Talloo, Advaith Peddi, So Hyun Park, Gang Bao, Xue Gao
{"title":"Precision A3G Base Editors for Disease Modeling and Correction","authors":"Hongzhi Zeng, Aidi Liu, Tyler C. Daniel, Devin A. Golla, Zhenyu Lu, Rebecca Serodio, Brigid A. Millette, Ananya Lingineni, Peretz Gilberd, Kelly Chee, Komal Talloo, Advaith Peddi, So Hyun Park, Gang Bao, Xue Gao","doi":"10.1016/j.ymthe.2026.01.003","DOIUrl":"https://doi.org/10.1016/j.ymthe.2026.01.003","url":null,"abstract":"","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":"55 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145962411","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-10DOI: 10.1016/j.ymthe.2026.01.001
Gabriel A Barragán Bravo,David A de la Cerda,Elisa Landoni,Kshiti Dholakia,Piotr Humeniuk,Leidy D Caraballo Galva,Ying Wang,Gengwen Tian,Boning Yang,Linjie Guo,Michael S Wood,Xavier Rios,Xin Xu,Amy N Courtney,Erica J Di Pierro,Joan Jacob,Yi-Han Li,Akshaya Adaikkalavan,Norihiro Watanabe,Sufeng Mao,George Miles,Gianpietro Dotti,Leonid S Metelitsa
Invariant natural killer T cells (NKTs) have intrinsic anti-tumor properties that make them promising candidates for chimeric antigen receptor (CAR) immunotherapies. Transgenic cytokine expression can enhance cellular therapy potency, and we hypothesized that co-expressing IL-18 alone or with IL-15 would boost CAR-NKT therapeutic potential. To test this, we generated retroviral constructs expressing IL-15 and/or IL-18 with an inducible caspase 9 safety switch and co-transduced them with a GD2-specific CAR into human NKTs. Co-expression of IL-18 or IL-15/IL-18 increased CAR-NKT cytotoxicity, proliferation, and cytokine secretion in vitro compared to IL-15 alone. IL-18 also enhanced GPC3.CAR and CD19.CAR NKT activity against hepatocellular carcinoma and B cell leukemia cells, respectively. In a metastatic neuroblastoma model, IL-18-expressing GD2.CAR-NKTs controlled tumors more effectively than IL-15-only cells, but mice in the IL-15/IL-18 group developed severe toxicities not observed in the IL-18-only group. Mechanistically, IL-18 induced a transcriptional program distinct from IL-15, marked by lower exhaustion signatures and enrichment of metabolic pathways. Finally, targeted metabolomics showed that IL-18 drives broad metabolic reprogramming in CAR-NKTs including increased oxidative phosphorylation, glycolysis, glutaminolysis, and purine metabolism. These findings support the use of IL-18 in developing the next generation of cytokine-armed CAR-NKT cancer immunotherapies.
{"title":"IL-18 metabolically reprograms CAR-expressing natural killer T cells and enhances their antitumor activity.","authors":"Gabriel A Barragán Bravo,David A de la Cerda,Elisa Landoni,Kshiti Dholakia,Piotr Humeniuk,Leidy D Caraballo Galva,Ying Wang,Gengwen Tian,Boning Yang,Linjie Guo,Michael S Wood,Xavier Rios,Xin Xu,Amy N Courtney,Erica J Di Pierro,Joan Jacob,Yi-Han Li,Akshaya Adaikkalavan,Norihiro Watanabe,Sufeng Mao,George Miles,Gianpietro Dotti,Leonid S Metelitsa","doi":"10.1016/j.ymthe.2026.01.001","DOIUrl":"https://doi.org/10.1016/j.ymthe.2026.01.001","url":null,"abstract":"Invariant natural killer T cells (NKTs) have intrinsic anti-tumor properties that make them promising candidates for chimeric antigen receptor (CAR) immunotherapies. Transgenic cytokine expression can enhance cellular therapy potency, and we hypothesized that co-expressing IL-18 alone or with IL-15 would boost CAR-NKT therapeutic potential. To test this, we generated retroviral constructs expressing IL-15 and/or IL-18 with an inducible caspase 9 safety switch and co-transduced them with a GD2-specific CAR into human NKTs. Co-expression of IL-18 or IL-15/IL-18 increased CAR-NKT cytotoxicity, proliferation, and cytokine secretion in vitro compared to IL-15 alone. IL-18 also enhanced GPC3.CAR and CD19.CAR NKT activity against hepatocellular carcinoma and B cell leukemia cells, respectively. In a metastatic neuroblastoma model, IL-18-expressing GD2.CAR-NKTs controlled tumors more effectively than IL-15-only cells, but mice in the IL-15/IL-18 group developed severe toxicities not observed in the IL-18-only group. Mechanistically, IL-18 induced a transcriptional program distinct from IL-15, marked by lower exhaustion signatures and enrichment of metabolic pathways. Finally, targeted metabolomics showed that IL-18 drives broad metabolic reprogramming in CAR-NKTs including increased oxidative phosphorylation, glycolysis, glutaminolysis, and purine metabolism. These findings support the use of IL-18 in developing the next generation of cytokine-armed CAR-NKT cancer immunotherapies.","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":"3 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145947414","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bietti crystalline corneoretinal dystrophy (BCD) is an inherited retinal degeneration caused by biallelic variants in the CYP4V2 gene. Here, we report a phase 1/2, dose-escalation clinical study to assess the safety and efficacy of ZVS101e, an adeno-associated viral (AAV) mediated gene augmentation therapy (rAAV2/8-hCYP4V2), in 11 BCD patients with up to 365 days of follow-up (NCT05832684). ZVS101e showed a favorable safety profile, with no dose-limiting toxicities or drug-related serious adverse events. Inflammatory response was observed, primarily in the medium- and high-dose groups, which were effectively managed with corticosteroids. Efficacy was optimal in the low-dose group, with mean best-corrected visual acuity (BCVA) improvement of 14.0 letters and 50% of participants achieving clinically meaningful gains (≥15 letters). Functional assessments, including a multi-luminance mobility test (MLMT) and visual function questionnaire-25 (VFQ-25), along with retinal structure, demonstrated consistent improvements. These results highlight ZVS101e's therapeutic potential for BCD, supporting further clinical development.
{"title":"Gene therapy for Bietti crystalline corneoretinal dystrophy: a phase 1/2 clinical trial.","authors":"Liping Yang,Dongjun Xing,Jinlu Zhang,Boshi Liu,Xiaoshuang Jiang,Shaohong Chen,Wang Li,Baoyuan Qu,Jing Qiao,Xuefeng Feng,Haicun Jia,Xianjun Hu,Shicheng Yu,Ruixuan Jia,Likun Wang,Vinit B Mahajan,Hongliang Dou,Fang Lu,Xiaorong Li","doi":"10.1016/j.ymthe.2026.01.005","DOIUrl":"https://doi.org/10.1016/j.ymthe.2026.01.005","url":null,"abstract":"Bietti crystalline corneoretinal dystrophy (BCD) is an inherited retinal degeneration caused by biallelic variants in the CYP4V2 gene. Here, we report a phase 1/2, dose-escalation clinical study to assess the safety and efficacy of ZVS101e, an adeno-associated viral (AAV) mediated gene augmentation therapy (rAAV2/8-hCYP4V2), in 11 BCD patients with up to 365 days of follow-up (NCT05832684). ZVS101e showed a favorable safety profile, with no dose-limiting toxicities or drug-related serious adverse events. Inflammatory response was observed, primarily in the medium- and high-dose groups, which were effectively managed with corticosteroids. Efficacy was optimal in the low-dose group, with mean best-corrected visual acuity (BCVA) improvement of 14.0 letters and 50% of participants achieving clinically meaningful gains (≥15 letters). Functional assessments, including a multi-luminance mobility test (MLMT) and visual function questionnaire-25 (VFQ-25), along with retinal structure, demonstrated consistent improvements. These results highlight ZVS101e's therapeutic potential for BCD, supporting further clinical development.","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":"185 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145947430","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-10DOI: 10.1016/j.ymthe.2026.01.008
Wei Liu,Dan Wang,Jagadeesh K Venkatesan,Gertrud Schmitt,Henning Madry,Magali Cucchiarini
Osteoarthritis is a degenerative joint disease with an irreversible degradation of the articular cartilage that cannot be healed. We explored a novel gene therapy incorporating recombinant adeno-associated viral (rAAV) vectors in an alginate hydrogel to deliver reparative sox9 and TGF-β genes in human osteoarthritic articular chondrocytes (hOACs) and cartilage explants versus control (lacZ, RFP) genes. The rAAV/alginate hydrogel systems safely supported a prolonged rAAV vector release and overexpression of rAAV transgenes for up to 21 days in vitro and ex vivo. sox9 and TGF-β overexpression increased the deposition of extracellular matrix components (glycosaminoglycans/proteoglycans, type-II collagen) for up to 21 days versus respective controls (no vector, rAAV-lacZ in hydrogels) and relatively cell proliferation while reducing undesirable type-I and type-X collagen production although increasing mineralization. Stronger effects were noted with sox9 on glycosaminoglycan/proteoglycan deposition while TGF-β was more potent for cell proliferation and type-II collagen deposition. Multivariable mapping analyses validated the efficacy of the systems by segregating treatment groups from controls while elucidating the interrelationships of marker gene expression in vitro and across compartments ex vivo. These results show the value of rAAV/alginate hydrogel systems to promote the functional expression of reparative factors in human osteoarthritic cells and tissues for human osteoarthritis.
{"title":"Innovative In Vitro and Ex Vivo Human Osteoarthritis Gene Therapy via Alginate Hydrogel-Guided rAAV sox9 and TGF-β Gene Vector Delivery.","authors":"Wei Liu,Dan Wang,Jagadeesh K Venkatesan,Gertrud Schmitt,Henning Madry,Magali Cucchiarini","doi":"10.1016/j.ymthe.2026.01.008","DOIUrl":"https://doi.org/10.1016/j.ymthe.2026.01.008","url":null,"abstract":"Osteoarthritis is a degenerative joint disease with an irreversible degradation of the articular cartilage that cannot be healed. We explored a novel gene therapy incorporating recombinant adeno-associated viral (rAAV) vectors in an alginate hydrogel to deliver reparative sox9 and TGF-β genes in human osteoarthritic articular chondrocytes (hOACs) and cartilage explants versus control (lacZ, RFP) genes. The rAAV/alginate hydrogel systems safely supported a prolonged rAAV vector release and overexpression of rAAV transgenes for up to 21 days in vitro and ex vivo. sox9 and TGF-β overexpression increased the deposition of extracellular matrix components (glycosaminoglycans/proteoglycans, type-II collagen) for up to 21 days versus respective controls (no vector, rAAV-lacZ in hydrogels) and relatively cell proliferation while reducing undesirable type-I and type-X collagen production although increasing mineralization. Stronger effects were noted with sox9 on glycosaminoglycan/proteoglycan deposition while TGF-β was more potent for cell proliferation and type-II collagen deposition. Multivariable mapping analyses validated the efficacy of the systems by segregating treatment groups from controls while elucidating the interrelationships of marker gene expression in vitro and across compartments ex vivo. These results show the value of rAAV/alginate hydrogel systems to promote the functional expression of reparative factors in human osteoarthritic cells and tissues for human osteoarthritis.","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":"28 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145947416","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-10DOI: 10.1016/j.ymthe.2026.01.004
Niccolò Maurizi,Enrico Ammirati,Elizabeth Silver,Kimberly Hong,Quan Bui,Alessia Argirò,Iacopo Olivotto,Eric D Adler
Adeno-associated virus (AAV)-based gene replacement has emerged as a transformative platform for severe genetic disorders, yet immune-mediated adverse events (AEs) pose significant barriers to widespread clinical adoption. We performed a systematic review and meta-analysis of prospective and retrospective studies of AAV gene therapy published between January 2005 and March 2025 (PROSPERO CRD420251046546). Data from 801studies encompassing 1,972 patients and 2,142 patient-years were pooled to estimate the incidence and clinical impact of immunotoxicity. Random-effects meta-analysis yielded a 30.0 % (95 % CI 22.5-38.9; I2 = 83.1 %) overall AE rate, including hepatotoxicity in 23.8 % (17.4-31.7; I2 = 81.7 %), myocarditis in 6.2 % (4.6-8.1; I2 = 46 %), thrombotic microangiopathy (TMA) in 4.7 % (4.4-6.5; I2 = 18.5 %), and treatment-related death in 4.7 % (3.0-5.3; I2 = 46.1 %). Hepatotoxicity and myocarditis were generally mild (97 % and 96 % non-serious), whereas all TMA episodes carried substantial morbidity. Time-course analyses revealed TMA clustered in week 1, myocarditis at week 2, and hepatotoxicity up to six months post-infusion. In individual-patient analyses, vector serotype and doses > 1 × 1012 vg/kg significantly increased AE risk (OR 5.59 [1.35-12.2], p = 0.018; OR 2.31 [1.04-5.53], p = 0.041), whereas combined corticosteroid, anti-CD20, mTOR- and calcineurin-inhibitor regimens were protective (OR 0.67 [0.47-0.96], p = 0.040). At least 5 cases of TMA, 1 of myocarditis and three deaths could have not been included in the present analysis because these events were described in company statements. These findings underscore that one-third of AAV recipients experience immunotoxicity, predominantly early and mild, and support proactive immunosuppression and vector optimization to enhance safety.
基于腺相关病毒(AAV)的基因替代已成为严重遗传疾病的变革性平台,但免疫介导的不良事件(ae)对广泛的临床应用构成重大障碍。我们对2005年1月至2025年3月期间发表的AAV基因治疗的前瞻性和回顾性研究进行了系统回顾和荟萃分析(PROSPERO CRD420251046546)。来自801项研究的数据,包括1972例患者和2142例患者年,用于估计免疫毒性的发生率和临床影响。随机效应荟萃分析得出30.0% (95% CI 22.5-38.9; I2 = 83.1%)的AE总发生率,包括肝毒性23.8% (17.4-31.7;I2 = 81.7%)、心肌炎6.2% (4.6-8.1;I2 = 46%)、血栓性微血管病变(TMA) 4.7% (4.4-6.5; I2 = 18.5%)和治疗相关死亡4.7% (3.0-5.3;I2 = 46.1%)。肝毒性和心肌炎通常是轻微的(97%和96%非严重),而所有TMA发作都有很高的发病率。时间过程分析显示TMA聚集在第1周,第2周出现心肌炎,肝毒性持续到输注后6个月。在个体患者分析中,载体血清型和剂量> × 1012vg /kg显著增加AE风险(OR 5.59 [1.35-12.2], p = 0.018; OR 2.31 [1.04-5.53], p = 0.041),而皮质类固醇、抗cd20、mTOR和钙调磷酸酶抑制剂联合治疗方案具有保护作用(OR 0.67 [0.47-0.96], p = 0.040)。至少有5例TMA, 1例心肌炎和3例死亡病例可能未包括在本分析中,因为这些事件已在公司声明中描述。这些发现强调,三分之一的AAV受体出现免疫毒性,主要是早期和轻度的,支持主动免疫抑制和载体优化以提高安全性。
{"title":"Incidence, Timing and Clinical Significance of Adverse Immune Events after Gene Replacement Therapy: a Systematic Review and Meta-Analysis.","authors":"Niccolò Maurizi,Enrico Ammirati,Elizabeth Silver,Kimberly Hong,Quan Bui,Alessia Argirò,Iacopo Olivotto,Eric D Adler","doi":"10.1016/j.ymthe.2026.01.004","DOIUrl":"https://doi.org/10.1016/j.ymthe.2026.01.004","url":null,"abstract":"Adeno-associated virus (AAV)-based gene replacement has emerged as a transformative platform for severe genetic disorders, yet immune-mediated adverse events (AEs) pose significant barriers to widespread clinical adoption. We performed a systematic review and meta-analysis of prospective and retrospective studies of AAV gene therapy published between January 2005 and March 2025 (PROSPERO CRD420251046546). Data from 801studies encompassing 1,972 patients and 2,142 patient-years were pooled to estimate the incidence and clinical impact of immunotoxicity. Random-effects meta-analysis yielded a 30.0 % (95 % CI 22.5-38.9; I2 = 83.1 %) overall AE rate, including hepatotoxicity in 23.8 % (17.4-31.7; I2 = 81.7 %), myocarditis in 6.2 % (4.6-8.1; I2 = 46 %), thrombotic microangiopathy (TMA) in 4.7 % (4.4-6.5; I2 = 18.5 %), and treatment-related death in 4.7 % (3.0-5.3; I2 = 46.1 %). Hepatotoxicity and myocarditis were generally mild (97 % and 96 % non-serious), whereas all TMA episodes carried substantial morbidity. Time-course analyses revealed TMA clustered in week 1, myocarditis at week 2, and hepatotoxicity up to six months post-infusion. In individual-patient analyses, vector serotype and doses > 1 × 1012 vg/kg significantly increased AE risk (OR 5.59 [1.35-12.2], p = 0.018; OR 2.31 [1.04-5.53], p = 0.041), whereas combined corticosteroid, anti-CD20, mTOR- and calcineurin-inhibitor regimens were protective (OR 0.67 [0.47-0.96], p = 0.040). At least 5 cases of TMA, 1 of myocarditis and three deaths could have not been included in the present analysis because these events were described in company statements. These findings underscore that one-third of AAV recipients experience immunotoxicity, predominantly early and mild, and support proactive immunosuppression and vector optimization to enhance safety.","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":"57 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145947417","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-10DOI: 10.1016/j.ymthe.2026.01.007
John T Keane,David A Degaramo,Heather M Sosnoski,Shawna K Brookens,Hyeon-Gyu S Lewis,Fang Liu,Avery D Posey
Chimeric antigen receptor (CAR) T cell therapy achieves durable remissions in hematological malignancies, yet its success against solid tumors is blunted in part by the tumors' highly immunosuppressive microenvironment. Fourth-generation "armored" CAR T cells are engineered to secrete pro-inflammatory molecules to counteract this barrier. Here, we engineered TnMUC1-targeted CAR T cells that constitutively secrete either single-chain interleukin-12 (scIL-12) or single-chain interleukin-23 (scIL-23). Both cytokine-armored CAR T cell formats improved effector function in vitro, increasing interferon-gamma production and cytotoxicity compared with their unarmored counterparts. scIL-12- and scIL23-secreting CAR T cells significantly delayed tumor growth and prolonged survival in mouse xenograft models of human breast and prostate cancer, while scIL-23 secretion led to increases in vivo persistence and retention of early differentiation states. These findings nominate scIL-23 armoring as a promising strategy to extend CAR T cell therapy to solid tumors.
{"title":"Single-Chain IL-23 Secretion by CAR T Cells Improves Tumor Control and Persistence Against Solid Tumors.","authors":"John T Keane,David A Degaramo,Heather M Sosnoski,Shawna K Brookens,Hyeon-Gyu S Lewis,Fang Liu,Avery D Posey","doi":"10.1016/j.ymthe.2026.01.007","DOIUrl":"https://doi.org/10.1016/j.ymthe.2026.01.007","url":null,"abstract":"Chimeric antigen receptor (CAR) T cell therapy achieves durable remissions in hematological malignancies, yet its success against solid tumors is blunted in part by the tumors' highly immunosuppressive microenvironment. Fourth-generation \"armored\" CAR T cells are engineered to secrete pro-inflammatory molecules to counteract this barrier. Here, we engineered TnMUC1-targeted CAR T cells that constitutively secrete either single-chain interleukin-12 (scIL-12) or single-chain interleukin-23 (scIL-23). Both cytokine-armored CAR T cell formats improved effector function in vitro, increasing interferon-gamma production and cytotoxicity compared with their unarmored counterparts. scIL-12- and scIL23-secreting CAR T cells significantly delayed tumor growth and prolonged survival in mouse xenograft models of human breast and prostate cancer, while scIL-23 secretion led to increases in vivo persistence and retention of early differentiation states. These findings nominate scIL-23 armoring as a promising strategy to extend CAR T cell therapy to solid tumors.","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":"19 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145947413","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Restoring immune tolerance by engineered regulatory T cell (Treg) therapy is a promising strategy to treat patients suffering from autoimmune and inflammatory diseases. However, in many of these conditions, relevant disease-driving antigens are unknown. Therefore, suitable target (auto-)antigens for antigen-specific Treg cell therapy are rarely available. We present a novel artificial immune biosensor for Treg cells that circumvents this limitation by targeting the immune costimulatory protein CD40-ligand (CD40L), transiently expressed by activated T cells. The artificial immune receptor (AIR) comprises a CD40-derived extracellular binding domain, an intracellular costimulatory signaling domain and a T cell receptor signaling domain of the CD3-ζ chain. After interaction with its membrane-bound ligand, this synthetic receptor triggers a TCR-like activation program in Treg cells including induction of Treg effector molecules and cell proliferation. In a mouse model of graft-versus-host disease (GvHD), transfer of CD40-AIR Treg cells significantly improved survival and demonstrated immune control of the alloantigen-reactive T cell compartment. Expression and signaling of the corresponding human CD40-AIR illustrate the potential for translating this concept. Engineering Treg cells with a CD40L-targeting sensor, that detects activated T cells presents a promising therapeutic approach for a broad range of T cell mediated inflammatory diseases.
{"title":"Engineered CD40-biosensor-expressing Treg cells as a cell therapy approach for inflammatory diseases.","authors":"Sebastian Bittner,Lisa Schmidleithner,Brigitte Ruhland,Veronika Hofmann,Philipp Stüve,Frauke Hoffmann,Bernd Echtenacher,Petra Hoffmann,Matthias Edinger,Andreas Beilhack,Nicholas Strieder,Inmaculada Hernandez-Lopez,Michael Rehli,Thomas Hehlgans,Markus Feuerer","doi":"10.1016/j.ymthe.2026.01.002","DOIUrl":"https://doi.org/10.1016/j.ymthe.2026.01.002","url":null,"abstract":"Restoring immune tolerance by engineered regulatory T cell (Treg) therapy is a promising strategy to treat patients suffering from autoimmune and inflammatory diseases. However, in many of these conditions, relevant disease-driving antigens are unknown. Therefore, suitable target (auto-)antigens for antigen-specific Treg cell therapy are rarely available. We present a novel artificial immune biosensor for Treg cells that circumvents this limitation by targeting the immune costimulatory protein CD40-ligand (CD40L), transiently expressed by activated T cells. The artificial immune receptor (AIR) comprises a CD40-derived extracellular binding domain, an intracellular costimulatory signaling domain and a T cell receptor signaling domain of the CD3-ζ chain. After interaction with its membrane-bound ligand, this synthetic receptor triggers a TCR-like activation program in Treg cells including induction of Treg effector molecules and cell proliferation. In a mouse model of graft-versus-host disease (GvHD), transfer of CD40-AIR Treg cells significantly improved survival and demonstrated immune control of the alloantigen-reactive T cell compartment. Expression and signaling of the corresponding human CD40-AIR illustrate the potential for translating this concept. Engineering Treg cells with a CD40L-targeting sensor, that detects activated T cells presents a promising therapeutic approach for a broad range of T cell mediated inflammatory diseases.","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":"19 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145949675","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Innovative chimeric antigen receptor (CAR) T-cell designs and combinational approaches are needed for enhancing therapeutic effectiveness in solid tumors. We developed and assessed a novel dual-targeting CAR-T therapy that combines an αPDL1.CD28 chimeric receptor with a second-generation αCD133 CAR to target CD133+ tumors. The αPDL1.CD28 structure activated the CD3ζ signaling in cis by clustering with αCD133 CAR via CD28 dimerization. Binding to PD-L1 through αPD-L1 CAR improved the CD133-targeted cytotoxic function of T cells by enhancing activation signals and countering inhibitory signals. Combination with PD-1 blockade further disrupted the PD-L1/PD-1 inhibitory signal, achieving prolonged therapeutic efficacy. Moreover, radiation pre-conditioning (10 Gy/1 fraction or 4 Gy/2 f) maximized the antitumor effects of CAR-T plus PD-1 blockade, inducing complete tumor regression in mice. Radiation induced a unique tissue-resident memory CAR-T cell phenotype with high CXCR6 and CD103 expression. As the ligand of CD103, E-cadherin expression increased in tumor cells after irradiation, potentially mediating E-cadherin-CD103 interactions between tumor cells and tissue-resident memory T cells. Our study introduces a novel dual-targeting CD133/PD-L1 CAR-T cell, and further demonstrates the efficacy and rationale of the triple combination approach in solid tumors.
{"title":"Dual-Targeting CD133/PD-L1 CAR-T plus αPD-1 Overcomes Immunosuppressive Microenvironment and Enhanced by Radiation Pre-conditioning through Induction of a Tissue-resident Memory Phenotype.","authors":"Zhuoran Yao,Kai Kang,Pei-Heng Li,Limei Yin,Ruizhan Tong,Linglu Yi,Yonghong Song,Ren Luo,Yijun Wu,Shanghai Liu,Zichong Peng,Xianming Mo,Wenbo Wang,Jianxin Xue,You Lu","doi":"10.1016/j.ymthe.2025.12.068","DOIUrl":"https://doi.org/10.1016/j.ymthe.2025.12.068","url":null,"abstract":"Innovative chimeric antigen receptor (CAR) T-cell designs and combinational approaches are needed for enhancing therapeutic effectiveness in solid tumors. We developed and assessed a novel dual-targeting CAR-T therapy that combines an αPDL1.CD28 chimeric receptor with a second-generation αCD133 CAR to target CD133+ tumors. The αPDL1.CD28 structure activated the CD3ζ signaling in cis by clustering with αCD133 CAR via CD28 dimerization. Binding to PD-L1 through αPD-L1 CAR improved the CD133-targeted cytotoxic function of T cells by enhancing activation signals and countering inhibitory signals. Combination with PD-1 blockade further disrupted the PD-L1/PD-1 inhibitory signal, achieving prolonged therapeutic efficacy. Moreover, radiation pre-conditioning (10 Gy/1 fraction or 4 Gy/2 f) maximized the antitumor effects of CAR-T plus PD-1 blockade, inducing complete tumor regression in mice. Radiation induced a unique tissue-resident memory CAR-T cell phenotype with high CXCR6 and CD103 expression. As the ligand of CD103, E-cadherin expression increased in tumor cells after irradiation, potentially mediating E-cadherin-CD103 interactions between tumor cells and tissue-resident memory T cells. Our study introduces a novel dual-targeting CD133/PD-L1 CAR-T cell, and further demonstrates the efficacy and rationale of the triple combination approach in solid tumors.","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":"65 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145949674","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-10DOI: 10.1016/j.ymthe.2026.01.006
Pablo Tebas,Julie K Jadlowsky,Yuqi Zhou,M Betina Pampena,George J Leslie,Nan Liu,Sriram Srivatsa,Colby R Maldini,Jingxin Wang,Chungdhak Jaming Tsang,Ola Mohamed,Josephine Romano,Beth Haggarty,Gabriela Plesa,Avery L Gaymon,Andrea L Brennan,Elizabeth Veloso,Bruce L Levine,Mary E Putt,Irina Kulikovskaya,Vanessa Gonzalez,Joseph A Fraietta,Michael R Betts,James A Hoxie,James L Riley
Coupling the HIV fusion inhibitor C34 with CXCR4 (C34-CXCR4) protected CD4 T cells from all strains of HIV. Escape from C34-CXCR4 expression proved difficult, requiring more than 21 serial passages, mutations at conserved gp41 sites, and a complete loss of Vpu activity. A dose-escalation, Phase I clinical trial was performed in which up to 10 billion C34-CXCR4 expressing T cells were infused into people with HIV (PWH). While these infusions were safe, we observed limited persistence of these C34-CXCR4 T cells, lack of reconstitution of HIV-specific CD8 T cell responses, and no effect on time to viral rebound after an analytical treatment interruption. We hypothesized that inappropriate expression of a signaling competent chemokine receptor resulted in the limited persistence of these engineered T cells so we screened several C34-CXCR4 mutants to identify those that could not respond to CXCL12 and still could mediate potent anti-viral activity. A single mutation (D97N) in CXCR4 fulfilled both criteria. T cells co-expressing Dual HIV-specific CARs and C34-CXCR4 D97N maintained high C34-expression in vivo and controlled HIV replication better than unprotected Dual CAR T cells, suggesting that this C34-CXCR4 D97N construct should be considered for future clinical development.
{"title":"Clinical Trial Results Provide the Rationale to Protect Dual HIV-specific T Cells with a Signaling-Defective HIV Fusion Inhibitor.","authors":"Pablo Tebas,Julie K Jadlowsky,Yuqi Zhou,M Betina Pampena,George J Leslie,Nan Liu,Sriram Srivatsa,Colby R Maldini,Jingxin Wang,Chungdhak Jaming Tsang,Ola Mohamed,Josephine Romano,Beth Haggarty,Gabriela Plesa,Avery L Gaymon,Andrea L Brennan,Elizabeth Veloso,Bruce L Levine,Mary E Putt,Irina Kulikovskaya,Vanessa Gonzalez,Joseph A Fraietta,Michael R Betts,James A Hoxie,James L Riley","doi":"10.1016/j.ymthe.2026.01.006","DOIUrl":"https://doi.org/10.1016/j.ymthe.2026.01.006","url":null,"abstract":"Coupling the HIV fusion inhibitor C34 with CXCR4 (C34-CXCR4) protected CD4 T cells from all strains of HIV. Escape from C34-CXCR4 expression proved difficult, requiring more than 21 serial passages, mutations at conserved gp41 sites, and a complete loss of Vpu activity. A dose-escalation, Phase I clinical trial was performed in which up to 10 billion C34-CXCR4 expressing T cells were infused into people with HIV (PWH). While these infusions were safe, we observed limited persistence of these C34-CXCR4 T cells, lack of reconstitution of HIV-specific CD8 T cell responses, and no effect on time to viral rebound after an analytical treatment interruption. We hypothesized that inappropriate expression of a signaling competent chemokine receptor resulted in the limited persistence of these engineered T cells so we screened several C34-CXCR4 mutants to identify those that could not respond to CXCL12 and still could mediate potent anti-viral activity. A single mutation (D97N) in CXCR4 fulfilled both criteria. T cells co-expressing Dual HIV-specific CARs and C34-CXCR4 D97N maintained high C34-expression in vivo and controlled HIV replication better than unprotected Dual CAR T cells, suggesting that this C34-CXCR4 D97N construct should be considered for future clinical development.","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":"253 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145947415","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-10DOI: 10.1016/j.ymthe.2026.01.010
Yong Won Choi,Jae Ho Choi,Young-Sam Lee,Jinju Jeong,Eunho Kang,So Hyun Park,Young-Kyoung Lee,Soon Sang Park,Hee Young Kang,Young Hwa Kim,Tae Jun Park
Drug-induced liver injury accounts for approximately 10% of acute hepatitis and up to 50% of acute liver failure. Despite its clinical significance, treatment remains largely limited to cessation of the offending agent. SLIT/ROBO signaling, known for roles in organ development, angiogenesis, leukocyte migration, and cancer metastasis, has demonstrated protective effects against various organ damage. In mouse models of liver injury induced by acetaminophen (APAP), thioacetamide, bile duct ligation, and serum from patients with toxic liver disease, Slit2 expression significantly increases, while Slit1 and Slit3 remain unchanged. Liver-specific Slit2 knockdown exacerbates liver injury, whereas recombinant SLIT2 alleviates liver damage by reducing oxidative stress via CYP2E1 downregulation and suppressing inflammation through NF-κB inhibition. Notably, among ROBO receptors, only ROBO4 was induced in hepatocytes after APAP exposure. ROBO4 knockdown eliminates the hepatoprotective effects of SLIT2, highlighting the importance of SLIT2/BOBO4 signaling in toxic liver injury. Furthermore, a novel Slit2-derived peptide-5 (SP5), designed from the ROBO4-binding LRR2 domain, significantly reduces liver damage and inflammation. Notably, both recombinant SLIT2 and SP5 confer hepatoprotection even when administered 24 hours after APAP challenge. These findings suggest that SLIT2/ROBO4-targeted therapies may offer a promising approach for preventing fulminant hepatitis in the context of toxic liver injury.
{"title":"SLIT2 as a Key Regulator and Therapeutic Target in Liver Injury.","authors":"Yong Won Choi,Jae Ho Choi,Young-Sam Lee,Jinju Jeong,Eunho Kang,So Hyun Park,Young-Kyoung Lee,Soon Sang Park,Hee Young Kang,Young Hwa Kim,Tae Jun Park","doi":"10.1016/j.ymthe.2026.01.010","DOIUrl":"https://doi.org/10.1016/j.ymthe.2026.01.010","url":null,"abstract":"Drug-induced liver injury accounts for approximately 10% of acute hepatitis and up to 50% of acute liver failure. Despite its clinical significance, treatment remains largely limited to cessation of the offending agent. SLIT/ROBO signaling, known for roles in organ development, angiogenesis, leukocyte migration, and cancer metastasis, has demonstrated protective effects against various organ damage. In mouse models of liver injury induced by acetaminophen (APAP), thioacetamide, bile duct ligation, and serum from patients with toxic liver disease, Slit2 expression significantly increases, while Slit1 and Slit3 remain unchanged. Liver-specific Slit2 knockdown exacerbates liver injury, whereas recombinant SLIT2 alleviates liver damage by reducing oxidative stress via CYP2E1 downregulation and suppressing inflammation through NF-κB inhibition. Notably, among ROBO receptors, only ROBO4 was induced in hepatocytes after APAP exposure. ROBO4 knockdown eliminates the hepatoprotective effects of SLIT2, highlighting the importance of SLIT2/BOBO4 signaling in toxic liver injury. Furthermore, a novel Slit2-derived peptide-5 (SP5), designed from the ROBO4-binding LRR2 domain, significantly reduces liver damage and inflammation. Notably, both recombinant SLIT2 and SP5 confer hepatoprotection even when administered 24 hours after APAP challenge. These findings suggest that SLIT2/ROBO4-targeted therapies may offer a promising approach for preventing fulminant hepatitis in the context of toxic liver injury.","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":"6 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2026-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145949673","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}