首页 > 最新文献

Molecular Cancer Therapeutics最新文献

英文 中文
ASCL1 restrains ERK1/2 to promote survival of a subset of neuroendocrine lung cancers. ASCL1抑制ERK1/2,促进神经内分泌肺癌亚群的存活。
IF 5.7 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-19 DOI: 10.1158/1535-7163.mct-24-0355
Ana Martin-Vega,Svetlana A Earnest,Alexander Augustyn,Chonlarat Wichaidit,Luc Girard,Michael Peyton,John D Minna,Jane E Johnson,Melanie H Cobb
The transcription factor achaete-scute complex homolog 1 (ASCL1) is a lineage oncogene that is central in growth and survival of the majority of small cell lung cancers (SCLC) and neuroendocrine non-small cell lung cancers (NSCLC-NE) that express it. Targeting ASCL1, or its downstream pathways, remains a challenge. SCLCs and NSCLC-NE that express ASCL1 exhibit relatively low ERK1/2 activity, in dramatic contrast to NSCLCs in which the ERK pathway has a major role in pathogenesis. ERK1/2 inhibition in ASCL1-expressing lung tumor cells revealed down-regulation of ERK1/2 pathway suppressors SPRY4, SPRED1, DUSP6, and the transcription factor ETV5, which regulates DUSP6. CHIP-seq demonstrated that these genes are bound by ASCL1. Availability of a pharmacological inhibitor directed mechanistic studies towards DUSP6, an ERK1/2-selective phosphatase, in a subset of ASCL1-high NE lung tumors. Inhibition of DUSP6 increased active ERK1/2, which accumulated in the nucleus. Pharmacologic and genetic inhibition of DUSP6 reduced proliferation and survival of these cancers. Resistance developed in DUSP6 KO cells, indicating a bypass mechanism. Although targeting ASCL1 remains a challenge, our findings suggest that expression of ASCL1, DUSP6 and low phospho-ERK1/2 identify neuroendocrine lung cancers for which DUSP6 may be a therapeutic target.
转录因子Achaete-scute复合体同源物1(ASCL1)是一种系癌基因,在大多数表达它的小细胞肺癌(SCLC)和神经内分泌性非小细胞肺癌(NSCLC-NE)的生长和存活中起着核心作用。靶向 ASCL1 或其下游通路仍然是一项挑战。表达ASCL1的SCLC和NSCLC-NE的ERK1/2活性相对较低,这与ERK通路在发病机制中起主要作用的NSCLC形成鲜明对比。在表达 ASCL1 的肺肿瘤细胞中抑制 ERK1/2 发现,ERK1/2 通路抑制因子 SPRY4、SPRED1、DUSP6 和调控 DUSP6 的转录因子 ETV5 下调。CHIP-seq表明,这些基因与ASCL1结合。一种药理抑制剂的出现将机理研究引向了 ASCL1 高 NE 肺肿瘤亚群中的 DUSP6(一种 ERK1/2 选择性磷酸酶)。抑制 DUSP6 会增加活性 ERK1/2,使其在细胞核中积聚。药物和基因抑制 DUSP6 可减少这些癌症的增殖和存活。DUSP6 KO细胞产生了抗药性,这表明存在一种旁路机制。尽管以ASCL1为靶点仍是一个挑战,但我们的研究结果表明,ASCL1、DUSP6和低磷酸化ERK1/2的表达确定了神经内分泌性肺癌,DUSP6可能是其治疗靶点。
{"title":"ASCL1 restrains ERK1/2 to promote survival of a subset of neuroendocrine lung cancers.","authors":"Ana Martin-Vega,Svetlana A Earnest,Alexander Augustyn,Chonlarat Wichaidit,Luc Girard,Michael Peyton,John D Minna,Jane E Johnson,Melanie H Cobb","doi":"10.1158/1535-7163.mct-24-0355","DOIUrl":"https://doi.org/10.1158/1535-7163.mct-24-0355","url":null,"abstract":"The transcription factor achaete-scute complex homolog 1 (ASCL1) is a lineage oncogene that is central in growth and survival of the majority of small cell lung cancers (SCLC) and neuroendocrine non-small cell lung cancers (NSCLC-NE) that express it. Targeting ASCL1, or its downstream pathways, remains a challenge. SCLCs and NSCLC-NE that express ASCL1 exhibit relatively low ERK1/2 activity, in dramatic contrast to NSCLCs in which the ERK pathway has a major role in pathogenesis. ERK1/2 inhibition in ASCL1-expressing lung tumor cells revealed down-regulation of ERK1/2 pathway suppressors SPRY4, SPRED1, DUSP6, and the transcription factor ETV5, which regulates DUSP6. CHIP-seq demonstrated that these genes are bound by ASCL1. Availability of a pharmacological inhibitor directed mechanistic studies towards DUSP6, an ERK1/2-selective phosphatase, in a subset of ASCL1-high NE lung tumors. Inhibition of DUSP6 increased active ERK1/2, which accumulated in the nucleus. Pharmacologic and genetic inhibition of DUSP6 reduced proliferation and survival of these cancers. Resistance developed in DUSP6 KO cells, indicating a bypass mechanism. Although targeting ASCL1 remains a challenge, our findings suggest that expression of ASCL1, DUSP6 and low phospho-ERK1/2 identify neuroendocrine lung cancers for which DUSP6 may be a therapeutic target.","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142268794","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
Emerging Tumor-Agnostic Molecular Targets. 新出现的肿瘤诊断分子靶标。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-16 DOI: 10.1158/1535-7163.MCT-23-0725
Dedipya Bhamidipati, Alison M Schram

Advances in tumor molecular profiling have uncovered shared genomic and proteomic alterations across tumor types that can be exploited therapeutically. A biomarker-driven, disease-agnostic approach to oncology drug development can maximize the reach of novel therapeutics. To date, eight drug-biomarker pairs have been approved for the treatment of patients with advanced solid tumors with specific molecular profiles. Emerging biomarkers with the potential for clinical actionability across tumor types include gene fusions involving NRG1, FGFR1/2/3, BRAF, and ALK, mutations in TP53 Y220C, KRAS G12C, FGFR2/3, and BRAF non-V600 (Class II). We explore the growing evidence for clinical actionability of these biomarkers in patients with advanced solid tumors.

肿瘤分子图谱分析技术的进步发现了不同肿瘤类型共有的基因组和蛋白质组改变,这些改变可用于治疗。以生物标志物为驱动、以疾病诊断为导向的肿瘤药物开发方法可以最大限度地扩大新型疗法的覆盖范围。迄今为止,已有八种药物-生物标记物配对获批用于治疗具有特定分子特征的晚期实体瘤患者。具有跨肿瘤类型临床作用潜力的新兴生物标记物包括涉及 NRG1、FGFR1/2/3、BRAF 和 ALK 的基因融合、TP53 Y220C 突变、KRAS G12C、FGFR2/3 和 BRAF 非 V600(II 类)。我们将探讨越来越多的证据表明这些生物标记物在晚期实体瘤患者中的临床可操作性。
{"title":"Emerging Tumor-Agnostic Molecular Targets.","authors":"Dedipya Bhamidipati, Alison M Schram","doi":"10.1158/1535-7163.MCT-23-0725","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-23-0725","url":null,"abstract":"<p><p>Advances in tumor molecular profiling have uncovered shared genomic and proteomic alterations across tumor types that can be exploited therapeutically. A biomarker-driven, disease-agnostic approach to oncology drug development can maximize the reach of novel therapeutics. To date, eight drug-biomarker pairs have been approved for the treatment of patients with advanced solid tumors with specific molecular profiles. Emerging biomarkers with the potential for clinical actionability across tumor types include gene fusions involving NRG1, FGFR1/2/3, BRAF, and ALK, mutations in TP53 Y220C, KRAS G12C, FGFR2/3, and BRAF non-V600 (Class II). We explore the growing evidence for clinical actionability of these biomarkers in patients with advanced solid tumors.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142291433","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
Discovery and Characterization of Brigimadlin, a Novel and Highly Potent MDM2–p53 Antagonist Suitable for Intermittent Dose Schedules 发现并鉴定适合间歇性给药的新型强效 MDM2-p53 拮抗剂 Brigimadlin
IF 5.7 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-11 DOI: 10.1158/1535-7163.mct-23-0783
Andreas Gollner, Dorothea Rudolph, Ulrike Weyer-Czernilofsky, Rosa Baumgartinger, Peter Jung, Harald Weinstabl, Jürgen Ramharter, Rolf Grempler, Jens Quant, Jörg Rinnenthal, Alejandro Pérez Pitarch, Bojana Golubovic, Daniel Gerlach, Gerd Bader, Kristiane Wetzel, Sebastian Otto, Christian Mandl, Guido Boehmelt, Darryl B. McConnell, Norbert Kraut, Patrizia Sini
p53 is known as the guardian of the genome and is one of the most important tumor-suppressors. It is inactivated in most tumors, either via tumor protein p53 (TP53) gene mutation or copy number amplification of key negative regulators, e.g., mouse double minute 2 (MDM2). Compounds that bind to the MDM2 protein and disrupt its interaction with p53 restore p53 tumor suppressor activity, thereby promoting cell cycle arrest and apoptosis. Previous clinical experience with MDM2–p53 protein–protein interaction antagonists (MDM2–p53 antagonists) have demonstrated that thrombocytopenia and neutropenia represent on-target dose-limiting toxicities that might restrict their therapeutic utility. Dosing less frequently, while maintaining efficacious exposure, represents an approach to mitigate toxicity and improve the therapeutic window of MDM2–p53 antagonists. However, to achieve this, a molecule possessing excellent potency and ideal pharmacokinetic properties is required. Here, we present the discovery and characterization of brigimadlin (BI 907828), a novel, investigational spiro-oxindole MDM2–p53 antagonist. Brigimadlin exhibited high bioavailability and exposure, as well as dose-linear pharmacokinetics in preclinical models. Brigimadlin treatment restored p53 activity and led to apoptosis induction in preclinical models of TP53 wild-type, MDM2-amplified cancer. Oral administration of brigimadlin in an intermittent dosing schedule induced potent tumor growth inhibition in several TP53 wild-type, MDM2-amplified xenograft models. Exploratory clinical pharmacokinetic studies (NCT03449381) showed high systemic exposure and a long plasma elimination half-life in cancer patients who received oral brigimadlin. These findings support the continued clinical evaluation of brigimadlin in patients with MDM2-amplified cancers, such as dedifferentiated liposarcoma.
p53 被称为基因组的守护者,是最重要的肿瘤抑制因子之一。在大多数肿瘤中,由于肿瘤蛋白 p53(TP53)基因突变或关键负调控因子(如小鼠双分化 2(MDM2))的拷贝数扩增,p53 失活。与 MDM2 蛋白结合并破坏其与 p53 相互作用的化合物可恢复 p53 的肿瘤抑制活性,从而促进细胞周期停滞和细胞凋亡。以往使用 MDM2 蛋白-p53 蛋白相互作用拮抗剂(MDM2-p53 拮抗剂)的临床经验表明,血小板减少症和中性粒细胞减少症是靶向剂量限制性毒性,可能会限制其治疗效用。在保持有效暴露的同时减少给药次数,是减轻毒性和改善 MDM2-p53 拮抗剂治疗窗口期的一种方法。然而,要实现这一目标,需要一种具有卓越药效和理想药代动力学特性的分子。在此,我们介绍了一种新型、在研螺吲哚类 MDM2-p53 拮抗剂 Brigimadlin(BI 907828)的发现和表征。在临床前模型中,Brigimadlin 表现出高生物利用度和暴露度,以及剂量线性药代动力学。在 TP53 野生型、MDM2-扩增的癌症临床前模型中,Brigimadlin 治疗可恢复 p53 活性并诱导细胞凋亡。在几种 TP53 野生型、MDM2-扩增的异种移植模型中,以间歇给药方式口服布瑞吉玛德林能有效抑制肿瘤生长。探索性临床药代动力学研究(NCT03449381)显示,口服布瑞吉玛德林的癌症患者全身暴露率高,血浆消除半衰期长。这些研究结果支持继续对MDM2-扩增的癌症患者(如已分化的脂肪肉瘤)进行临床评估。
{"title":"Discovery and Characterization of Brigimadlin, a Novel and Highly Potent MDM2–p53 Antagonist Suitable for Intermittent Dose Schedules","authors":"Andreas Gollner, Dorothea Rudolph, Ulrike Weyer-Czernilofsky, Rosa Baumgartinger, Peter Jung, Harald Weinstabl, Jürgen Ramharter, Rolf Grempler, Jens Quant, Jörg Rinnenthal, Alejandro Pérez Pitarch, Bojana Golubovic, Daniel Gerlach, Gerd Bader, Kristiane Wetzel, Sebastian Otto, Christian Mandl, Guido Boehmelt, Darryl B. McConnell, Norbert Kraut, Patrizia Sini","doi":"10.1158/1535-7163.mct-23-0783","DOIUrl":"https://doi.org/10.1158/1535-7163.mct-23-0783","url":null,"abstract":"p53 is known as the guardian of the genome and is one of the most important tumor-suppressors. It is inactivated in most tumors, either via tumor protein p53 (TP53) gene mutation or copy number amplification of key negative regulators, e.g., mouse double minute 2 (MDM2). Compounds that bind to the MDM2 protein and disrupt its interaction with p53 restore p53 tumor suppressor activity, thereby promoting cell cycle arrest and apoptosis. Previous clinical experience with MDM2–p53 protein–protein interaction antagonists (MDM2–p53 antagonists) have demonstrated that thrombocytopenia and neutropenia represent on-target dose-limiting toxicities that might restrict their therapeutic utility. Dosing less frequently, while maintaining efficacious exposure, represents an approach to mitigate toxicity and improve the therapeutic window of MDM2–p53 antagonists. However, to achieve this, a molecule possessing excellent potency and ideal pharmacokinetic properties is required. Here, we present the discovery and characterization of brigimadlin (BI 907828), a novel, investigational spiro-oxindole MDM2–p53 antagonist. Brigimadlin exhibited high bioavailability and exposure, as well as dose-linear pharmacokinetics in preclinical models. Brigimadlin treatment restored p53 activity and led to apoptosis induction in preclinical models of TP53 wild-type, MDM2-amplified cancer. Oral administration of brigimadlin in an intermittent dosing schedule induced potent tumor growth inhibition in several TP53 wild-type, MDM2-amplified xenograft models. Exploratory clinical pharmacokinetic studies (NCT03449381) showed high systemic exposure and a long plasma elimination half-life in cancer patients who received oral brigimadlin. These findings support the continued clinical evaluation of brigimadlin in patients with MDM2-amplified cancers, such as dedifferentiated liposarcoma.","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142194516","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 the synthetic lethal relationship between FOCAD and TUT7 represents a potential therapeutic opportunity for TUT4/7 small molecule inhibitors in cancer. 针对 FOCAD 和 TUT7 之间的合成致死关系,是 TUT4/7 小分子抑制剂治疗癌症的潜在机会。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-05 DOI: 10.1158/1535-7163.MCT-24-0176
Robinson Triboulet, Khikmet Sadykov, Darren M Harvey, David M Wilson, Michael J Steinbaugh, Christopher B Mayo, Dillon Hawley, Andrew Madanjian, Corey Fyfe, Christina Bracken, Izarys Rivera-Rivera, Anna Ericsson, Andrew R Snyder, Sarah K Knutson, Ross L Stein, Veronica Gibaja, Shomir Ghosh, Robert M Campbell

Targeting synthetic lethal interactions between genes has emerged as a promising strategy for cancer therapy. This study explores the intricate interplay between terminal uridyltransferase 4 (TUT4) and terminal uridyltransferase 7 (TUT7), the 3'-5' exoribonuclease DIS3L2, and the SKI complex-interacting factor Focadhesin (FOCAD) in the context of cancer vulnerability. Using CRISPR and public functional genomics data, we show impairment of cell proliferation upon knockout of TUT7 or DIS3L2, but not TUT4, on cancer cells with FOCAD loss. Moreover, we report the characterization of the first potent and selective TUT4/7 inhibitors that substantially reduce uridylation and demonstrate in vitro and in vivo antiproliferative activity specifically in FOCAD-deleted cancer. FOCAD deficiency post-transcriptionally disrupts the stability of the SKI complex, whose role is to safeguard cells against aberrant RNA. Re-introduction of FOCAD restores the SKI complex and makes these cells less sensitive to TUT4/7 inhibitors, indicating that TUT7 dependency is FOCAD loss-driven. We propose a model where, in absence of FOCAD, TUT7 and DIS3L2 function as a salvage mechanism that degrades aberrant RNA, and genetic or pharmacological inhibition of this pathway leads to cell death. Our findings underscore the significance of FOCAD loss as a genetic driver of TUT7 vulnerability and provide insights into the potential utility of TUT4/7 inhibitors for cancer treatment.

靶向基因间的合成致死相互作用已成为一种很有前景的癌症治疗策略。本研究探讨了末端尿苷基转移酶4(TUT4)和末端尿苷基转移酶7(TUT7)、3'-5'外切核酸酶DIS3L2和SKI复合物相互作用因子Focadhesin(FOCAD)之间在癌症易感性方面错综复杂的相互作用。利用 CRISPR 和公开的功能基因组学数据,我们发现在 FOCAD 缺失的癌细胞上敲除 TUT7 或 DIS3L2 后,细胞增殖会受到影响,而敲除 TUT4 则不会。此外,我们还报告了第一种强效选择性 TUT4/7 抑制剂的特性,这种抑制剂能显著降低尿苷酸化,并在体外和体内显示出专门针对 FOCAD 缺失癌症的抗增殖活性。FOCAD 的缺乏会在转录后破坏 SKI 复合物的稳定性,而 SKI 复合物的作用是保护细胞免受异常 RNA 的侵害。重新引入FOCAD可恢复SKI复合物,并使这些细胞对TUT4/7抑制剂不那么敏感,这表明TUT7依赖性是由FOCAD缺失驱动的。我们提出了这样一个模型:在缺乏FOCAD的情况下,TUT7和DIS3L2发挥着降解异常RNA的挽救机制的作用,而对这一途径的遗传或药物抑制会导致细胞死亡。我们的发现强调了FOCAD缺失作为TUT7脆弱性遗传驱动因素的重要性,并为TUT4/7抑制剂在癌症治疗中的潜在作用提供了启示。
{"title":"Targeting the synthetic lethal relationship between FOCAD and TUT7 represents a potential therapeutic opportunity for TUT4/7 small molecule inhibitors in cancer.","authors":"Robinson Triboulet, Khikmet Sadykov, Darren M Harvey, David M Wilson, Michael J Steinbaugh, Christopher B Mayo, Dillon Hawley, Andrew Madanjian, Corey Fyfe, Christina Bracken, Izarys Rivera-Rivera, Anna Ericsson, Andrew R Snyder, Sarah K Knutson, Ross L Stein, Veronica Gibaja, Shomir Ghosh, Robert M Campbell","doi":"10.1158/1535-7163.MCT-24-0176","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0176","url":null,"abstract":"<p><p>Targeting synthetic lethal interactions between genes has emerged as a promising strategy for cancer therapy. This study explores the intricate interplay between terminal uridyltransferase 4 (TUT4) and terminal uridyltransferase 7 (TUT7), the 3'-5' exoribonuclease DIS3L2, and the SKI complex-interacting factor Focadhesin (FOCAD) in the context of cancer vulnerability. Using CRISPR and public functional genomics data, we show impairment of cell proliferation upon knockout of TUT7 or DIS3L2, but not TUT4, on cancer cells with FOCAD loss. Moreover, we report the characterization of the first potent and selective TUT4/7 inhibitors that substantially reduce uridylation and demonstrate in vitro and in vivo antiproliferative activity specifically in FOCAD-deleted cancer. FOCAD deficiency post-transcriptionally disrupts the stability of the SKI complex, whose role is to safeguard cells against aberrant RNA. Re-introduction of FOCAD restores the SKI complex and makes these cells less sensitive to TUT4/7 inhibitors, indicating that TUT7 dependency is FOCAD loss-driven. We propose a model where, in absence of FOCAD, TUT7 and DIS3L2 function as a salvage mechanism that degrades aberrant RNA, and genetic or pharmacological inhibition of this pathway leads to cell death. Our findings underscore the significance of FOCAD loss as a genetic driver of TUT7 vulnerability and provide insights into the potential utility of TUT4/7 inhibitors for cancer treatment.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142133245","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
TR-107, An Agonist of Caseinolytic Peptidase Proteolytic Subunit, Disrupts Mitochondrial Metabolism and Inhibits the Growth of Human Colorectal Cancer Cells. TR-107是酪酸肽酶蛋白水解亚基的一种激动剂,它能破坏线粒体代谢并抑制人类结直肠癌细胞的生长。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-05 DOI: 10.1158/1535-7163.MCT-24-0170
Michael Giarrizzo, Joseph F LaComb, Hetvi R Patel, Rohan G Reddy, John D Haley, Lee M Graves, Edwin J Iwanowicz, Agnieszka B Bialkowska

Oxidative phosphorylation (OXPHOS) is an essential metabolic process for cancer proliferation and therapy resistance. The ClpXP complex maintains mitochondrial proteostasis by degrading misfolded proteins. Madera Therapeutics has developed a class of highly potent and selective small-molecule activators (TR compounds) of the ClpXP component caseinolytic peptidase proteolytic subunit (ClpP). This approach to cancer therapy eliminates substrate recognition and activates non-specific protease function within mitochondria, which has shown encouraging preclinical efficacy in multiple malignancies. The class-leading compound, TR-107, has demonstrated significantly improved potency in ClpP affinity and activation and enhanced pharmacokinetic properties over the multi-targeting clinical agent ONC201. In this study, we investigate the in vitro efficacy of TR-107 against human colorectal cancer (CRC) cells. TR-107 inhibited CRC cell proliferation in a dose- and time-dependent manner and induced cell cycle arrest at low nanomolar concentrations. Mechanistically, TR-107 downregulated the expression of proteins involved in the mitochondrial unfolded protein response (UPRmt) and mtDNA transcription and translation. TR-107 attenuated oxygen consumption rate and glycolytic compensation, confirming inactivation of OXPHOS and a reduction in total cellular respiration. Multi-omics analysis of treated cells indicated a downregulation of respiratory chain complex subunits and an upregulation of mitophagy and ferroptosis pathways. Further evaluation of ferroptosis revealed a depletion of antioxidant and iron toxicity defenses that could potentiate sensitivity to combinatory chemotherapeutics. Together, this study provides evidence and insight into the subcellular mechanisms employed by CRC cells in response to potent ClpP agonism. Our findings demonstrate a productive approach to disrupting mitochondrial metabolism, supporting the translational potential of TR-107.

氧化磷酸化(OXPHOS)是癌症增殖和抗药性的重要代谢过程。ClpXP 复合物通过降解折叠错误的蛋白质来维持线粒体蛋白稳态。Madera Therapeutics 公司已开发出一类 ClpXP 成分酪蛋白溶肽酶蛋白水解亚基(ClpP)的高效、选择性小分子激活剂(TR 化合物)。这种癌症治疗方法消除了底物识别,激活了线粒体内的非特异性蛋白酶功能,在多种恶性肿瘤中显示出令人鼓舞的临床前疗效。与多靶点临床药物ONC201相比,同类领先化合物TR-107在ClpP亲和力和激活方面的效力显著提高,药代动力学特性也有所增强。在本研究中,我们研究了 TR-107 对人类结直肠癌(CRC)细胞的体外疗效。在低纳摩尔浓度下,TR-107以剂量和时间依赖性方式抑制CRC细胞增殖,并诱导细胞周期停滞。从机理上讲,TR-107 下调了参与线粒体未折叠蛋白反应(UPRmt)以及 mtDNA 转录和翻译的蛋白质的表达。TR-107 降低了耗氧率和糖酵解补偿,证实了 OXPHOS 失活和细胞总呼吸量减少。对处理过的细胞进行的多组学分析表明,呼吸链复合物亚基下调,有丝分裂和铁凋亡途径上调。对铁变态反应的进一步评估显示,抗氧化和铁毒性防御功能耗竭,这可能会增强对联合化疗药物的敏感性。总之,这项研究为 CRC 细胞应对强效 ClpP 激动所采用的亚细胞机制提供了证据和见解。我们的研究结果表明了破坏线粒体代谢的有效方法,支持 TR-107 的转化潜力。
{"title":"TR-107, An Agonist of Caseinolytic Peptidase Proteolytic Subunit, Disrupts Mitochondrial Metabolism and Inhibits the Growth of Human Colorectal Cancer Cells.","authors":"Michael Giarrizzo, Joseph F LaComb, Hetvi R Patel, Rohan G Reddy, John D Haley, Lee M Graves, Edwin J Iwanowicz, Agnieszka B Bialkowska","doi":"10.1158/1535-7163.MCT-24-0170","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0170","url":null,"abstract":"<p><p>Oxidative phosphorylation (OXPHOS) is an essential metabolic process for cancer proliferation and therapy resistance. The ClpXP complex maintains mitochondrial proteostasis by degrading misfolded proteins. Madera Therapeutics has developed a class of highly potent and selective small-molecule activators (TR compounds) of the ClpXP component caseinolytic peptidase proteolytic subunit (ClpP). This approach to cancer therapy eliminates substrate recognition and activates non-specific protease function within mitochondria, which has shown encouraging preclinical efficacy in multiple malignancies. The class-leading compound, TR-107, has demonstrated significantly improved potency in ClpP affinity and activation and enhanced pharmacokinetic properties over the multi-targeting clinical agent ONC201. In this study, we investigate the in vitro efficacy of TR-107 against human colorectal cancer (CRC) cells. TR-107 inhibited CRC cell proliferation in a dose- and time-dependent manner and induced cell cycle arrest at low nanomolar concentrations. Mechanistically, TR-107 downregulated the expression of proteins involved in the mitochondrial unfolded protein response (UPRmt) and mtDNA transcription and translation. TR-107 attenuated oxygen consumption rate and glycolytic compensation, confirming inactivation of OXPHOS and a reduction in total cellular respiration. Multi-omics analysis of treated cells indicated a downregulation of respiratory chain complex subunits and an upregulation of mitophagy and ferroptosis pathways. Further evaluation of ferroptosis revealed a depletion of antioxidant and iron toxicity defenses that could potentiate sensitivity to combinatory chemotherapeutics. Together, this study provides evidence and insight into the subcellular mechanisms employed by CRC cells in response to potent ClpP agonism. Our findings demonstrate a productive approach to disrupting mitochondrial metabolism, supporting the translational potential of TR-107.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142133246","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
IL-15/IL-15Rα-Fc-Fusion Protein XmAb24306 Potentiates Activity of CD3 Bispecific Antibodies through Enhancing T-Cell Expansion. IL-15/IL-15Rα-Fc融合蛋白XmAb24306通过增强T细胞扩增来提高CD3双特异性抗体的活性。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-04 DOI: 10.1158/1535-7163.MCT-23-0910
Ji Li, Robyn Clark, Dionysos Slaga, Kendra Avery, Ke Liu, Suzanne Schubbert, Rajat Varma, Eugene Chiang, Klara Totpal, Matthew J Bernett, Patrick G Holder, Teemu T Junttila

An insufficient quantity of functional T cells is a likely factor limiting the clinical activity of T-cell bispecific antibodies, especially in solid tumor indications. We hypothesized that XmAb24306 (efbalropendekin alfa), a lymphoproliferative interleukin (IL)-15/IL-15 receptor α (IL-15Rα) Fc-fusion protein, may potentiate the activity of T-cell dependent (TDB) antibodies. The activation of human peripheral T cells by cevostamab, an anti-FcRH5/CD3 TDB, or anti-HER2/CD3 TDB resulted in the upregulation of the IL-2/15Rβ (CD122) receptor subunit in nearly all CD8+ and majority of CD4+ T cells, suggesting that TDB treatment may sensitize T cells to IL-15. XmAb24306 enhanced T-cell bispecific antibody-induced CD8+ and CD4+ T-cell proliferation and expansion. In vitro combination of XmAb24306 with cevostamab or anti-HER2/CD3 TDB resulted in significant enhancement of tumor cell killing, which was reversed when T-cell numbers were normalized, suggesting that T-cell expansion is the main mechanism of the observed benefit. Pretreatment of immunocompetent mice with a mouse-reactive surrogate of XmAb24306 (mIL-15-Fc) resulted in a significant increase of T cells in the blood, spleen, and tumors and converted transient anti-HER2/CD3 TDB responses to complete durable responses. In summary, our results support the hypothesis that the number of tumor-infiltrating T cells is rate limiting for the activity of solid tumor-targeting TDBs. Upregulation of CD122 by TDB treatment and the observed synergy with XmAb24306 and T-cell bispecific antibodies support clinical evaluation of this novel immunotherapy combination.

功能性 T 细胞数量不足可能是限制 T 细胞双特异性抗体临床活性的一个因素,尤其是在实体瘤适应症中。我们假设XmAb24306(efbalropendekin alfa)--一种淋巴细胞增殖性白细胞介素(IL)-15/IL-15受体α(IL-15Rα)Fc融合蛋白--可能会增强T细胞依赖性(TDB)抗体的活性。用塞伏司他(一种抗 FcRH5/CD3 TDB 或抗 HER2/CD3 TDB)激活人外周 T 细胞后,几乎所有 CD8+ T 细胞和大多数 CD4+ T 细胞中的 IL-2/15Rβ (CD122)受体亚基都会上调,这表明 TDB 处理可能会使 T 细胞对 IL-15 敏感。XmAb24306 可增强 T 细胞双特异性抗体诱导的 CD8+ 和 CD4+ T 细胞的增殖和扩增。体外将 XmAb24306 与塞夫司他单抗或抗 HER2/CD3 TDB 结合使用可显著增强对肿瘤细胞的杀伤力,而当 T 细胞数量正常化后,杀伤力又会逆转,这表明 T 细胞扩增是观察到的益处的主要机制。用XmAb24306的小鼠反应代用品(mIL-15-Fc)预处理免疫功能正常的小鼠会导致血液、脾脏和肿瘤中的T细胞显著增加,并将短暂的抗HER2/CD3 TDB反应转化为完全持久的反应。总之,我们的研究结果支持这样一个假设,即肿瘤浸润T细胞的数量限制了实体瘤靶向TDB的活性。TDB治疗对CD122的上调以及观察到的与XmAb24306和T细胞双特异性抗体的协同作用支持对这种新型免疫疗法组合进行临床评估。
{"title":"IL-15/IL-15Rα-Fc-Fusion Protein XmAb24306 Potentiates Activity of CD3 Bispecific Antibodies through Enhancing T-Cell Expansion.","authors":"Ji Li, Robyn Clark, Dionysos Slaga, Kendra Avery, Ke Liu, Suzanne Schubbert, Rajat Varma, Eugene Chiang, Klara Totpal, Matthew J Bernett, Patrick G Holder, Teemu T Junttila","doi":"10.1158/1535-7163.MCT-23-0910","DOIUrl":"10.1158/1535-7163.MCT-23-0910","url":null,"abstract":"<p><p>An insufficient quantity of functional T cells is a likely factor limiting the clinical activity of T-cell bispecific antibodies, especially in solid tumor indications. We hypothesized that XmAb24306 (efbalropendekin alfa), a lymphoproliferative interleukin (IL)-15/IL-15 receptor α (IL-15Rα) Fc-fusion protein, may potentiate the activity of T-cell dependent (TDB) antibodies. The activation of human peripheral T cells by cevostamab, an anti-FcRH5/CD3 TDB, or anti-HER2/CD3 TDB resulted in the upregulation of the IL-2/15Rβ (CD122) receptor subunit in nearly all CD8+ and majority of CD4+ T cells, suggesting that TDB treatment may sensitize T cells to IL-15. XmAb24306 enhanced T-cell bispecific antibody-induced CD8+ and CD4+ T-cell proliferation and expansion. In vitro combination of XmAb24306 with cevostamab or anti-HER2/CD3 TDB resulted in significant enhancement of tumor cell killing, which was reversed when T-cell numbers were normalized, suggesting that T-cell expansion is the main mechanism of the observed benefit. Pretreatment of immunocompetent mice with a mouse-reactive surrogate of XmAb24306 (mIL-15-Fc) resulted in a significant increase of T cells in the blood, spleen, and tumors and converted transient anti-HER2/CD3 TDB responses to complete durable responses. In summary, our results support the hypothesis that the number of tumor-infiltrating T cells is rate limiting for the activity of solid tumor-targeting TDBs. Upregulation of CD122 by TDB treatment and the observed synergy with XmAb24306 and T-cell bispecific antibodies support clinical evaluation of this novel immunotherapy combination.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140912524","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
Correction: Oleanolic Acid Inhibits Epithelial-Mesenchymal Transition of Hepatocellular Carcinoma by Promoting iNOS Dimerization. 更正:齐墩果酸通过促进 iNOS 二聚化抑制肝细胞癌的上皮-间质转化
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-04 DOI: 10.1158/1535-7163.MCT-24-0565
Hongzhi Wang, Weilong Zhong, Jianmin Zhao, Heng Zhang, Qiang Zhang, Yuan Liang, Shuang Chen, Huijuan Liu, Shumin Zong, Yixuan Tian, Honggang Zhou, Tao Sun, Yanrong Liu, Cheng Yang
{"title":"Correction: Oleanolic Acid Inhibits Epithelial-Mesenchymal Transition of Hepatocellular Carcinoma by Promoting iNOS Dimerization.","authors":"Hongzhi Wang, Weilong Zhong, Jianmin Zhao, Heng Zhang, Qiang Zhang, Yuan Liang, Shuang Chen, Huijuan Liu, Shumin Zong, Yixuan Tian, Honggang Zhou, Tao Sun, Yanrong Liu, Cheng Yang","doi":"10.1158/1535-7163.MCT-24-0565","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0565","url":null,"abstract":"","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142126165","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
Enhancing Neutrophil Cytotoxicity of a Panel of Clinical EGFR Antibodies by Fc Engineering to IgA3.0. 通过 IgA3.0 的 Fc 工程增强一组临床表皮生长因子受体抗体的中性粒细胞毒性。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-04 DOI: 10.1158/1535-7163.MCT-24-0217
Chilam Chan, J H Marco Jansen, Ilona S T Hendriks, Ida C van der Peet, Meggy E L Verdonschot, Elsemieke M Passchier, Maria Tsioumpekou, Maaike Nederend, Sharon A Klomp, Thomas Valerius, Matthias Peipp, Jeanette H W Leusen, Patricia A Olofsen

EGFR plays an essential role in cellular signaling pathways that regulate cell growth, proliferation, and survival and is often dysregulated in cancer. Several monoclonal IgG antibodies have been clinically tested over the years, which exert their function via blocking the ligand binding domain (thereby inhibiting downstream signaling) and inducing Fc-related effector functions, such as antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). However, these IgG antibodies do not optimally recruit neutrophils, which are the most abundant white blood cell population in humans. Therefore, we reformatted six therapeutic EGFR antibodies (cetuximab, panitumumab, nimotuzumab, necitumumab, zalutumumab, and matuzumab) into the IgA3.0 format, which is an IgA2 isotype adapted for clinical application. Reformatting these antibodies preserved Fab-mediated functions such as EGFR binding, growth inhibition, and ligand blockade. In addition, whole leukocyte ADCC was significantly increased when using this panel of IgA3.0 antibodies compared with their respective IgG counterparts, with no major differences between IgA3.0 antibodies. In vivo, IgA3.0 matuzumab outperformed the other antibodies, resulting in the strongest suppression of tumor outgrowth in a long intraperitoneal model. We showed that neutrophils are important for the suppression of tumor outgrowth. IgA3.0 matuzumab exhibited reduced receptor internalization compared with the other antibodies, possibly accounting for its superior in vivo Fc-mediated tumor cell killing efficacy. In conclusion, reformatting EGFR antibodies into an IgA3.0 format increased Fc-mediated killing while retaining Fab-mediated functions and could therefore be a good alternative for the currently available antibody therapies.

表皮生长因子受体(EGFR)在调控细胞生长、增殖和存活的细胞信号通路中发挥着重要作用,在癌症中经常出现失调。多年来,临床试验了多种单克隆 IgG 抗体,它们通过阻断配体结合域(从而抑制下游信号传导)和诱导 Fc 相关效应器功能(如抗体依赖性细胞毒性(ADCC)和抗体依赖性细胞吞噬(ADCP))来发挥其功能。然而,这些 IgG 抗体并不能最佳地募集中性粒细胞,而中性粒细胞是迄今为止人类数量最多的白细胞群。因此,我们将六种治疗性表皮生长因子受体(EGFR)抗体(西妥昔单抗、帕尼单抗、尼妥珠单抗、奈替单抗、扎鲁珠单抗和马妥珠单抗)重新格式化为 IgA3.0 格式,这是一种 IgA2 同工型,已被应用于临床。将这些抗体重新格式化后,它们仍保留了 Fab 介导的功能,如表皮生长因子受体结合、生长抑制和配体阻断。此外,与各自的 IgG 抗体相比,使用这组 IgA3.0 抗体时,整个白细胞的 ADCC 显著增加,而 IgA3.0 抗体之间没有重大差异。在体内,IgA3.0 matuzumab的表现优于其他抗体,在长腹膜模型中对肿瘤生长的抑制作用最强。我们的研究表明,中性粒细胞对抑制肿瘤生长非常重要。与其他抗体相比,IgA3.0 matuzumab的受体内化程度降低,这可能是其体内Fc介导的肿瘤细胞杀伤效力更强的原因。总之,将表皮生长因子受体抗体重新格式化为 IgA3.0 格式可提高 Fc 介导的杀伤力,同时保留 Fab 介导的功能,因此可作为现有抗体疗法的良好替代品。
{"title":"Enhancing Neutrophil Cytotoxicity of a Panel of Clinical EGFR Antibodies by Fc Engineering to IgA3.0.","authors":"Chilam Chan, J H Marco Jansen, Ilona S T Hendriks, Ida C van der Peet, Meggy E L Verdonschot, Elsemieke M Passchier, Maria Tsioumpekou, Maaike Nederend, Sharon A Klomp, Thomas Valerius, Matthias Peipp, Jeanette H W Leusen, Patricia A Olofsen","doi":"10.1158/1535-7163.MCT-24-0217","DOIUrl":"10.1158/1535-7163.MCT-24-0217","url":null,"abstract":"<p><p>EGFR plays an essential role in cellular signaling pathways that regulate cell growth, proliferation, and survival and is often dysregulated in cancer. Several monoclonal IgG antibodies have been clinically tested over the years, which exert their function via blocking the ligand binding domain (thereby inhibiting downstream signaling) and inducing Fc-related effector functions, such as antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). However, these IgG antibodies do not optimally recruit neutrophils, which are the most abundant white blood cell population in humans. Therefore, we reformatted six therapeutic EGFR antibodies (cetuximab, panitumumab, nimotuzumab, necitumumab, zalutumumab, and matuzumab) into the IgA3.0 format, which is an IgA2 isotype adapted for clinical application. Reformatting these antibodies preserved Fab-mediated functions such as EGFR binding, growth inhibition, and ligand blockade. In addition, whole leukocyte ADCC was significantly increased when using this panel of IgA3.0 antibodies compared with their respective IgG counterparts, with no major differences between IgA3.0 antibodies. In vivo, IgA3.0 matuzumab outperformed the other antibodies, resulting in the strongest suppression of tumor outgrowth in a long intraperitoneal model. We showed that neutrophils are important for the suppression of tumor outgrowth. IgA3.0 matuzumab exhibited reduced receptor internalization compared with the other antibodies, possibly accounting for its superior in vivo Fc-mediated tumor cell killing efficacy. In conclusion, reformatting EGFR antibodies into an IgA3.0 format increased Fc-mediated killing while retaining Fab-mediated functions and could therefore be a good alternative for the currently available antibody therapies.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141492665","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
MEK Inhibition Enhances the Antitumor Effect of Radiotherapy in NF1-Deficient Glioblastoma. 抑制MEK可增强放疗对NF1缺陷胶质母细胞瘤的抗肿瘤效果。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-04 DOI: 10.1158/1535-7163.MCT-23-0510
Maria Ioannou, Kriti Lalwani, Abiola A Ayanlaja, Viveka Chinnasamy, Christine A Pratilas, Karisa C Schreck

Individuals with neurofibromatosis type 1, an autosomal dominant neurogenetic and tumor predisposition syndrome, are susceptible to developing low-grade glioma and less commonly high-grade glioma. These gliomas exhibit loss of the neurofibromin gene [neurofibromin type 1 (NF1)], and 10% to 15% of sporadic high-grade gliomas have somatic NF1 alterations. Loss of NF1 leads to hyperactive RAS signaling, creating opportunity given the established efficacy of MEK inhibitors in plexiform neurofibromas and some individuals with low-grade glioma. We observed that NF1-deficient glioblastoma neurospheres were sensitive to the combination of an MEK inhibitor (mirdametinib) with irradiation, as evidenced by synergistic inhibition of cell growth, colony formation, and increased cell death. In contrast, NF1-intact neurospheres were not sensitive to the combination, despite complete ERK pathway inhibition. No neurosphere lines exhibited enhanced sensitivity to temozolomide combined with mirdametinib. Mirdametinib decreased transcription of homologous recombination genes and RAD51 foci, associated with DNA damage repair, in sensitive models. Heterotopic xenograft models displayed synergistic growth inhibition to mirdametinib combined with irradiation in NF1-deficient glioma xenografts but not in those with intact NF1. In sensitive models, benefits were observed at least 3 weeks beyond the completion of treatment, including sustained phosphor-ERK inhibition on immunoblot and decreased Ki-67 expression. These observations demonstrate synergistic activity between mirdametinib and irradiation in NF1-deficient glioma models and may have clinical implications for patients with gliomas that harbor germline or somatic NF1 alterations.

神经纤维瘤病 1 型(NF-1)是一种常染色体显性神经遗传和肿瘤易感综合征,患者易患低级别胶质瘤(LGG),较少见的是高级别胶质瘤(HGG)。这些胶质瘤表现为神经纤维色素基因(NF1)缺失,10%-15%的散发性 HGG 存在体细胞 NF1 改变。NF1 基因缺失会导致 RAS 信号亢进,鉴于 MEK 抑制剂(MEKi)在丛状神经纤维瘤和一些 LGG 患者中已确立的疗效,这为我们创造了机会。我们观察到,NF1缺陷胶质母细胞瘤神经球对MEKi(mirdametinib)与辐照的联合治疗很敏感,这体现在对细胞生长、集落形成和细胞死亡增加的协同抑制上。与此相反,尽管ERK通路完全受到抑制,但NF1-intact神经球对这一组合并不敏感。没有神经球系对替莫唑胺联合米达替尼表现出更高的敏感性。在敏感模型中,米达替尼减少了同源重组基因的转录和与DNA损伤修复相关的RAD51病灶。在异位异种移植模型中,米达替尼与辐照联合治疗对NF1缺陷胶质瘤异种移植的生长有协同抑制作用,但对完整NF1的异种移植没有抑制作用。在敏感模型中,治疗结束后至少三周仍能观察到益处,包括免疫印迹上持续的磷酸-ERK抑制和Ki-67表达的降低。这些观察结果表明,在NF1缺陷胶质瘤模型中,mirdametinib和辐照具有协同活性,可能对携带种系或体细胞NF1改变的胶质瘤患者有临床意义。
{"title":"MEK Inhibition Enhances the Antitumor Effect of Radiotherapy in NF1-Deficient Glioblastoma.","authors":"Maria Ioannou, Kriti Lalwani, Abiola A Ayanlaja, Viveka Chinnasamy, Christine A Pratilas, Karisa C Schreck","doi":"10.1158/1535-7163.MCT-23-0510","DOIUrl":"10.1158/1535-7163.MCT-23-0510","url":null,"abstract":"<p><p>Individuals with neurofibromatosis type 1, an autosomal dominant neurogenetic and tumor predisposition syndrome, are susceptible to developing low-grade glioma and less commonly high-grade glioma. These gliomas exhibit loss of the neurofibromin gene [neurofibromin type 1 (NF1)], and 10% to 15% of sporadic high-grade gliomas have somatic NF1 alterations. Loss of NF1 leads to hyperactive RAS signaling, creating opportunity given the established efficacy of MEK inhibitors in plexiform neurofibromas and some individuals with low-grade glioma. We observed that NF1-deficient glioblastoma neurospheres were sensitive to the combination of an MEK inhibitor (mirdametinib) with irradiation, as evidenced by synergistic inhibition of cell growth, colony formation, and increased cell death. In contrast, NF1-intact neurospheres were not sensitive to the combination, despite complete ERK pathway inhibition. No neurosphere lines exhibited enhanced sensitivity to temozolomide combined with mirdametinib. Mirdametinib decreased transcription of homologous recombination genes and RAD51 foci, associated with DNA damage repair, in sensitive models. Heterotopic xenograft models displayed synergistic growth inhibition to mirdametinib combined with irradiation in NF1-deficient glioma xenografts but not in those with intact NF1. In sensitive models, benefits were observed at least 3 weeks beyond the completion of treatment, including sustained phosphor-ERK inhibition on immunoblot and decreased Ki-67 expression. These observations demonstrate synergistic activity between mirdametinib and irradiation in NF1-deficient glioma models and may have clinical implications for patients with gliomas that harbor germline or somatic NF1 alterations.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11374499/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140876835","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
Combination Immunotherapy with Vaccine and Oncolytic HSV Virotherapy Is Time Dependent. 疫苗与溶解性 HSV 病毒疗法的联合免疫疗法取决于时间。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-04 DOI: 10.1158/1535-7163.MCT-23-0873
Stacie K Totsch, Andrew S Ishizuka, Kyung-Don Kang, Sam E Gary, Abbey Rocco, Aaron E Fan, Li Zhou, Pablo A Valdes, SeungHo Lee, Jason Li, Luca Peruzzotti-Jametti, Sarah Blitz, Christopher M Garliss, James M Johnston, James M Markert, Geoffrey M Lynn, Joshua D Bernstock, Gregory K Friedman

Oncolytic virotherapy or immunovirotherapy is a strategy that utilizes viruses to selectively infect and kill tumor cells while also stimulating an immune response against the tumor. Early clinical trials in both pediatric and adult patients using oncolytic herpes simplex viruses (oHSV) have demonstrated safety and promising efficacy; however, combinatorial strategies designed to enhance oncolysis while also promoting durable T-cell responses for sustaining disease remission are likely required. We hypothesized that combining the direct tumor cell killing and innate immune stimulation by oHSV with a vaccine that promotes T cell-mediated immunity may lead to more durable tumor regression. To this end, we investigated the preclinical efficacy and potential synergy of combining oHSV with a self-assembling nanoparticle vaccine codelivering peptide antigens and Toll-like receptor 7 and 8 agonists (referred to as SNAPvax),which induces robust tumor-specific T-cell immunity. We then assessed how timing of the treatments (i.e., vaccine before or after oHSV) impacts T-cell responses, viral replication, and preclinical efficacy. The sequence of treatments was critical, as survival was significantly enhanced when the SNAPvax vaccine was given prior to oHSV. Increased clinical efficacy was associated with reduced tumor volume and increases in virus replication and tumor antigen-specific CD8+ T cells. These findings substantiate the criticality of combination immunotherapy timing and provide preclinical support for combining SNAPvax with oHSV as a promising treatment approach for both pediatric and adult tumors.

目的:溶瘤病毒疗法或免疫疗法是一种利用病毒有选择性地感染和杀死肿瘤细胞,同时激发针对肿瘤的免疫反应的策略。使用溶瘤性单纯疱疹病毒(oHSVs)对儿童和成人患者进行的早期临床试验已经证明了其安全性和良好的疗效;然而,可能需要采取组合策略,在增强溶瘤作用的同时促进持久的 T 细胞反应,以维持疾病缓解。我们假设,将 oHSV 对肿瘤细胞的直接杀伤和先天性免疫刺激与促进 T 细胞介导免疫的疫苗相结合,可能会导致更持久的肿瘤消退:为此,我们研究了将 oHSV 与共同递送多肽抗原和 Toll 样受体-7 和-8 激动剂(TLR-7/8a)的自组装纳米粒子疫苗(简称 SNAPvax™)相结合的临床前疗效和潜在协同作用,该疫苗可诱导强大的肿瘤特异性 T 细胞免疫。然后,我们评估了治疗时机(即在 oHSV 之前或之后接种疫苗)对 T 细胞反应、病毒复制和临床前疗效的影响:结果:治疗的顺序至关重要,因为在oHSV之前接种SNAPvax™疫苗可显著提高存活率。临床疗效的提高与肿瘤体积缩小、病毒复制和肿瘤抗原特异性 CD8+ T 细胞增加有关:这些研究结果证明了联合免疫疗法时机的重要性,并为将 SNAPvax 与 oHSV 作为一种治疗儿童和成人肿瘤的有效方法提供了临床前支持。
{"title":"Combination Immunotherapy with Vaccine and Oncolytic HSV Virotherapy Is Time Dependent.","authors":"Stacie K Totsch, Andrew S Ishizuka, Kyung-Don Kang, Sam E Gary, Abbey Rocco, Aaron E Fan, Li Zhou, Pablo A Valdes, SeungHo Lee, Jason Li, Luca Peruzzotti-Jametti, Sarah Blitz, Christopher M Garliss, James M Johnston, James M Markert, Geoffrey M Lynn, Joshua D Bernstock, Gregory K Friedman","doi":"10.1158/1535-7163.MCT-23-0873","DOIUrl":"10.1158/1535-7163.MCT-23-0873","url":null,"abstract":"<p><p>Oncolytic virotherapy or immunovirotherapy is a strategy that utilizes viruses to selectively infect and kill tumor cells while also stimulating an immune response against the tumor. Early clinical trials in both pediatric and adult patients using oncolytic herpes simplex viruses (oHSV) have demonstrated safety and promising efficacy; however, combinatorial strategies designed to enhance oncolysis while also promoting durable T-cell responses for sustaining disease remission are likely required. We hypothesized that combining the direct tumor cell killing and innate immune stimulation by oHSV with a vaccine that promotes T cell-mediated immunity may lead to more durable tumor regression. To this end, we investigated the preclinical efficacy and potential synergy of combining oHSV with a self-assembling nanoparticle vaccine codelivering peptide antigens and Toll-like receptor 7 and 8 agonists (referred to as SNAPvax),which induces robust tumor-specific T-cell immunity. We then assessed how timing of the treatments (i.e., vaccine before or after oHSV) impacts T-cell responses, viral replication, and preclinical efficacy. The sequence of treatments was critical, as survival was significantly enhanced when the SNAPvax vaccine was given prior to oHSV. Increased clinical efficacy was associated with reduced tumor volume and increases in virus replication and tumor antigen-specific CD8+ T cells. These findings substantiate the criticality of combination immunotherapy timing and provide preclinical support for combining SNAPvax with oHSV as a promising treatment approach for both pediatric and adult tumors.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11374504/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140867304","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
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:481959085
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