首页 > 最新文献

Molecular Cancer Therapeutics最新文献

英文 中文
In vivo Auto-tuning of Antibody-Drug Conjugate Delivery to Maximize Efficacy using High-Avidity, Low-Affinity Antibodies. 体内自动调整抗体-药物结合递送,以最大限度地利用高亲和力,低亲和力抗体的疗效。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-30 DOI: 10.1158/1535-7163.MCT-25-0576
Anna Kopp, Shujun Dong, Hyeyoung Kwon, Tiexin Wang, Alec A Desai, Andrew Philippart, Dong Jun Koo, Jennifer J Linderman, Peter M Tessier, Greg M Thurber

Antibody-drug conjugates (ADCs) have experienced a surge in clinical approvals in the past few years. Despite this success, a major limitation to ADC efficacy in solid tumors is poor tumor penetration, which leaves many cancer cells untargeted. Co-administration of unconjugated antibody can improve tumor penetration and increase efficacy when target receptor expression is high. However, it can also reduce efficacy in low-expression tumors where ADC delivery is limited by insufficient cellular uptake. This creates an intrinsic problem because many patients express different levels of target between and within tumors. Here, we show how unconjugated High-Avidity, Low-Affinity (HALA) antibodies can automatically tune the cellular ADC delivery to match the local expression level. Using HER2 ADCs as a model, the tumor distribution of trastuzumab emtansine and trastuzumab deruxtecan co-administered with the HALA antibody was improved in vivo, translating to equal or greater ADC efficacy across a range of HER2 expression levels. Furthermore, Fc-enhanced HALA antibodies elicited a strong response in an immunocompetent mouse model. These results demonstrate that HALA antibodies can expand treatment ranges beyond high-expression targets and leverage strong immune responses.

抗体-药物偶联物(adc)在过去几年中经历了临床批准的激增。尽管取得了这一成功,但ADC在实体肿瘤中的疗效的一个主要限制是肿瘤穿透性差,这使得许多癌细胞无法靶向。当靶受体表达高时,非偶联抗体联合给药可促进肿瘤穿透,提高疗效。然而,在ADC递送因细胞摄取不足而受到限制的低表达肿瘤中,它也会降低疗效。这就产生了一个固有的问题,因为许多患者在肿瘤之间和肿瘤内部表达不同水平的靶蛋白。在这里,我们展示了未偶联的高亲和度,低亲和力(HALA)抗体如何自动调整细胞ADC递送以匹配局部表达水平。以HER2 ADC为模型,曲妥珠单抗emtansine和曲妥珠单抗deruxtecan与HALA抗体共同给药的肿瘤分布在体内得到改善,在HER2表达水平范围内转化为相同或更高的ADC疗效。此外,fc增强的HALA抗体在免疫功能小鼠模型中引起强烈反应。这些结果表明,HALA抗体可以扩大治疗范围,超越高表达靶点,并利用强大的免疫反应。
{"title":"In vivo Auto-tuning of Antibody-Drug Conjugate Delivery to Maximize Efficacy using High-Avidity, Low-Affinity Antibodies.","authors":"Anna Kopp, Shujun Dong, Hyeyoung Kwon, Tiexin Wang, Alec A Desai, Andrew Philippart, Dong Jun Koo, Jennifer J Linderman, Peter M Tessier, Greg M Thurber","doi":"10.1158/1535-7163.MCT-25-0576","DOIUrl":"10.1158/1535-7163.MCT-25-0576","url":null,"abstract":"<p><p>Antibody-drug conjugates (ADCs) have experienced a surge in clinical approvals in the past few years. Despite this success, a major limitation to ADC efficacy in solid tumors is poor tumor penetration, which leaves many cancer cells untargeted. Co-administration of unconjugated antibody can improve tumor penetration and increase efficacy when target receptor expression is high. However, it can also reduce efficacy in low-expression tumors where ADC delivery is limited by insufficient cellular uptake. This creates an intrinsic problem because many patients express different levels of target between and within tumors. Here, we show how unconjugated High-Avidity, Low-Affinity (HALA) antibodies can automatically tune the cellular ADC delivery to match the local expression level. Using HER2 ADCs as a model, the tumor distribution of trastuzumab emtansine and trastuzumab deruxtecan co-administered with the HALA antibody was improved in vivo, translating to equal or greater ADC efficacy across a range of HER2 expression levels. Furthermore, Fc-enhanced HALA antibodies elicited a strong response in an immunocompetent mouse model. These results demonstrate that HALA antibodies can expand treatment ranges beyond high-expression targets and leverage strong immune responses.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145864135","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting Tumor Microenvironment-Derived NRG1-HER2/3 Signaling with Zenocutuzumab Restores Sensitivity to AR Inhibition in PTEN Wild-type Prostate Cancer. Zenocutuzumab靶向肿瘤微环境来源的NRG1-HER2/3信号通路可恢复PTEN野生型前列腺癌对AR抑制的敏感性
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-30 DOI: 10.1158/1535-7163.MCT-25-0505
Brian M Shinder, Young Sun Lee, Ninghui Mao, Nazifa Salsabeel, Zeda Zhang, Harmanpreet Kaur, Xiaoping Chen, Qing Chang, Elisa de Stanchina, Anuradha Gopalan, Charles L Sawyers, Brett S Carver

Investigating the mechanisms of acquired resistance to antiandrogens remains a critical clinical need as patients with prostate cancer inevitably develop resistance to androgen receptor (AR)-targeted therapies. Previously, we demonstrated that neuregulin 1 (NRG1) derived from cancer-associated fibroblasts (CAF) promotes antiandrogen resistance through human epidermal growth factor receptor 3 (HER3)-AKT signaling. In this study, we sought to further dissect the molecular context in which NRG1-induced PI3K signaling activation plays a dominant role in driving resistance and evaluate whether targeting HER2/3 dimerization can influence sensitivity to AR inhibition. IHC analysis of radical prostatectomy specimens from patients with prostate cancer treated with or without neoadjuvant hormonal therapy shows that NRG1 was significantly upregulated following AR inhibition, independent of PTEN status. However, we found that stimulation with recombinant NRG1 or CAF-conditioned media induced resistance to AR inhibition only in PTEN wild-type prostate cancer cells and not in PTEN-deficient cells. Selective inhibition of NRG1 using the clinical-grade bispecific humanized immunoglobulin G1, zenocutuzumab (Zeno, MCLA-128), restored sensitivity to AR-targeted therapies in PTEN wild-type tumors, demonstrating its efficacy as a potential therapeutic agent to block the effects of NRG1. In the context of PTEN loss and AR inhibitor resistance, Zeno did not restore sensitivity. These findings highlight the critical molecular context in which tumor microenvironment-derived NRG1 affects responsiveness to AR inhibition and suggest that targeting NRG1 is a promising strategy for overcoming resistance to androgen blockade in PTEN wild-type prostate cancers.

随着前列腺癌患者不可避免地对雄激素受体(AR)靶向治疗产生耐药性,研究抗雄激素获得性耐药的机制仍然是一个关键的临床需要。之前,我们证明了来自癌症相关成纤维细胞(CAF)的神经调节蛋白1 (NRG1)通过人表皮生长因子受体3 (HER3)-AKT信号通路促进抗雄激素抗性。在这项研究中,我们试图进一步分析nrg1诱导的PI3K信号激活在驱动耐药中起主导作用的分子背景,并评估靶向HER2/3二聚化是否会影响对AR抑制的敏感性。对接受或未接受新辅助激素治疗的前列腺癌根治性前列腺切除术标本的免疫组化分析显示,NRG1在AR抑制后显著上调,与PTEN状态无关。然而,我们发现重组NRG1或cafo条件培养基刺激仅在PTEN野生型前列腺癌细胞中诱导对AR抑制的抗性,而在PTEN缺陷细胞中则没有。使用临床级双特异性人源化免疫球蛋白G1选择性抑制NRG1, zenocutuzumab (Zeno, MCLA-128)恢复了PTEN野生型肿瘤对ar靶向治疗的敏感性,证明了其作为阻断NRG1作用的潜在治疗药物的有效性。在PTEN缺失和AR抑制剂耐药的情况下,Zeno没有恢复敏感性。这些发现强调了肿瘤微环境源性NRG1影响AR抑制反应的关键分子背景,并表明靶向NRG1是克服PTEN野生型前列腺癌雄激素阻断抗性的一种有希望的策略。
{"title":"Targeting Tumor Microenvironment-Derived NRG1-HER2/3 Signaling with Zenocutuzumab Restores Sensitivity to AR Inhibition in PTEN Wild-type Prostate Cancer.","authors":"Brian M Shinder, Young Sun Lee, Ninghui Mao, Nazifa Salsabeel, Zeda Zhang, Harmanpreet Kaur, Xiaoping Chen, Qing Chang, Elisa de Stanchina, Anuradha Gopalan, Charles L Sawyers, Brett S Carver","doi":"10.1158/1535-7163.MCT-25-0505","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0505","url":null,"abstract":"<p><p>Investigating the mechanisms of acquired resistance to antiandrogens remains a critical clinical need as patients with prostate cancer inevitably develop resistance to androgen receptor (AR)-targeted therapies. Previously, we demonstrated that neuregulin 1 (NRG1) derived from cancer-associated fibroblasts (CAF) promotes antiandrogen resistance through human epidermal growth factor receptor 3 (HER3)-AKT signaling. In this study, we sought to further dissect the molecular context in which NRG1-induced PI3K signaling activation plays a dominant role in driving resistance and evaluate whether targeting HER2/3 dimerization can influence sensitivity to AR inhibition. IHC analysis of radical prostatectomy specimens from patients with prostate cancer treated with or without neoadjuvant hormonal therapy shows that NRG1 was significantly upregulated following AR inhibition, independent of PTEN status. However, we found that stimulation with recombinant NRG1 or CAF-conditioned media induced resistance to AR inhibition only in PTEN wild-type prostate cancer cells and not in PTEN-deficient cells. Selective inhibition of NRG1 using the clinical-grade bispecific humanized immunoglobulin G1, zenocutuzumab (Zeno, MCLA-128), restored sensitivity to AR-targeted therapies in PTEN wild-type tumors, demonstrating its efficacy as a potential therapeutic agent to block the effects of NRG1. In the context of PTEN loss and AR inhibitor resistance, Zeno did not restore sensitivity. These findings highlight the critical molecular context in which tumor microenvironment-derived NRG1 affects responsiveness to AR inhibition and suggest that targeting NRG1 is a promising strategy for overcoming resistance to androgen blockade in PTEN wild-type prostate cancers.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"OF1-OF10"},"PeriodicalIF":5.5,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145857325","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The proteasome is revealed as a therapeutic target in recurrent glioblastoma xenografts. 蛋白酶体是复发性胶质母细胞瘤异种移植物的治疗靶点。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-24 DOI: 10.1158/1535-7163.MCT-25-0770
Charlotte Degorre, Philip J Tofilon

Radiation remains a primary treatment for glioblastoma, yet tumors frequently recur within two years. In this study, orthotopic xenografts initiated from glioma stem-like cells (GSCs) implanted into the right striatum of nude mice were used to investigate the biology of recurrent GBM. In this model, untreated tumors showed diffuse growth pattern across the right hemisphere (RH) and olfactory bulb (OB), whereas post-irradiation tumors (10 Gy) regrew predominantly within the OB, exhibiting increased cell density and a well-demarcated border indicative of an altered growth pattern. Transcriptomes of untreated and recurrent tumors were assessed using spatial profiling. Comparison of gene expression across regions of interest revealed that recurrent tumors are less heterogeneous and exhibit a distinct transcriptional profile compared to untreated tumors. 463 genes were differentially expressed and GSEA analysis revealed significant enrichment of pathways related to cell cycle regulation in the recurrent as compared to untreated tumors. Further analysis of those pathways revealed a significant upregulation of 22 proteasome-related genes in recurrent tumors. Moreover, functional assays revealed significantly higher proteasome activity in recurrent compared to untreated tumors, suggesting the proteasome as a potential therapeutic target unique to recurrent GBM. To evaluate the therapeutic relevance, mice were treated with the combination of radiation followed by the proteasome inhibitor ixazomib. While ixazomib had no effect on untreated tumors, its administration post-irradiation significantly prolonged survival in two GSC xenograft models. These results illustrate how defining molecular alterations that develop in recurrent GBM xenografts can lead to the identification of a novel therapeutic target.

放疗仍然是胶质母细胞瘤的主要治疗方法,但肿瘤经常在两年内复发。本研究采用胶质瘤干细胞样细胞(GSCs)原位移植裸鼠右纹状体,研究复发性GBM的生物学特性。在该模型中,未经治疗的肿瘤显示右半球(RH)和嗅球(OB)的弥漫性生长模式,而辐照后肿瘤(10 Gy)主要在嗅球内再生,细胞密度增加,边界清晰,表明生长模式发生了改变。使用空间谱法评估未治疗和复发肿瘤的转录组。跨感兴趣区域的基因表达比较显示,与未治疗的肿瘤相比,复发肿瘤的异质性较小,表现出不同的转录谱。463个基因差异表达,GSEA分析显示,与未治疗的肿瘤相比,复发肿瘤中与细胞周期调节相关的途径显著富集。对这些通路的进一步分析显示,22个蛋白酶体相关基因在复发性肿瘤中显著上调。此外,功能分析显示,与未治疗的肿瘤相比,复发性肿瘤的蛋白酶体活性明显更高,这表明蛋白酶体是复发性GBM特有的潜在治疗靶点。为了评估治疗相关性,小鼠接受了放疗和蛋白酶体抑制剂ixazomib的联合治疗。虽然ixazomib对未治疗的肿瘤没有影响,但在两种GSC异种移植模型中,照射后给予ixazomib可显着延长生存期。这些结果说明了如何定义复发性GBM异种移植物中发生的分子改变可以导致鉴定新的治疗靶点。
{"title":"The proteasome is revealed as a therapeutic target in recurrent glioblastoma xenografts.","authors":"Charlotte Degorre, Philip J Tofilon","doi":"10.1158/1535-7163.MCT-25-0770","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0770","url":null,"abstract":"<p><p>Radiation remains a primary treatment for glioblastoma, yet tumors frequently recur within two years. In this study, orthotopic xenografts initiated from glioma stem-like cells (GSCs) implanted into the right striatum of nude mice were used to investigate the biology of recurrent GBM. In this model, untreated tumors showed diffuse growth pattern across the right hemisphere (RH) and olfactory bulb (OB), whereas post-irradiation tumors (10 Gy) regrew predominantly within the OB, exhibiting increased cell density and a well-demarcated border indicative of an altered growth pattern. Transcriptomes of untreated and recurrent tumors were assessed using spatial profiling. Comparison of gene expression across regions of interest revealed that recurrent tumors are less heterogeneous and exhibit a distinct transcriptional profile compared to untreated tumors. 463 genes were differentially expressed and GSEA analysis revealed significant enrichment of pathways related to cell cycle regulation in the recurrent as compared to untreated tumors. Further analysis of those pathways revealed a significant upregulation of 22 proteasome-related genes in recurrent tumors. Moreover, functional assays revealed significantly higher proteasome activity in recurrent compared to untreated tumors, suggesting the proteasome as a potential therapeutic target unique to recurrent GBM. To evaluate the therapeutic relevance, mice were treated with the combination of radiation followed by the proteasome inhibitor ixazomib. While ixazomib had no effect on untreated tumors, its administration post-irradiation significantly prolonged survival in two GSC xenograft models. These results illustrate how defining molecular alterations that develop in recurrent GBM xenografts can lead to the identification of a novel therapeutic target.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145820255","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Immune Reprogramming of Tumor Microenvironments by Cytotoxic Antibody-Drug Conjugate Payloads. 细胞毒性抗体-药物偶联有效载荷对肿瘤微环境的免疫重编程。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-23 DOI: 10.1158/1535-7163.MCT-25-0682
Srishti Chakravorty, Yulia Zybina, Eunseon Ahn, Komal Pradhan, Ni Yu, Marlene Taylor, Manoj Charati, Douglas C Wilson, Albert B Jeon, Wendy M Blumenschein, Jin-Hwan Han

Antibody-drug conjugates (ADC) have recently emerged as an effective treatment option for cancer. Although the fundamental mechanisms of direct tumor cell killing by various ADC payloads have been established, their impact on the tumor microenvironment (TME) remains underexplored. To investigate this fundamental question, we generated an immunocompetent murine tumor model that maintains the expression of a clinically validated tumor-associated antigen, human HER2. We evaluated two ADCs with a shared antibody framework: trastuzumab linked to the microtubule inhibitor monomethyl auristatin E (T-MMAE) and the topoisomerase inhibitor deruxtecan (T-DXd). Treatment with T-MMAE led to a significant increase in immune cell infiltration, whereas T-DXd-treated tumors had fewer immune cells albeit comparable tumor cytotoxicity. When combined with anti-PD-1 immunotherapy, similar additive effects on the primary antitumor response were observed for both ADCs. A key qualitative difference between the two ADCs was observed in the phenotypes of myeloid APCs; T-MMAE treatment resulted in greater immune cell infiltration within the tumor, including macrophages that showed increased gene expression of F4/80, CD206, and IL10RA. In contrast, tumors treated with T-DXd exhibited a lower proportion of macrophages, but APCs in these tumors displayed heightened levels of the CD80 costimulatory molecule. The secondary antitumor response mediated by memory CD8+ T cells was crucial for the formation of immunologic memory induced by both ADCs. Therefore, our findings reveal that, after ADC-mediated tumor cytotoxicity, different ADC payloads elicit distinct immunologic responses characterized by varying levels of myeloid cell activation within the TME.

抗体-药物偶联物(ADC)最近成为一种有效的癌症治疗选择。尽管各种ADC有效载荷直接杀伤肿瘤细胞的基本机制已经建立,但它们对肿瘤微环境(TME)的影响仍未得到充分探讨。为了研究这个基本问题,我们建立了一个免疫能力强的小鼠肿瘤模型,该模型维持了一种临床验证的肿瘤相关抗原——人HER2的表达。我们评估了两种具有共享抗体框架的adc:曲妥珠单抗与微管抑制剂monomethyl auristatin E (T-MMAE)和拓扑异构酶抑制剂deruxtecan (T-DXd)连接。T-MMAE治疗导致免疫细胞浸润显著增加,而t - dxd治疗的肿瘤免疫细胞较少,尽管肿瘤细胞毒性相当。当与抗pd -1免疫治疗联合使用时,观察到两种adc对原发性抗肿瘤反应的相似叠加效应。两种adc在髓系APCs的表型上存在关键的定性差异;T-MMAE治疗导致肿瘤内更大的免疫细胞浸润,包括巨噬细胞,F4/80、CD206和IL10RA基因表达增加。相比之下,用T-DXd治疗的肿瘤显示出较低比例的巨噬细胞,但这些肿瘤中的apc显示出CD80共刺激分子水平升高。记忆性CD8+ T细胞介导的继发性抗肿瘤反应对于两种adc诱导的免疫记忆的形成至关重要。因此,我们的研究结果表明,在ADC介导的肿瘤细胞毒性后,不同的ADC有效载荷引起不同的免疫反应,其特征是TME内不同水平的骨髓细胞活化。
{"title":"Immune Reprogramming of Tumor Microenvironments by Cytotoxic Antibody-Drug Conjugate Payloads.","authors":"Srishti Chakravorty, Yulia Zybina, Eunseon Ahn, Komal Pradhan, Ni Yu, Marlene Taylor, Manoj Charati, Douglas C Wilson, Albert B Jeon, Wendy M Blumenschein, Jin-Hwan Han","doi":"10.1158/1535-7163.MCT-25-0682","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0682","url":null,"abstract":"<p><p>Antibody-drug conjugates (ADC) have recently emerged as an effective treatment option for cancer. Although the fundamental mechanisms of direct tumor cell killing by various ADC payloads have been established, their impact on the tumor microenvironment (TME) remains underexplored. To investigate this fundamental question, we generated an immunocompetent murine tumor model that maintains the expression of a clinically validated tumor-associated antigen, human HER2. We evaluated two ADCs with a shared antibody framework: trastuzumab linked to the microtubule inhibitor monomethyl auristatin E (T-MMAE) and the topoisomerase inhibitor deruxtecan (T-DXd). Treatment with T-MMAE led to a significant increase in immune cell infiltration, whereas T-DXd-treated tumors had fewer immune cells albeit comparable tumor cytotoxicity. When combined with anti-PD-1 immunotherapy, similar additive effects on the primary antitumor response were observed for both ADCs. A key qualitative difference between the two ADCs was observed in the phenotypes of myeloid APCs; T-MMAE treatment resulted in greater immune cell infiltration within the tumor, including macrophages that showed increased gene expression of F4/80, CD206, and IL10RA. In contrast, tumors treated with T-DXd exhibited a lower proportion of macrophages, but APCs in these tumors displayed heightened levels of the CD80 costimulatory molecule. The secondary antitumor response mediated by memory CD8+ T cells was crucial for the formation of immunologic memory induced by both ADCs. Therefore, our findings reveal that, after ADC-mediated tumor cytotoxicity, different ADC payloads elicit distinct immunologic responses characterized by varying levels of myeloid cell activation within the TME.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"OF1-OF16"},"PeriodicalIF":5.5,"publicationDate":"2025-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145810626","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Development of a SIN1 targeting inhibitor as a novel therapeutic approach for the treatment of malignancies. 开发一种靶向SIN1的抑制剂作为治疗恶性肿瘤的新方法。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-22 DOI: 10.1158/1535-7163.MCT-25-0298
Elspeth M Beauchamp, Atul Jain, Matt Clutter, Dominik Nahotko, Emely Lopez Fajardo, Purav P Vagadia, Sara F Dunne, Alain Mina, Mariafausta Fischietti, Chidera V Oku, Gavin T Blyth, Rama K Mishra, Elena Karras, Sara G Radecki, Connor Lantz, Sidharth Addepalli, Mirage Modi, Lihua Zou, Byoung-Kyu Cho, Young Ah Goo, Jenna Rossoff, Eleanor N Fish, Diana Saleiro, Gary E Schiltz, Leonidas C Platanias

The essential role of mTOR in promoting tumorigenesis of many cancers makes it an attractive therapeutic target. However, catalytic mTOR inhibitors, which block both mTORC1 and mTORC2, result in activation of negative feedback loops as a resistance mechanism. Selective mTORC2 inhibitors are expected to have the desired anti-tumor effects without engaging resistance mechanisms; however, to date, no such mTORC2 inhibitors have been developed. Using in-silico screening and medicinal chemistry optimization, we identified several small molecules that bind to the unique mTORC2 component, SIN1. We demonstrate that this SIN1 inhibitor alters post-translational modification, protein-protein interactions, and blocks mTORC2- and rapamycin sensitive (RS) mTORC1-mediated signaling. The SIN1 inhibitor also inhibits wildtype RAS activation and downstream MAPK signaling, as well as cell proliferation of multiple cancer cell line types. SIN1 inhibition can enhance the efficacy of FDA- approved anti-neoplastic agents in vitro and may provide a novel approach for the treatment of different types of malignancies.

mTOR在促进多种癌症发生中的重要作用使其成为一个有吸引力的治疗靶点。然而,催化mTOR抑制剂,阻断mTORC1和mTORC2,导致负反馈回路的激活作为一种抗性机制。选择性mTORC2抑制剂有望在不涉及耐药机制的情况下具有理想的抗肿瘤作用;然而,到目前为止,还没有开发出这样的mTORC2抑制剂。通过硅筛选和药物化学优化,我们确定了几个与mTORC2独特成分SIN1结合的小分子。我们证明这种SIN1抑制剂可以改变翻译后修饰、蛋白-蛋白相互作用,并阻断mTORC2和雷帕霉素敏感(RS) mtorc1介导的信号传导。SIN1抑制剂还抑制野生型RAS激活和下游MAPK信号传导,以及多种癌细胞系类型的细胞增殖。抑制SIN1可以提高FDA批准的抗肿瘤药物的体外疗效,并可能为治疗不同类型的恶性肿瘤提供新的途径。
{"title":"Development of a SIN1 targeting inhibitor as a novel therapeutic approach for the treatment of malignancies.","authors":"Elspeth M Beauchamp, Atul Jain, Matt Clutter, Dominik Nahotko, Emely Lopez Fajardo, Purav P Vagadia, Sara F Dunne, Alain Mina, Mariafausta Fischietti, Chidera V Oku, Gavin T Blyth, Rama K Mishra, Elena Karras, Sara G Radecki, Connor Lantz, Sidharth Addepalli, Mirage Modi, Lihua Zou, Byoung-Kyu Cho, Young Ah Goo, Jenna Rossoff, Eleanor N Fish, Diana Saleiro, Gary E Schiltz, Leonidas C Platanias","doi":"10.1158/1535-7163.MCT-25-0298","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0298","url":null,"abstract":"<p><p>The essential role of mTOR in promoting tumorigenesis of many cancers makes it an attractive therapeutic target. However, catalytic mTOR inhibitors, which block both mTORC1 and mTORC2, result in activation of negative feedback loops as a resistance mechanism. Selective mTORC2 inhibitors are expected to have the desired anti-tumor effects without engaging resistance mechanisms; however, to date, no such mTORC2 inhibitors have been developed. Using in-silico screening and medicinal chemistry optimization, we identified several small molecules that bind to the unique mTORC2 component, SIN1. We demonstrate that this SIN1 inhibitor alters post-translational modification, protein-protein interactions, and blocks mTORC2- and rapamycin sensitive (RS) mTORC1-mediated signaling. The SIN1 inhibitor also inhibits wildtype RAS activation and downstream MAPK signaling, as well as cell proliferation of multiple cancer cell line types. SIN1 inhibition can enhance the efficacy of FDA- approved anti-neoplastic agents in vitro and may provide a novel approach for the treatment of different types of malignancies.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145804814","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CHS-114: an afucosylated anti-CCR8 monoclonal antibody that selectively depletes intratumoral Treg cells and induces antitumor immune responses. CHS-114:一种聚焦抗ccr8单克隆抗体,选择性地消耗肿瘤内Treg细胞并诱导抗肿瘤免疫反应。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-22 DOI: 10.1158/1535-7163.MCT-25-0367
Xiaoguang Wang, Varun N Kapoor, Daniel J Chin, Scott L Klakamp, Federico Baruffaldi, James F Mohan, Robert Haines, Austin Dulak, Marisella Panduro, Yue Ren, Ricard Masia, Jonathan A Hill, Theresa M LaVallee, Narendiran Rajasekaran

Intratumoral T regulatory cells (Tregs) promote an immunosuppressive tumor microenvironment and are frequently associated with a lack of response to immunotherapy. Selective targeting of intratumoral Tregs while sparing broader Tregs and effector T cell populations is an attractive strategy to enhance antitumor immune responses. CCR8 is a G protein-coupled receptor (GPCR) that is predominantly upregulated on tumor resident Tregs in a range of human solid tumors making it a promising target for their selective depletion. In preclinical studies using the mouse tumor models, anti-mouse CCR8 antibody treatment resulted in depletion of CCR8+ intratumoral Tregs, significant antitumor activity and enhanced survival in combination with anti-PD-1. CHS-114 is a highly selective, afucosylated human anti-CCR8 monoclonal antibody that is being developed as a cancer immunotherapy. CHS-114 selectively binds human CCR8 and potently kills CCR8 expressing cells by inducing antibody dependent cellular cytotoxicity (ADCC) and antibody dependent cellular phagocytosis (ADCP). Ex vivo studies evaluating human dissociated tumor cells (DTCs) demonstrated the selectivity of CHS-114 in depleting intratumoral Tregs while sparing CCR8 negative Tregs and effector T cells. Treatment of tumor bearing human CCR8 knock-in (huCCR8KI) mice with CHS-114 resulted in significant tumor growth inhibition (62.6%) accompanied by remodeling of the tumor immune microenvironment and enhanced differentiation of a subset of cytotoxic CD8+ T cells. Based on the promising preclinical data, we are evaluating CHS-114 in clinical trials as an investigational agent for the treatment of solid tumors with and without the anti-PD-1 antibody toripalimab (NCT05635643, NCT06657144).

肿瘤内T调节细胞(Tregs)促进免疫抑制肿瘤微环境,并且经常与免疫治疗缺乏反应相关。选择性靶向肿瘤内treg,同时保留更广泛的treg和效应T细胞群,是增强抗肿瘤免疫反应的一种有吸引力的策略。CCR8是一种G蛋白偶联受体(GPCR),在一系列人类实体肿瘤中主要在肿瘤驻留treg上表达上调,使其成为选择性清除treg的一个有希望的靶点。在使用小鼠肿瘤模型的临床前研究中,抗小鼠CCR8抗体治疗导致CCR8+瘤内Tregs的消耗,显著的抗肿瘤活性和抗pd -1联合提高生存率。CHS-114是一种高选择性的,集中的人抗ccr8单克隆抗体,正在开发作为癌症免疫治疗。CHS-114选择性结合人CCR8,通过诱导抗体依赖性细胞毒性(ADCC)和抗体依赖性细胞吞噬(ADCP),有效杀伤表达CCR8的细胞。评估人游离肿瘤细胞(dtc)的离体研究表明,CHS-114在消耗瘤内Tregs方面具有选择性,同时保留CCR8阴性Tregs和效应T细胞。用CHS-114治疗携带肿瘤的人CCR8敲入(huCCR8KI)小鼠,可显著抑制肿瘤生长(62.6%),并伴有肿瘤免疫微环境的重塑和细胞毒性CD8+ T细胞亚群的分化增强。基于有前景的临床前数据,我们正在临床试验中评估CHS-114作为联合或不联合抗pd -1抗体torpalimab (NCT05635643, NCT06657144)治疗实体肿瘤的研究药物。
{"title":"CHS-114: an afucosylated anti-CCR8 monoclonal antibody that selectively depletes intratumoral Treg cells and induces antitumor immune responses.","authors":"Xiaoguang Wang, Varun N Kapoor, Daniel J Chin, Scott L Klakamp, Federico Baruffaldi, James F Mohan, Robert Haines, Austin Dulak, Marisella Panduro, Yue Ren, Ricard Masia, Jonathan A Hill, Theresa M LaVallee, Narendiran Rajasekaran","doi":"10.1158/1535-7163.MCT-25-0367","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0367","url":null,"abstract":"<p><p>Intratumoral T regulatory cells (Tregs) promote an immunosuppressive tumor microenvironment and are frequently associated with a lack of response to immunotherapy. Selective targeting of intratumoral Tregs while sparing broader Tregs and effector T cell populations is an attractive strategy to enhance antitumor immune responses. CCR8 is a G protein-coupled receptor (GPCR) that is predominantly upregulated on tumor resident Tregs in a range of human solid tumors making it a promising target for their selective depletion. In preclinical studies using the mouse tumor models, anti-mouse CCR8 antibody treatment resulted in depletion of CCR8+ intratumoral Tregs, significant antitumor activity and enhanced survival in combination with anti-PD-1. CHS-114 is a highly selective, afucosylated human anti-CCR8 monoclonal antibody that is being developed as a cancer immunotherapy. CHS-114 selectively binds human CCR8 and potently kills CCR8 expressing cells by inducing antibody dependent cellular cytotoxicity (ADCC) and antibody dependent cellular phagocytosis (ADCP). Ex vivo studies evaluating human dissociated tumor cells (DTCs) demonstrated the selectivity of CHS-114 in depleting intratumoral Tregs while sparing CCR8 negative Tregs and effector T cells. Treatment of tumor bearing human CCR8 knock-in (huCCR8KI) mice with CHS-114 resulted in significant tumor growth inhibition (62.6%) accompanied by remodeling of the tumor immune microenvironment and enhanced differentiation of a subset of cytotoxic CD8+ T cells. Based on the promising preclinical data, we are evaluating CHS-114 in clinical trials as an investigational agent for the treatment of solid tumors with and without the anti-PD-1 antibody toripalimab (NCT05635643, NCT06657144).</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145804777","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Lipophilic Statins Deplete GPX4 to Promote Ferroptosis and Sensitize Cancer Cells to Immune Checkpoint Blockade. 亲脂性他汀类药物消耗GPX4促进铁凋亡并使癌细胞对免疫检查点封锁敏感。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-22 DOI: 10.1158/1535-7163.MCT-25-0667
Ronan Talty, Veronica T Brooks, Meaghan K McGeary, Simon Milette, Sijin Zheng, Karine Flem-Karlsen, Andrew Daniels, Maya Deshmukh, Koonam Park, Will Caraccio, Hong Yan, Caroline Echeandia-Francis, Madeline McNamara, Suyeon Hong, David S Kirwin, Kelly Olino, Caroline H Johnson, Marcus Bosenberg, Goran Micevic, Simon F Roy

Statins have been reported to exert anticancer activity, varying with cancer type and specific statins. These findings suggest that more mechanistic insights into the anticancer effects of statins are needed. In this study, we interrogated the ability of statins to induce cell death and ferroptosis in melanoma and colorectal cancer. First, we showed that statins induce cell death in patient-derived melanoma cell lines and that lower expression of mevalonate pathway genes correlates with increased CD8+ T-cell infiltration and improved overall survival in patients with melanoma. We found that lipophilic statins induce cell death with features of ferroptosis. Transcriptional data also revealed system-level changes to a variety of ferroptosis-related pathways. We found that mevalonate rescued statin-induced cell death. Mechanistically, mevalonate-derived isopentyl pyrophosphate is necessary for isopentylation of tRNA [Ser]Sec, which is required for efficient synthesis of the selenoprotein ferroptosis suppressor GPX4. Given the emerging role for ferroptosis in antitumor immunity, we tested lipophilic statins, including simvastatin, alone and in combination with α-PD1 in vivo and found that simvastatin and α-PD1 promoted tumor clearance and extended survival in 20% to 60% of mice alone but in nearly 100% of mice when administered together. Simvastatin also depleted GPX4 in vivo. These results highlight the therapeutic potential of statin use in combination with immunotherapies.

据报道,他汀类药物具有抗癌活性,但因癌症类型和特定的他汀类药物而异。这些发现表明,需要对他汀类药物的抗癌作用进行更多的机制研究。在这项研究中,我们探讨了他汀类药物在黑色素瘤和结直肠癌中诱导细胞死亡和铁下垂的能力。首先,我们发现他汀类药物诱导患者源性黑色素瘤细胞系的细胞死亡,甲羟戊酸途径基因的低表达与黑色素瘤患者CD8+ t细胞浸润增加和总体生存率提高相关。我们发现亲脂性他汀类药物诱导细胞死亡并伴有铁下垂的特征。转录数据还揭示了系统水平的变化,各种铁衰相关的途径。我们发现甲羟戊酸挽救了他汀类药物诱导的细胞死亡。从机制上说,甲戊酸衍生的焦磷酸异戊基是tRNA [Ser]Sec异戊基化所必需的,而tRNA [Ser]Sec异戊基化是硒蛋白铁沉抑制因子GPX4有效合成所必需的。鉴于铁中毒在抗肿瘤免疫中的新作用,我们在体内测试了亲脂性他汀类药物,包括辛伐他汀单独使用和与α-PD1联合使用,发现辛伐他汀和α-PD1在单独使用的小鼠中可促进20%至60%的肿瘤清除和延长生存期,而在联合使用的小鼠中,这一比例接近100%。辛伐他汀也在体内减少GPX4。这些结果突出了他汀类药物与免疫疗法联合使用的治疗潜力。
{"title":"Lipophilic Statins Deplete GPX4 to Promote Ferroptosis and Sensitize Cancer Cells to Immune Checkpoint Blockade.","authors":"Ronan Talty, Veronica T Brooks, Meaghan K McGeary, Simon Milette, Sijin Zheng, Karine Flem-Karlsen, Andrew Daniels, Maya Deshmukh, Koonam Park, Will Caraccio, Hong Yan, Caroline Echeandia-Francis, Madeline McNamara, Suyeon Hong, David S Kirwin, Kelly Olino, Caroline H Johnson, Marcus Bosenberg, Goran Micevic, Simon F Roy","doi":"10.1158/1535-7163.MCT-25-0667","DOIUrl":"10.1158/1535-7163.MCT-25-0667","url":null,"abstract":"<p><p>Statins have been reported to exert anticancer activity, varying with cancer type and specific statins. These findings suggest that more mechanistic insights into the anticancer effects of statins are needed. In this study, we interrogated the ability of statins to induce cell death and ferroptosis in melanoma and colorectal cancer. First, we showed that statins induce cell death in patient-derived melanoma cell lines and that lower expression of mevalonate pathway genes correlates with increased CD8+ T-cell infiltration and improved overall survival in patients with melanoma. We found that lipophilic statins induce cell death with features of ferroptosis. Transcriptional data also revealed system-level changes to a variety of ferroptosis-related pathways. We found that mevalonate rescued statin-induced cell death. Mechanistically, mevalonate-derived isopentyl pyrophosphate is necessary for isopentylation of tRNA [Ser]Sec, which is required for efficient synthesis of the selenoprotein ferroptosis suppressor GPX4. Given the emerging role for ferroptosis in antitumor immunity, we tested lipophilic statins, including simvastatin, alone and in combination with α-PD1 in vivo and found that simvastatin and α-PD1 promoted tumor clearance and extended survival in 20% to 60% of mice alone but in nearly 100% of mice when administered together. Simvastatin also depleted GPX4 in vivo. These results highlight the therapeutic potential of statin use in combination with immunotherapies.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"OF1-OF12"},"PeriodicalIF":5.5,"publicationDate":"2025-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12752129/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145804775","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Histone Modifier KANSL2 Is an Actionable Biomarker in Multiple Myeloma. 组蛋白修饰因子KANSL2是多发性骨髓瘤的可操作生物标志物。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-19 DOI: 10.1158/1535-7163.MCT-25-0379
Kaiting Jiang, Marieluise Kirchner, Frederik Herzberg, Yan Zhao, Amelie Gasper, Francis Baumgartner, Paul Jung, Jan Braune, Veronika Schulze, Konstandina Isaakidis, Philipp Mertins, Jan Krönke, Matthias Wirth, Ulrich Keller, Stefan Habringer

Epigenetic aberrations are key drivers of multiple myeloma, yet targeted therapies exploiting epigenetic alterations have not been established. By integrating clinical and molecular datasets of patients with multiple myeloma with an unbiased genetic in vivo screen, we identified KAT8 regulatory NSL complex subunit 2 (KANSL2) as a histone posttranslational modification-associated candidate oncogene. High expression of KANSL2 was associated with adverse prognosis in patients with multiple myeloma. Genetic gain- and loss-of-function models identified a protective role of KANSL2 toward genotoxic stress. By transcriptomics, proteomics, and quantitative acetylome profiling, we identified a KANSL2-dependent specific molecular program targetable by acetylation-related modifiers. High KANSL2 levels increased sensitivity to the histone deacetylase (HDAC) inhibitor panobinostat and bromodomain and extra-terminal motif (BET) inhibitor OTX-015 and their combination. Ex vivo drug response profiling in samples from patients with relapsed/refractory multiple myeloma confirmed that high KANSL2 expression is associated with selective multiple myeloma cell killing by HDAC and BET inhibitors. Collectively, these findings position KANSL2 as a mediator of chemotherapy resistance and actionable biomarker for response to drugs targeting its epigenetic program.

表观遗传畸变是多发性骨髓瘤的关键驱动因素,但利用表观遗传改变的靶向治疗尚未建立。通过整合多发性骨髓瘤患者的临床和分子数据集以及无偏倚的体内遗传筛选,我们确定了KAT8调节性NSL复合物亚基2 (KANSL2)作为组蛋白翻译后修饰相关的候选癌基因。高表达的KANSL2与多发性骨髓瘤患者的不良预后相关。遗传功能增益和功能丧失模型确定了KANSL2对遗传毒性应激的保护作用。通过转录组学、蛋白质组学和定量乙酰化分析,我们确定了一个依赖于kansl2的特定分子程序,可被乙酰化相关修饰物靶向。高KANSL2水平增加了对组蛋白去乙酰化酶(HDAC)抑制剂panobinostat和bromodomain和extra-terminal motif (BET)抑制剂OTX-015及其组合的敏感性。来自复发/难治性多发性骨髓瘤患者样本的体外药物反应分析证实,高KANSL2表达与HDAC和BET抑制剂选择性杀死多发性骨髓瘤细胞有关。总的来说,这些发现表明KANSL2是化疗耐药的中介,也是针对其表观遗传程序的药物反应的可操作的生物标志物。
{"title":"The Histone Modifier KANSL2 Is an Actionable Biomarker in Multiple Myeloma.","authors":"Kaiting Jiang, Marieluise Kirchner, Frederik Herzberg, Yan Zhao, Amelie Gasper, Francis Baumgartner, Paul Jung, Jan Braune, Veronika Schulze, Konstandina Isaakidis, Philipp Mertins, Jan Krönke, Matthias Wirth, Ulrich Keller, Stefan Habringer","doi":"10.1158/1535-7163.MCT-25-0379","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0379","url":null,"abstract":"<p><p>Epigenetic aberrations are key drivers of multiple myeloma, yet targeted therapies exploiting epigenetic alterations have not been established. By integrating clinical and molecular datasets of patients with multiple myeloma with an unbiased genetic in vivo screen, we identified KAT8 regulatory NSL complex subunit 2 (KANSL2) as a histone posttranslational modification-associated candidate oncogene. High expression of KANSL2 was associated with adverse prognosis in patients with multiple myeloma. Genetic gain- and loss-of-function models identified a protective role of KANSL2 toward genotoxic stress. By transcriptomics, proteomics, and quantitative acetylome profiling, we identified a KANSL2-dependent specific molecular program targetable by acetylation-related modifiers. High KANSL2 levels increased sensitivity to the histone deacetylase (HDAC) inhibitor panobinostat and bromodomain and extra-terminal motif (BET) inhibitor OTX-015 and their combination. Ex vivo drug response profiling in samples from patients with relapsed/refractory multiple myeloma confirmed that high KANSL2 expression is associated with selective multiple myeloma cell killing by HDAC and BET inhibitors. Collectively, these findings position KANSL2 as a mediator of chemotherapy resistance and actionable biomarker for response to drugs targeting its epigenetic program.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"OF1-OF14"},"PeriodicalIF":5.5,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145781580","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Vitamin C conditioning generates tumor-targeting CAR T cells with superior cytotoxicity and fitness in a post-transplant lymphoproliferative disorder tumor model. 在移植后淋巴增生性疾病肿瘤模型中,维生素C调节产生具有优越细胞毒性和适应性的肿瘤靶向CAR - T细胞。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-19 DOI: 10.1158/1535-7163.MCT-25-0005
Pegah Rahmati, Agnes Bonifacius, Anna Christina Dragon, Chiara Malinconico, Friederike Floegel, Anne Halenius, Rainer Blasczyk, Britta Maecker-Kolhoff, Michael Hudecek, Stefan Floess, Jochen Huehn, Britta Eiz-Vesper

CAR T-cell therapy has shown efficacy in hematological malignancies but faces challenges in solid tumors and virus-associated malignancies such as post-transplant lymphoproliferative disorder (PTLD). Various strategies, including optimization of receptor design, genetic modifications addressing immunomodulatory mechanisms and refining the manufacturing process have been explored to overcome limited in vivo persistence and tumor infiltration, antigen escape and the immunosuppressive tumor microenvironment (TME). This study investigated the effect of vitamin C (vitC) conditioning on CD19-targeting CAR T cells (vitC-CAR19-Ts) to improve efficacy of CAR T-cell therapy. VitC has been shown to influence immune responses through epigenetic regulation and oxidative stress reduction. Enhanced transduction efficiency and proliferative capacity by vitC conditioning resulted in a higher yield of CD4+ and CD8+ CAR19-Ts. VitC-CAR19-Ts exhibited faster and improved cytotoxic response towards CD19+ Nalm-6 cells and Epstein-Barr virus (EBV)-infected B-lymphoblastoid cell lines (B-LCLs), the in vitro model of PTLD. Increased demethylation was observed in TBX21 regions, which was in line with a type 1-like phenotype and higher expression of effector molecules such as Granulysin in both CD4+ and CD8+ in vitC-CAR19-Ts, providing insights into the effects of vitC conditioning. Importantly, vitC-CAR19-Ts outperformed CAR19-Ts in long-term antigen stress assays and 3D multicellular spheroid models, indicating a potentially improved in vivo functionality and tumor infiltration capacity. In summary, vitC conditioning represents a promising strategy to enhance CAR T-cell yield, cytotoxic potential and durability, complementing existing approaches to overcome the limitations of CAR T cells in the treatment of hematological malignancies and solid tumors.

CAR -t细胞疗法在血液系统恶性肿瘤中显示出疗效,但在实体瘤和病毒相关恶性肿瘤(如移植后淋巴细胞增生性疾病(PTLD))中面临挑战。为了克服有限的体内持久性和肿瘤浸润、抗原逃逸和免疫抑制肿瘤微环境(TME),研究人员探索了各种策略,包括优化受体设计、针对免疫调节机制的遗传修饰和改进制造工艺。本研究探讨了维生素C (vitC)调节cd19靶向CAR -T细胞(vitC- car19 - ts)以提高CAR -T细胞治疗效果的作用。维生素c已被证明通过表观遗传调节和氧化应激减少影响免疫反应。通过维生素c调节提高转导效率和增殖能力,导致CD4+和CD8+ CAR19-Ts的产量增加。VitC-CAR19-Ts对CD19+ n- 6细胞和eb病毒(EBV)感染的b淋巴母细胞样细胞系(B-LCLs) (PTLD的体外模型)表现出更快和更好的细胞毒性反应。在TBX21区域观察到去甲基化增加,这与维生素c - car19 - ts的1型样表型和效应分子如Granulysin在CD4+和CD8+中的高表达一致,为维生素c调节的作用提供了见解。重要的是,vitC-CAR19-Ts在长期抗原应激试验和3D多细胞球体模型中表现优于CAR19-Ts,这表明vitC-CAR19-Ts可能改善体内功能和肿瘤浸润能力。综上所述,维生素c调节是提高CAR - T细胞产量、细胞毒性潜能和持久性的一种有前景的策略,补充了克服CAR - T细胞治疗血液系统恶性肿瘤和实体瘤局限性的现有方法。
{"title":"Vitamin C conditioning generates tumor-targeting CAR T cells with superior cytotoxicity and fitness in a post-transplant lymphoproliferative disorder tumor model.","authors":"Pegah Rahmati, Agnes Bonifacius, Anna Christina Dragon, Chiara Malinconico, Friederike Floegel, Anne Halenius, Rainer Blasczyk, Britta Maecker-Kolhoff, Michael Hudecek, Stefan Floess, Jochen Huehn, Britta Eiz-Vesper","doi":"10.1158/1535-7163.MCT-25-0005","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0005","url":null,"abstract":"<p><p>CAR T-cell therapy has shown efficacy in hematological malignancies but faces challenges in solid tumors and virus-associated malignancies such as post-transplant lymphoproliferative disorder (PTLD). Various strategies, including optimization of receptor design, genetic modifications addressing immunomodulatory mechanisms and refining the manufacturing process have been explored to overcome limited in vivo persistence and tumor infiltration, antigen escape and the immunosuppressive tumor microenvironment (TME). This study investigated the effect of vitamin C (vitC) conditioning on CD19-targeting CAR T cells (vitC-CAR19-Ts) to improve efficacy of CAR T-cell therapy. VitC has been shown to influence immune responses through epigenetic regulation and oxidative stress reduction. Enhanced transduction efficiency and proliferative capacity by vitC conditioning resulted in a higher yield of CD4+ and CD8+ CAR19-Ts. VitC-CAR19-Ts exhibited faster and improved cytotoxic response towards CD19+ Nalm-6 cells and Epstein-Barr virus (EBV)-infected B-lymphoblastoid cell lines (B-LCLs), the in vitro model of PTLD. Increased demethylation was observed in TBX21 regions, which was in line with a type 1-like phenotype and higher expression of effector molecules such as Granulysin in both CD4+ and CD8+ in vitC-CAR19-Ts, providing insights into the effects of vitC conditioning. Importantly, vitC-CAR19-Ts outperformed CAR19-Ts in long-term antigen stress assays and 3D multicellular spheroid models, indicating a potentially improved in vivo functionality and tumor infiltration capacity. In summary, vitC conditioning represents a promising strategy to enhance CAR T-cell yield, cytotoxic potential and durability, complementing existing approaches to overcome the limitations of CAR T cells in the treatment of hematological malignancies and solid tumors.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145781637","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cross-resistance of Belinostat and Romidepsin in Non-T Follicular Helper Peripheral T-cell Lymphoma Models Suggests Subtype-Specific Implications for Belinostat-CHOP. Belinostat和罗米地辛在非t滤泡辅助外周t细胞淋巴瘤模型中的交叉耐药提示Belinostat- chop的亚型特异性意义
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-17 DOI: 10.1158/1535-7163.MCT-25-0941
Jacob C Pantazis, Adam C Palmer

The phase III study of romidepsin plus CHOP for peripheral T-cell lymphoma (PTCL) yielded negative results in the overall analysis and showed that outcomes were only improved in the T follicular helper (TFH) subgroup of PTCL, with no benefit in non-TFH PTCL. Belinostat, another histone deacetylase (HDAC) inhibitor, is now the primary candidate to test whether HDAC inhibition can improve treatment outcomes in first-line PTCL. Whether belinostat shares romidepsin's TFH-specific clinical activity remains uncertain, and therefore whether it is better to include or exclude non-TFH subtypes may be a crucial choice for a forthcoming confirmatory trial of belinostat-CHOP in first-line PTCL. We evaluated whether these agents are cross-resistant in non-TFH lymphomas by conducting in vitro drug sensitivity profiling with belinostat, romidepsin, and other pairs of mechanistically related agents in 30 non-TFH human T- and NK-cell lymphoma cultures. Sensitivities to romidepsin and belinostat were strongly correlated (ρ = 0.77, P < 10-6) and comparable with correlations observed for other same-mechanism drug pairs (e.g., doxorubicin/etoposide) and significantly higher than for drug pairs without mechanistic similarity. These findings indicate substantial cross-resistance between romidepsin and belinostat in non-TFH PTCL. If belinostat shares romidepsin's subtype-specific activity, a first-line trial of belinostat plus CHOP with broad patient enrollment may risk diluting benefit in TFH lymphomas by including non-TFH cases, whereas a TFH-focused design could maximize the likelihood of the trial's success.

罗米地辛联合CHOP治疗外周T细胞淋巴瘤(PTCL)的III期研究在总体分析中得出阴性结果,并且表明仅在PTCL的T滤泡辅助(TFH)亚组中预后得到改善,而在非TFH PTCL中没有获益。Belinostat是另一种组蛋白去乙酰化酶(HDAC)抑制剂,目前是测试HDAC抑制是否可以改善一线PTCL治疗结果的主要候选药物。belinostat是否与罗米地辛具有相同的tfh特异性临床活性仍不确定,因此,纳入或排除非tfh亚型是否更好,可能是即将进行的一线PTCL中belinostat- chop验证性试验的关键选择。我们通过在30例非tfh人T细胞和nk细胞淋巴瘤培养物中使用belinostat、roidepsin和其他机制相关药物对体外药物敏感性分析,评估了这些药物在非tfh淋巴瘤中是否具有交叉耐药。对罗米地辛和贝利诺他的敏感性强相关(ρ = 0.77, P < 10-6),与其他同机制药物对(如阿霉素/依泊苷)的相关性相当,且显著高于无机制相似性的药物对。这些发现表明,在非tfh PTCL中,罗米地辛和贝林他汀存在明显的交叉耐药。如果belinostat与罗米地辛具有相同的亚型特异性活性,那么在广泛患者入组的一线试验中,belinostat加CHOP可能会因为纳入非TFH病例而有稀释TFH淋巴瘤疗效的风险,而以TFH为重点的设计可以最大限度地提高试验成功的可能性。
{"title":"Cross-resistance of Belinostat and Romidepsin in Non-T Follicular Helper Peripheral T-cell Lymphoma Models Suggests Subtype-Specific Implications for Belinostat-CHOP.","authors":"Jacob C Pantazis, Adam C Palmer","doi":"10.1158/1535-7163.MCT-25-0941","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0941","url":null,"abstract":"<p><p>The phase III study of romidepsin plus CHOP for peripheral T-cell lymphoma (PTCL) yielded negative results in the overall analysis and showed that outcomes were only improved in the T follicular helper (TFH) subgroup of PTCL, with no benefit in non-TFH PTCL. Belinostat, another histone deacetylase (HDAC) inhibitor, is now the primary candidate to test whether HDAC inhibition can improve treatment outcomes in first-line PTCL. Whether belinostat shares romidepsin's TFH-specific clinical activity remains uncertain, and therefore whether it is better to include or exclude non-TFH subtypes may be a crucial choice for a forthcoming confirmatory trial of belinostat-CHOP in first-line PTCL. We evaluated whether these agents are cross-resistant in non-TFH lymphomas by conducting in vitro drug sensitivity profiling with belinostat, romidepsin, and other pairs of mechanistically related agents in 30 non-TFH human T- and NK-cell lymphoma cultures. Sensitivities to romidepsin and belinostat were strongly correlated (ρ = 0.77, P < 10-6) and comparable with correlations observed for other same-mechanism drug pairs (e.g., doxorubicin/etoposide) and significantly higher than for drug pairs without mechanistic similarity. These findings indicate substantial cross-resistance between romidepsin and belinostat in non-TFH PTCL. If belinostat shares romidepsin's subtype-specific activity, a first-line trial of belinostat plus CHOP with broad patient enrollment may risk diluting benefit in TFH lymphomas by including non-TFH cases, whereas a TFH-focused design could maximize the likelihood of the trial's success.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"OF1-OF6"},"PeriodicalIF":5.5,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145768484","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Molecular Cancer Therapeutics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1