Pub Date : 2025-01-08Epub Date: 2024-11-26DOI: 10.1016/j.ymthe.2024.11.033
Bryce D Holt, Samuel J Elliott, Rebecca Meyer, Daniela Reyes, Karyn O'Neil, Zhanna Druzina, Swapnil Kulkarni, Beth L Thurberg, Steven G Nadler, Bartholomew A Pederson
Pompe disease is caused by acid alpha-glucosidase (GAA) deficiency, resulting in lysosomal glycogen accumulation. This disease is characterized by progressive skeletal muscle weakness, respiratory distress, and in the infantile-onset form, cardiomyopathy. The only approved treatment is enzyme replacement therapy (ERT) with human recombinant GAA. While ERT therapy extends life span, residual symptoms remain, with poor muscle uptake and immunogenicity limiting efficacy. We examined a novel Centyrin protein-short interfering ribonucleic acid (siRNA) conjugate targeting CD71 (transferrin receptor type 1, TfR1) and GYS1, a key enzyme involved in glycogen synthesis. Unlike existing ERTs designed to degrade aberrant glycogen deposits observed in Pompe patients, the CD71 Centyrin:Gys1 siRNA is designed to restore glycogen balance by inhibiting glycogen synthesis. To this end, we administered the CD71 Centyrin:Gys1 siRNA conjugate to the 6neo/6neo Pompe mouse model. Once bound to TfR1, siRNA-conjugated Centyrin is internalized into cells to facilitate gene knockdown. We found that treatment with this conjugate significantly reduced GYS1 protein expression, glycogen synthase enzymatic activity, and glycogen levels in muscle. In addition, impaired treadmill exercise performance of male Pompe mice was improved. These data suggest that Centyrin-mediated delivery of Gys1 siRNA may be an effective next generation therapy for late-onset Pompe disease or, in combination with ERT, for infantile-onset Pompe disease.
{"title":"A novel CD71 Centyrin:Gys1 siRNA conjugate reduces glycogen synthesis and glycogen levels in a mouse model of Pompe disease.","authors":"Bryce D Holt, Samuel J Elliott, Rebecca Meyer, Daniela Reyes, Karyn O'Neil, Zhanna Druzina, Swapnil Kulkarni, Beth L Thurberg, Steven G Nadler, Bartholomew A Pederson","doi":"10.1016/j.ymthe.2024.11.033","DOIUrl":"10.1016/j.ymthe.2024.11.033","url":null,"abstract":"<p><p>Pompe disease is caused by acid alpha-glucosidase (GAA) deficiency, resulting in lysosomal glycogen accumulation. This disease is characterized by progressive skeletal muscle weakness, respiratory distress, and in the infantile-onset form, cardiomyopathy. The only approved treatment is enzyme replacement therapy (ERT) with human recombinant GAA. While ERT therapy extends life span, residual symptoms remain, with poor muscle uptake and immunogenicity limiting efficacy. We examined a novel Centyrin protein-short interfering ribonucleic acid (siRNA) conjugate targeting CD71 (transferrin receptor type 1, TfR1) and GYS1, a key enzyme involved in glycogen synthesis. Unlike existing ERTs designed to degrade aberrant glycogen deposits observed in Pompe patients, the CD71 Centyrin:Gys1 siRNA is designed to restore glycogen balance by inhibiting glycogen synthesis. To this end, we administered the CD71 Centyrin:Gys1 siRNA conjugate to the 6<sup>neo</sup>/6<sup>neo</sup> Pompe mouse model. Once bound to TfR1, siRNA-conjugated Centyrin is internalized into cells to facilitate gene knockdown. We found that treatment with this conjugate significantly reduced GYS1 protein expression, glycogen synthase enzymatic activity, and glycogen levels in muscle. In addition, impaired treadmill exercise performance of male Pompe mice was improved. These data suggest that Centyrin-mediated delivery of Gys1 siRNA may be an effective next generation therapy for late-onset Pompe disease or, in combination with ERT, for infantile-onset Pompe disease.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":"235-248"},"PeriodicalIF":12.1,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142739990","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 : 2025-01-08Epub Date: 2024-11-27DOI: 10.1016/j.ymthe.2024.11.031
Yanhong Jiang, Shuanghong Chen, Shenlin Hsiao, Haokun Zhang, Da Xie, Zi Jun Wang, Wendan Ren, Mingyao Liu, Jiaoyang Liao, Yuxuan Wu
{"title":"Efficient and safe in vivo treatment of primary hyperoxaluria type 1 via LNP-CRISPR-Cas9-mediated glycolate oxidase disruption.","authors":"Yanhong Jiang, Shuanghong Chen, Shenlin Hsiao, Haokun Zhang, Da Xie, Zi Jun Wang, Wendan Ren, Mingyao Liu, Jiaoyang Liao, Yuxuan Wu","doi":"10.1016/j.ymthe.2024.11.031","DOIUrl":"10.1016/j.ymthe.2024.11.031","url":null,"abstract":"","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":"417-418"},"PeriodicalIF":12.1,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142751343","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 : 2025-01-08Epub Date: 2024-11-07DOI: 10.1016/j.ymthe.2024.11.011
Jiaxuan Li, Naihui Mao, Ying Wang, Shuli Deng, Keda Chen
Acute upper respiratory tract infections are a major public health issue, with uncontrolled inflammation triggered by upper respiratory viruses being a significant cause of patient deterioration or death. This study focuses on the Janus kinase-signal transducer and activator of transcription Rho-associated coiled-coil containing protein kinase (JAK-STAT-ROCK) signaling pathway, providing an in-depth analysis of the interplay between uncontrolled inflammation after upper respiratory tract infections and the development of neurodegenerative diseases. It offers a conceptual framework for understanding the lung-brain-related immune responses and potential interactions. The relationship between the ROCK-JAK-STAT signaling pathway and inflammatory immunity is a complex and multi-layered research area and exploring potential common targets could open new avenues for the prevention and treatment of related inflammation.
{"title":"Novel insights into the ROCK-JAK-STAT signaling pathway in upper respiratory tract infections and neurodegenerative diseases.","authors":"Jiaxuan Li, Naihui Mao, Ying Wang, Shuli Deng, Keda Chen","doi":"10.1016/j.ymthe.2024.11.011","DOIUrl":"10.1016/j.ymthe.2024.11.011","url":null,"abstract":"<p><p>Acute upper respiratory tract infections are a major public health issue, with uncontrolled inflammation triggered by upper respiratory viruses being a significant cause of patient deterioration or death. This study focuses on the Janus kinase-signal transducer and activator of transcription Rho-associated coiled-coil containing protein kinase (JAK-STAT-ROCK) signaling pathway, providing an in-depth analysis of the interplay between uncontrolled inflammation after upper respiratory tract infections and the development of neurodegenerative diseases. It offers a conceptual framework for understanding the lung-brain-related immune responses and potential interactions. The relationship between the ROCK-JAK-STAT signaling pathway and inflammatory immunity is a complex and multi-layered research area and exploring potential common targets could open new avenues for the prevention and treatment of related inflammation.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":"32-50"},"PeriodicalIF":12.1,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142604754","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 : 2025-01-08Epub Date: 2024-11-29DOI: 10.1016/j.ymthe.2024.11.026
Radoslaw Kaczmarek, Glenn F Pierce
{"title":"Second gene therapy for hemophilia B approved: More answers or questions?","authors":"Radoslaw Kaczmarek, Glenn F Pierce","doi":"10.1016/j.ymthe.2024.11.026","DOIUrl":"10.1016/j.ymthe.2024.11.026","url":null,"abstract":"","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":"1-2"},"PeriodicalIF":12.1,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142770527","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}
Intratumoral administration is a widely used method for oncolytic virotherapy, as it enables immediate access of virus particles to the target tumor and potentially lead to the suppression of untreated distant tumors via in situ vaccination. However, because the injection volume and concentration of the virus solution are physically limited, the dose level cannot be increased. Additionally, efficacy in distant tumors needs improvement to prolong patient survival. Here, we demonstrate the benefit of oxaliplatin, with detailed mechanisms revealed through transcriptome analysis, which may provide a solution for the crucial deficiencies of oncolytic virotherapy. In virus-injected tumors, oxaliplatin improved virus retention through suppression of type I interferons. In distant virus-naive tumors, oxaliplatin induced alterations in the intratumoral macrophage characteristics, leading to the chemotaxis and recruitment of activated T cells and subsequently inducing an inflammatory state in the non-injected tumors. Our findings can be a trigger to change the therapeutic paradigm of oncolytic virotherapy for patients with systemic metastases.
{"title":"Combination with oxaliplatin improves abscopal effect of oncolytic virotherapy through reorganization of intratumoral macrophages.","authors":"Kyoko Tomita, Midori Yamashita, Kentaro Ikegami, Yoshiko Shimizu, Nobuaki Amino, Shinsuke Nakao","doi":"10.1016/j.ymthe.2024.12.007","DOIUrl":"10.1016/j.ymthe.2024.12.007","url":null,"abstract":"<p><p>Intratumoral administration is a widely used method for oncolytic virotherapy, as it enables immediate access of virus particles to the target tumor and potentially lead to the suppression of untreated distant tumors via in situ vaccination. However, because the injection volume and concentration of the virus solution are physically limited, the dose level cannot be increased. Additionally, efficacy in distant tumors needs improvement to prolong patient survival. Here, we demonstrate the benefit of oxaliplatin, with detailed mechanisms revealed through transcriptome analysis, which may provide a solution for the crucial deficiencies of oncolytic virotherapy. In virus-injected tumors, oxaliplatin improved virus retention through suppression of type I interferons. In distant virus-naive tumors, oxaliplatin induced alterations in the intratumoral macrophage characteristics, leading to the chemotaxis and recruitment of activated T cells and subsequently inducing an inflammatory state in the non-injected tumors. Our findings can be a trigger to change the therapeutic paradigm of oncolytic virotherapy for patients with systemic metastases.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":"401-414"},"PeriodicalIF":12.1,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142813514","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 : 2025-01-08Epub Date: 2024-11-19DOI: 10.1016/j.ymthe.2024.11.028
Mireia Bachiller, Nina Barceló-Genestar, Alba Rodriguez-Garcia, Leticia Alserawan, Cèlia Dobaño-López, Marta Giménez-Alejandre, Joan Castellsagué, Salut Colell, Marc Otero-Mateo, Asier Antoñana-Vildosola, Marta Español-Rego, Noelia Ferruz, Mariona Pascal, Beatriz Martín-Antonio, Xavier M Anguela, Cristina Fillat, Eulàlia Olesti, Gonzalo Calvo, Manel Juan, Julio Delgado, Patricia Pérez-Galán, Álvaro Urbano-Ispizua, Sonia Guedan
CD19 CAR-T therapy has achieved remarkable responses in relapsed/refractory non-Hodgkin lymphoma (NHL). However, challenges persist, with refractory responses or relapses after CAR-T administration linked to CD19 loss or downregulation. Given the co-expression of CD19 and BCMA in NHL, we hypothesized that dual targeting could enhance long-term efficacy. We optimized different dual-targeting approaches, including co-transduction of two lentiviral vectors, bicistronic, tandem, and loop and pool strategies, based on our academic anti-CD19 (ARI0001) and anti-BCMA (ARI0002h) CAR-T cells. Comparison with anti-CD19/CD20 or anti-CD19/CD22 dual targeting was also performed. We demonstrate that anti-CD19/BCMA CAR-T cells can be effectively generated through the co-transduction of two lentiviral vectors after optimization to minimize competition for cellular resources. Co-transduced T cells, called ARI0003, effectively targeted NHL tumor cells with high avidity, outperforming anti-CD19 CAR-T cells and other dual-targeting approaches both in vitro and in vivo, particularly in low CD19 antigen density models. ARI0003 maintained effectiveness post-CD19 CAR-T treatment in xenograft models and in spheroids from relapsed CART-treated patients. ARI0003 CAR-T cells were effectively manufactured under Good Manufacturing Practice conditions, with a reduced risk of genotoxicity compared to other dual-targeting approaches. A first-in-human phase 1 clinical trial (CARTD-BG-01; this study was registered at ClinicalTrials.gov [NCT06097455]) has been initiated to evaluate the safety and efficacy of ARI0003 in NHL.
{"title":"ARI0003: Co-transduced CD19/BCMA dual-targeting CAR-T cells for the treatment of non-Hodgkin lymphoma.","authors":"Mireia Bachiller, Nina Barceló-Genestar, Alba Rodriguez-Garcia, Leticia Alserawan, Cèlia Dobaño-López, Marta Giménez-Alejandre, Joan Castellsagué, Salut Colell, Marc Otero-Mateo, Asier Antoñana-Vildosola, Marta Español-Rego, Noelia Ferruz, Mariona Pascal, Beatriz Martín-Antonio, Xavier M Anguela, Cristina Fillat, Eulàlia Olesti, Gonzalo Calvo, Manel Juan, Julio Delgado, Patricia Pérez-Galán, Álvaro Urbano-Ispizua, Sonia Guedan","doi":"10.1016/j.ymthe.2024.11.028","DOIUrl":"10.1016/j.ymthe.2024.11.028","url":null,"abstract":"<p><p>CD19 CAR-T therapy has achieved remarkable responses in relapsed/refractory non-Hodgkin lymphoma (NHL). However, challenges persist, with refractory responses or relapses after CAR-T administration linked to CD19 loss or downregulation. Given the co-expression of CD19 and BCMA in NHL, we hypothesized that dual targeting could enhance long-term efficacy. We optimized different dual-targeting approaches, including co-transduction of two lentiviral vectors, bicistronic, tandem, and loop and pool strategies, based on our academic anti-CD19 (ARI0001) and anti-BCMA (ARI0002h) CAR-T cells. Comparison with anti-CD19/CD20 or anti-CD19/CD22 dual targeting was also performed. We demonstrate that anti-CD19/BCMA CAR-T cells can be effectively generated through the co-transduction of two lentiviral vectors after optimization to minimize competition for cellular resources. Co-transduced T cells, called ARI0003, effectively targeted NHL tumor cells with high avidity, outperforming anti-CD19 CAR-T cells and other dual-targeting approaches both in vitro and in vivo, particularly in low CD19 antigen density models. ARI0003 maintained effectiveness post-CD19 CAR-T treatment in xenograft models and in spheroids from relapsed CART-treated patients. ARI0003 CAR-T cells were effectively manufactured under Good Manufacturing Practice conditions, with a reduced risk of genotoxicity compared to other dual-targeting approaches. A first-in-human phase 1 clinical trial (CARTD-BG-01; this study was registered at ClinicalTrials.gov [NCT06097455]) has been initiated to evaluate the safety and efficacy of ARI0003 in NHL.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":"317-335"},"PeriodicalIF":12.1,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142676016","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 : 2025-01-08Epub Date: 2024-11-19DOI: 10.1016/j.ymthe.2024.11.030
Michaela Helble, Jacqueline Chu, Kaitlyn Flowers, Abigail R Trachtman, Alana Huynh, Amber Kim, Nicholas Shupin, Casey E Hojecki, Ebony N Gary, Shahlo Solieva, Elizabeth M Parzych, David B Weiner, Daniel W Kulp, Ami Patel
Monoclonal antibodies are an important class of biologics with over 160 Food and Drug Administration/European Union-approved drugs. A significant bottleneck to global accessibility of recombinant monoclonal antibodies stems from complexities related to their production, storage, and distribution. Recently, gene-encoded approaches such as mRNA, DNA, or viral delivery have gained popularity, but ensuring biologically relevant levels of antibody expression in the host remains a critical issue. Using a synthetic DNA platform, we investigated the role of antibody structure and sequence toward in vivo expression. SARS-CoV-2 antibody 2196 was recently engineered as a DNA-encoded monoclonal antibody (DMAb-2196). Utilizing an immunoglobulin heavy and light chain "chain-swap" methodology, we interrogated features of DMAb-2196 that can modulate in vivo expression through rational design and structural modeling. Comparing these results to natural variation of antibody sequences resulted in development of an antibody frequency score that aids in the prediction of expression-improving mutations by leveraging antibody repertoire datasets. We demonstrate that a single amino acid mutation identified through this score increases in vivo expression up to 2-fold and that combinations of mutations can also enhance expression. This analysis has led to a generalized pipeline that can unlock the potential for in vivo delivery of therapeutic antibodies across many indications.
单克隆抗体是一类重要的生物制剂,美国食品和药物管理局(FDA)/欧盟共批准了 160 多种单克隆抗体药物。全球获得重组单克隆抗体的一个重要瓶颈是其生产、储存和销售的复杂性。最近,基因编码的方法(如 mRNA、DNA 或病毒递送)越来越受欢迎,但确保抗体在宿主体内的生物相关表达水平仍然是一个关键问题。利用合成 DNA 平台,我们研究了抗体结构和序列对体内表达的作用。SARS-COV2 抗体 2196 最近被设计成一种 DNA 编码的单克隆抗体(DMAb-2196)。利用免疫球蛋白重链和轻链 "换链 "的方法,我们通过合理的设计和结构建模研究了 DMAb-2196 可调节体内表达的特征。将这些结果与抗体序列的自然变异进行比较后,我们开发出了一种抗体频率评分,通过利用抗体库数据集来帮助预测可改善表达的突变。我们证明,通过该评分确定的单个氨基酸突变可将体内表达量提高 2 倍,突变组合也可提高表达量。通过这项分析,我们开发出了一种通用的管道,可以释放体内输送治疗性抗体的潜力,适用于多种适应症。
{"title":"Structure and sequence engineering approaches to improve in vivo expression of nucleic acid-delivered antibodies.","authors":"Michaela Helble, Jacqueline Chu, Kaitlyn Flowers, Abigail R Trachtman, Alana Huynh, Amber Kim, Nicholas Shupin, Casey E Hojecki, Ebony N Gary, Shahlo Solieva, Elizabeth M Parzych, David B Weiner, Daniel W Kulp, Ami Patel","doi":"10.1016/j.ymthe.2024.11.030","DOIUrl":"10.1016/j.ymthe.2024.11.030","url":null,"abstract":"<p><p>Monoclonal antibodies are an important class of biologics with over 160 Food and Drug Administration/European Union-approved drugs. A significant bottleneck to global accessibility of recombinant monoclonal antibodies stems from complexities related to their production, storage, and distribution. Recently, gene-encoded approaches such as mRNA, DNA, or viral delivery have gained popularity, but ensuring biologically relevant levels of antibody expression in the host remains a critical issue. Using a synthetic DNA platform, we investigated the role of antibody structure and sequence toward in vivo expression. SARS-CoV-2 antibody 2196 was recently engineered as a DNA-encoded monoclonal antibody (DMAb-2196). Utilizing an immunoglobulin heavy and light chain \"chain-swap\" methodology, we interrogated features of DMAb-2196 that can modulate in vivo expression through rational design and structural modeling. Comparing these results to natural variation of antibody sequences resulted in development of an antibody frequency score that aids in the prediction of expression-improving mutations by leveraging antibody repertoire datasets. We demonstrate that a single amino acid mutation identified through this score increases in vivo expression up to 2-fold and that combinations of mutations can also enhance expression. This analysis has led to a generalized pipeline that can unlock the potential for in vivo delivery of therapeutic antibodies across many indications.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":"152-167"},"PeriodicalIF":12.1,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142676189","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 : 2025-01-08Epub Date: 2024-11-07DOI: 10.1016/j.ymthe.2024.11.010
Xuejia Zhai, Ling Mao, Qingmei Kang, Jie Liu, Yu Zhou, Jun Wang, Xianyan Yang, Di Wang, Junhan Wang, Yao Li, Jiangjie Duan, Tao Zhang, Shuang Lin, Tingting Zhao, Jianjun Li, Min Wu, Shicang Yu
The incidence of brain metastasis (BM) is gradually increasing, and the prognosis and therapeutic effect are poor. The emergence of immunotherapy has brought hope for the development of BM treatments. This study revealed that compared with primary cancers, BMs have a colder and more acidic tumor microenvironment (TME), resulting in reduced protein levels of mesothelin (MSLN), a promising target for chimeric antigen receptor-T (CAR-T) cell therapy for triple-negative breast cancer (TNBC) with BMs. These factors could significantly decrease the efficiency of MSLN-CAR-T cells in TNBC BMs. Pantoprazole (PPZ) administration at the most commonly used dose in the clinic notably increased the pH of the TME, inhibited lysosomal activity, increased the membrane levels of the MSLN protein and improved the killing ability of MSLN-CAR-T cells both in vitro and in vivo. Similar results were obtained in non-small cell lung cancer BMs. Hence, when administered in combination with CAR-T cells, PPZ, which increases the protein levels of target antigens, may constitute a new immunotherapeutic strategy for treating solid tumors with BMs.
{"title":"Proton pump inhibitor attenuates acidic microenvironment to improve the therapeutic effects of MSLN-CAR-T cells on the brain metastasis.","authors":"Xuejia Zhai, Ling Mao, Qingmei Kang, Jie Liu, Yu Zhou, Jun Wang, Xianyan Yang, Di Wang, Junhan Wang, Yao Li, Jiangjie Duan, Tao Zhang, Shuang Lin, Tingting Zhao, Jianjun Li, Min Wu, Shicang Yu","doi":"10.1016/j.ymthe.2024.11.010","DOIUrl":"10.1016/j.ymthe.2024.11.010","url":null,"abstract":"<p><p>The incidence of brain metastasis (BM) is gradually increasing, and the prognosis and therapeutic effect are poor. The emergence of immunotherapy has brought hope for the development of BM treatments. This study revealed that compared with primary cancers, BMs have a colder and more acidic tumor microenvironment (TME), resulting in reduced protein levels of mesothelin (MSLN), a promising target for chimeric antigen receptor-T (CAR-T) cell therapy for triple-negative breast cancer (TNBC) with BMs. These factors could significantly decrease the efficiency of MSLN-CAR-T cells in TNBC BMs. Pantoprazole (PPZ) administration at the most commonly used dose in the clinic notably increased the pH of the TME, inhibited lysosomal activity, increased the membrane levels of the MSLN protein and improved the killing ability of MSLN-CAR-T cells both in vitro and in vivo. Similar results were obtained in non-small cell lung cancer BMs. Hence, when administered in combination with CAR-T cells, PPZ, which increases the protein levels of target antigens, may constitute a new immunotherapeutic strategy for treating solid tumors with BMs.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":"336-355"},"PeriodicalIF":12.1,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142604921","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 : 2025-01-08Epub Date: 2024-12-20DOI: 10.1016/j.ymthe.2024.12.026
Juan Cai, Zheng Dong
{"title":"C-reactive protein in diabetic kidney disease: A new therapeutic avenue?","authors":"Juan Cai, Zheng Dong","doi":"10.1016/j.ymthe.2024.12.026","DOIUrl":"10.1016/j.ymthe.2024.12.026","url":null,"abstract":"","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":" ","pages":"26-27"},"PeriodicalIF":12.1,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142872691","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}