Pub Date : 2025-12-30DOI: 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.
{"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}
Pub Date : 2025-12-30DOI: 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.
{"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}
Pub Date : 2025-12-24DOI: 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.
{"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}
Pub Date : 2025-12-23DOI: 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}
Pub Date : 2025-12-22DOI: 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.
{"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}
Pub Date : 2025-12-22DOI: 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).
{"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}
Pub Date : 2025-12-22DOI: 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.
{"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}
Pub Date : 2025-12-19DOI: 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.
{"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}
Pub Date : 2025-12-19DOI: 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.
{"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}
Pub Date : 2025-12-17DOI: 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}