首页 > 最新文献

Molecular Cancer Therapeutics最新文献

英文 中文
CRISPR Screen Identifies HDAC3 as a Novel Radiosensitizing Target in Small Cell Lung Cancer. CRISPR筛选确定HDAC3为小细胞肺癌新的放射增敏靶点
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-02 DOI: 10.1158/1535-7163.MCT-24-0861
Ujas A Patel, Mary Y Shi, Jalal M Kazan, Kevin C J Nixon, Xiaozhuo Ran, Sree N Nair, Olivia Huang, Lifang Song, Mansi K Aparnathi, Michael Y He, Mehran Bakhtiari, Rehna Krishnan, Razan K Hessenow, Vivek Philip, Troy Ketela, Verena Jendrossek, Razqallah Hakem, Housheng H He, Robert Kridel, Benjamin H Lok

Small cell lung cancer (SCLC) is an aggressive malignancy, with most patients presenting with prognostically poor extensive-stage disease. Limited progress in standard care stresses the urgent need for novel therapies. Radiotherapy offers some survival benefit for selected patients with SCLC but could be enhanced with radiosensitizers. In this study, we identify HDAC3 as a novel radiosensitizing target in SCLC using a CRISPR knockout screen and demonstrate its efficacy and mechanism. SBC5 cells were transduced with a custom EpiDrug single-guide RNA library and treated with ionizing radiation (IR) to identify radiosensitizing genes. HDAC3 emerged as a candidate and was validated through genetic knockdown and pharmacologic inhibition (RGFP966) in multiple SCLC cell lines. Both approaches enhanced radiosensitivity, as shown by cell viability (dose modification factor10 = 1.14-1.69) and clonogenic assays (dose modification factor10 = 1.16-1.41). We assessed changes in chromatin accessibility by assay for transposase-accessible chromatin using sequencing and IR-induced DNA damage and repair using γH2AX foci detection, double-strand break (DSB) repair assays, and immunoblotting of repair proteins. HDAC3-deficient cells exhibited increased chromatin accessibility, greater IR-induced DSBs, and impaired repair capacity, resulting in persistent DNA damage. This repair defect sensitized cells to PARP inhibitors, for which combining RGFP966 with olaparib or talazoparib produced additive to synergistic effects. In SCLC xenograft models, HDAC3 knockdown or RGFP966, combined with IR, achieved significant tumor growth inhibition. Collectively, we identified HDAC3 as a novel radiosensitizing target in SCLC. Its functional loss increased the generation and persistence of IR-induced DNA DSBs, effectively sensitizing SCLC cell lines and xenografts to IR, providing a potential radiosensitization strategy to treat SCLC.

小细胞肺癌(SCLC)是一种侵袭性恶性肿瘤,大多数患者表现为预后不良的广泛期疾病。标准治疗的有限进展强调了对新疗法的迫切需要。放射治疗为部分SCLC患者提供了一定的生存益处,但放射增敏剂可以增强。在这里,我们通过CRISPR敲除筛选确定了HDAC3作为SCLC中一种新的放射增敏靶点,并证明了其疗效和机制。用EpiDrug定制的sgRNA文库对SBC5细胞进行转导,并用电离辐射(IR)处理以鉴定放射致敏基因。HDAC3作为候选药物出现,并通过基因敲低(KD)和药理抑制(RGFP966)在多种SCLC细胞系中得到验证。细胞活力(剂量修饰因子[DMF]10 = 1.14-1.69)和克隆性测定(DMF10 = 1.16-1.41)表明,这两种方法都增强了放射敏感性。我们通过ATAC-seq评估了染色质可及性的变化,并使用γ - h2ax焦点检测、DSB修复试验和修复蛋白的免疫印迹技术评估了ir诱导的DNA损伤和修复。hdac3缺陷细胞表现出染色质可及性增加,ir诱导的dsb更大,修复能力受损,导致持续的DNA损伤。这种修复缺陷使细胞对PARP抑制剂敏感,其中RGFP966与奥拉帕尼或塔拉唑帕尼联合使用可产生增效效应。在SCLC异种移植模型中,HDAC3 KD或RGFP966联合IR,实现了显著的肿瘤生长抑制。总之,我们确定HDAC3是SCLC中一种新的放射增敏靶点。它的功能缺失增加了红外诱导的DNA dsb的产生和持久性,有效地使SCLC细胞系和异种移植物对红外敏感,为治疗SCLC提供了一种潜在的放射增敏策略。
{"title":"CRISPR Screen Identifies HDAC3 as a Novel Radiosensitizing Target in Small Cell Lung Cancer.","authors":"Ujas A Patel, Mary Y Shi, Jalal M Kazan, Kevin C J Nixon, Xiaozhuo Ran, Sree N Nair, Olivia Huang, Lifang Song, Mansi K Aparnathi, Michael Y He, Mehran Bakhtiari, Rehna Krishnan, Razan K Hessenow, Vivek Philip, Troy Ketela, Verena Jendrossek, Razqallah Hakem, Housheng H He, Robert Kridel, Benjamin H Lok","doi":"10.1158/1535-7163.MCT-24-0861","DOIUrl":"10.1158/1535-7163.MCT-24-0861","url":null,"abstract":"<p><p>Small cell lung cancer (SCLC) is an aggressive malignancy, with most patients presenting with prognostically poor extensive-stage disease. Limited progress in standard care stresses the urgent need for novel therapies. Radiotherapy offers some survival benefit for selected patients with SCLC but could be enhanced with radiosensitizers. In this study, we identify HDAC3 as a novel radiosensitizing target in SCLC using a CRISPR knockout screen and demonstrate its efficacy and mechanism. SBC5 cells were transduced with a custom EpiDrug single-guide RNA library and treated with ionizing radiation (IR) to identify radiosensitizing genes. HDAC3 emerged as a candidate and was validated through genetic knockdown and pharmacologic inhibition (RGFP966) in multiple SCLC cell lines. Both approaches enhanced radiosensitivity, as shown by cell viability (dose modification factor10 = 1.14-1.69) and clonogenic assays (dose modification factor10 = 1.16-1.41). We assessed changes in chromatin accessibility by assay for transposase-accessible chromatin using sequencing and IR-induced DNA damage and repair using γH2AX foci detection, double-strand break (DSB) repair assays, and immunoblotting of repair proteins. HDAC3-deficient cells exhibited increased chromatin accessibility, greater IR-induced DSBs, and impaired repair capacity, resulting in persistent DNA damage. This repair defect sensitized cells to PARP inhibitors, for which combining RGFP966 with olaparib or talazoparib produced additive to synergistic effects. In SCLC xenograft models, HDAC3 knockdown or RGFP966, combined with IR, achieved significant tumor growth inhibition. Collectively, we identified HDAC3 as a novel radiosensitizing target in SCLC. Its functional loss increased the generation and persistence of IR-induced DNA DSBs, effectively sensitizing SCLC cell lines and xenografts to IR, providing a potential radiosensitization strategy to treat SCLC.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"183-195"},"PeriodicalIF":5.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12493205/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144961727","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
A Novel Anti-CD47 Antibody TJH2201: Efficacious Tumor Suppression with Reduced RBC Toxicity via a SIRPα-Independent Mechanism. 一种新型抗cd47抗体TJH2201:通过sirp α-不依赖机制有效抑制肿瘤并降低红细胞毒性
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-02 DOI: 10.1158/1535-7163.MCT-25-0425
Huan Zhang, Fufan He, Lei Cao, Haiqing Ni, Ninghuan Li, Yang Liu, Min Wu, Ya Liu, Bing Wu, Li Li, ZhiHai Wu, Xiaomin Ling, Shuaixiang Zhou, Yiming Li, Shuxuan Deng, Weiwei Wu, Qian Chu

The CD47/SIRPα axis serves as a "do not eat me" signal, protecting normal cells from phagocytosis, but in the meantime, enabling immune evasion by tumor cells. Whereas substantial progress has been made in developing CD47 antagonists, achieving a balance between hematotoxicity and antitumor efficacy remains a critical challenge. In this study, we demonstrated that the bivalent anti-CD47 antibody, Hu5F9, caused severe anemia in both human CD47 knock-in mice lacking the CD47/SIRPα signal and human CD47/SIRPα double knock-in mice with complete signal, suggesting the CD47/SIRPα signal is not essential. Moreover, the single-arm CD47 antibody Hu5F9/gp120 exhibited only mild red blood cell (RBC) destruction in vitro and in vivo. These findings reveal that RBC toxicity induced by anti-CD47 antibodies is determined by the bivalency of the antibody rather than the CD47/SIRPα signal engagement. Based on this, we engineered TJH2201, a novel anti-CD47 antibody that avoids RBC agglutination while retaining high-affinity CD47 binding, robust signaling blockade, and enhanced pro-phagocytosis activity in vitro. In xenograft models with Raji and MV-4-11 cells, TJH2201 demonstrated potent antitumor activity without inducing body weight loss. These results suggest that TJH2201 is a promising CD47 antagonist that balances antitumor efficacy and hematologic safety, providing a new therapeutic approach for CD47-expressing malignancies.

CD47/SIRPα轴作为一个“不要吃我”的信号,保护正常细胞免受吞噬,但同时,使肿瘤细胞免疫逃避。虽然CD47拮抗剂的开发已经取得了实质性进展,但在血液毒性和抗肿瘤疗效之间取得平衡仍然是一个关键的挑战。在这里,我们证明了二价抗CD47抗体Hu5F9在缺乏CD47/SIRPα信号的人CD47敲入小鼠和具有完整CD47/SIRPα信号的人CD47/SIRPα双敲入小鼠中引起严重贫血,这表明CD47/SIRPα信号不是必需的。此外,单臂CD47抗体Hu5F9/gp120在体外和体内仅表现出轻微的红细胞破坏。这些发现表明,抗CD47抗体诱导的红细胞毒性是由抗体的二价决定的,而不是由CD47/SIRPα信号结合决定的。基于此,我们设计了一种新的抗CD47抗体TJH2201,它可以避免红细胞凝集,同时保持高亲和力的CD47结合,强大的信号阻断,并增强体外吞噬活性。在Raji和MV-4-11细胞的异种移植模型中,TJH2201显示出有效的抗肿瘤活性,而不引起体重减轻。这些结果表明,TJH2201是一种很有前景的CD47拮抗剂,能够平衡抗肿瘤疗效和血液学安全性,为CD47表达的恶性肿瘤提供新的治疗途径。
{"title":"A Novel Anti-CD47 Antibody TJH2201: Efficacious Tumor Suppression with Reduced RBC Toxicity via a SIRPα-Independent Mechanism.","authors":"Huan Zhang, Fufan He, Lei Cao, Haiqing Ni, Ninghuan Li, Yang Liu, Min Wu, Ya Liu, Bing Wu, Li Li, ZhiHai Wu, Xiaomin Ling, Shuaixiang Zhou, Yiming Li, Shuxuan Deng, Weiwei Wu, Qian Chu","doi":"10.1158/1535-7163.MCT-25-0425","DOIUrl":"10.1158/1535-7163.MCT-25-0425","url":null,"abstract":"<p><p>The CD47/SIRPα axis serves as a \"do not eat me\" signal, protecting normal cells from phagocytosis, but in the meantime, enabling immune evasion by tumor cells. Whereas substantial progress has been made in developing CD47 antagonists, achieving a balance between hematotoxicity and antitumor efficacy remains a critical challenge. In this study, we demonstrated that the bivalent anti-CD47 antibody, Hu5F9, caused severe anemia in both human CD47 knock-in mice lacking the CD47/SIRPα signal and human CD47/SIRPα double knock-in mice with complete signal, suggesting the CD47/SIRPα signal is not essential. Moreover, the single-arm CD47 antibody Hu5F9/gp120 exhibited only mild red blood cell (RBC) destruction in vitro and in vivo. These findings reveal that RBC toxicity induced by anti-CD47 antibodies is determined by the bivalency of the antibody rather than the CD47/SIRPα signal engagement. Based on this, we engineered TJH2201, a novel anti-CD47 antibody that avoids RBC agglutination while retaining high-affinity CD47 binding, robust signaling blockade, and enhanced pro-phagocytosis activity in vitro. In xenograft models with Raji and MV-4-11 cells, TJH2201 demonstrated potent antitumor activity without inducing body weight loss. These results suggest that TJH2201 is a promising CD47 antagonist that balances antitumor efficacy and hematologic safety, providing a new therapeutic approach for CD47-expressing malignancies.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"21-33"},"PeriodicalIF":5.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145192107","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 SUMOylation Triggers IFN-β-dependent Activation of Patient and Allogenic NK Cells in Preclinical Models of Acute Myeloid Leukemia. 在急性髓性白血病临床前模型中,靶向SUMOylation触发患者和同种异体自然杀伤细胞的干扰素ß依赖激活。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-02 DOI: 10.1158/1535-7163.MCT-25-0504
Rawan Hallal, Marion de Toledo, Denis Tempé, Rayane Berrahouane, Sara Zemiti, Loïs Coënon, Delphine Gitenay, Simon George, Moritz Schüssler, Nadine Laguette, Sarah Bonnet, Ludovic Gabellier, Guillaume Cartron, Mireia Pelegrin, Martin Villalba, Guillaume Bossis

NK cells can play a significant role in the antitumoral immune response. In patients with acute myeloid leukemia (AML), NK cells are, however, often found in low numbers and exhibit poor activity, contributing to leukemic progression. Allogenic NK cells are emerging as promising cellular therapies for hematologic cancer treatment. New strategies are however required to both reactivate NK cells in patients with AML and enhance the antitumor activity of transplanted NK cells. In this study, we demonstrate that targeting SUMOylation, a protein posttranslational modification, activates NK cells from both healthy donors and patients with AML. Subasumstat (TAK-981), a first-in-class inhibitor of SUMOylation used in phase I/II clinical trials, enhances NK cell degranulation, secretion of inflammatory cytokines (IFN-γ, TNF-α, and FasL), and cytotoxicity against AML cells. In vivo, TAK-981 improves the anti-leukemic efficacy of ex vivo expanded cord blood NK cells in leukemia-bearing mice. One early effect of TAK-981 is to specifically increase the accessibility and activation of cis-regulatory regions of IFN-I pathway genes and induce their transcription. TAK-981-induced secretion of IFN-β, mostly by NK cells and myeloid cells, is required for NK cell activation. Surprisingly, IFNB1 induction does not require its best-characterized activators MDA5, cGas, and IFN response factor-1, -3, and -7. Altogether, this suggests that targeting SUMOylation activates a noncanonical IFN-I pathway, which enhances the anti-leukemic potential of NK cells.

自然杀伤细胞(NK)在抗肿瘤免疫应答中发挥重要作用。然而,在急性髓性白血病(AML)患者中,NK细胞数量少,活性差,导致白血病进展。同种异体NK细胞正在成为血液肿瘤治疗的有前途的细胞疗法。然而,需要新的策略来重新激活AML患者的NK细胞并增强移植NK细胞的抗肿瘤活性。在这里,我们证明靶向SUMOylation(一种蛋白质翻译后修饰)可以激活来自健康供体和AML患者的NK细胞。subbasumstat (TAK-981)是一种用于I/II期临床试验的同类首个SUMOylation抑制剂,可增强NK细胞脱颗粒、炎性细胞因子(IFN-γ、TNF-α、FasL)的分泌和对AML细胞的细胞毒性。在体内,TAK-981可提高白血病小鼠体外扩增脐带血NK细胞的抗白血病功效。TAK-981的一个早期作用是特异性地增加I型干扰素(IFN-I)途径基因的顺式调控区域的可及性和激活,并诱导其转录。tak -981诱导的干扰素-ß分泌,主要由NK细胞和髓系细胞分泌,是NK细胞激活所必需的。令人惊讶的是,IFNB1诱导不需要其最具特征的激活剂MDA5、cGas、IRF-1、-3和-7。总之,这表明靶向SUMOylation激活非规范IFN-I通路,从而增强NK细胞的抗白血病潜能。
{"title":"Targeting SUMOylation Triggers IFN-β-dependent Activation of Patient and Allogenic NK Cells in Preclinical Models of Acute Myeloid Leukemia.","authors":"Rawan Hallal, Marion de Toledo, Denis Tempé, Rayane Berrahouane, Sara Zemiti, Loïs Coënon, Delphine Gitenay, Simon George, Moritz Schüssler, Nadine Laguette, Sarah Bonnet, Ludovic Gabellier, Guillaume Cartron, Mireia Pelegrin, Martin Villalba, Guillaume Bossis","doi":"10.1158/1535-7163.MCT-25-0504","DOIUrl":"10.1158/1535-7163.MCT-25-0504","url":null,"abstract":"<p><p>NK cells can play a significant role in the antitumoral immune response. In patients with acute myeloid leukemia (AML), NK cells are, however, often found in low numbers and exhibit poor activity, contributing to leukemic progression. Allogenic NK cells are emerging as promising cellular therapies for hematologic cancer treatment. New strategies are however required to both reactivate NK cells in patients with AML and enhance the antitumor activity of transplanted NK cells. In this study, we demonstrate that targeting SUMOylation, a protein posttranslational modification, activates NK cells from both healthy donors and patients with AML. Subasumstat (TAK-981), a first-in-class inhibitor of SUMOylation used in phase I/II clinical trials, enhances NK cell degranulation, secretion of inflammatory cytokines (IFN-γ, TNF-α, and FasL), and cytotoxicity against AML cells. In vivo, TAK-981 improves the anti-leukemic efficacy of ex vivo expanded cord blood NK cells in leukemia-bearing mice. One early effect of TAK-981 is to specifically increase the accessibility and activation of cis-regulatory regions of IFN-I pathway genes and induce their transcription. TAK-981-induced secretion of IFN-β, mostly by NK cells and myeloid cells, is required for NK cell activation. Surprisingly, IFNB1 induction does not require its best-characterized activators MDA5, cGas, and IFN response factor-1, -3, and -7. Altogether, this suggests that targeting SUMOylation activates a noncanonical IFN-I pathway, which enhances the anti-leukemic potential of NK cells.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"125-139"},"PeriodicalIF":5.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7618005/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144637671","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
Advancing Preclinical Biology for Ewing Sarcoma: An International Effort. 推进尤因肉瘤的临床前生物学:一项国际努力。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-02 DOI: 10.1158/1535-7163.MCT-25-0428
Filemon S Dela Cruz, Elizabeth A Stewart, Didier Surdez, Jessica D Daley, Alice Soragni, Eleni M Tomazou, Jaime Alvarez-Perez, Tamar Y Feinberg, James F Amatruda, Shireen S Ganapathi, Joyce E Ohm, Christine M Heske, Sarah Cohen-Gogo, Dusan Pesic, Joshua O Nash, Adam Shlien, Elizabeth A Roundhill, Susan A Burchill, Brian D Crompton, Elizabeth R Lawlor, David M Loeb, Olivier Delattre, Jaume Mora, Katia Scotlandi, Damon R Reed, Patrick J Grohar, Thomas G P Grünewald, Heinrich Kovar, Kelly M Bailey

Ewing sarcoma is an aggressive bone and soft-tissue cancer affecting adolescents and young adults. In vitro and in vivo models of Ewing sarcoma have been instrumental in advancing our understanding of Ewing sarcoma biology and essential in evaluating potential therapies, particularly for metastatic or relapsed disease for which effective treatment options remain limited. Through an international collaborative effort between the Children's Oncology Group Bone Tumor Committee and the Euro Ewing Consortium, we review the current landscape of preclinical modeling used in Ewing sarcoma research encompassing both in vitro (cell lines and tumor organoids) and in vivo (mouse and nonmammalian xenografts) model systems. We discuss factors that can influence experimental results, provide testing considerations for both in vitro and in vivo studies, and descriptions of existing preclinical data repositories. We highlight current needs in Ewing sarcoma modeling and the importance of enhanced international cooperative research and patient advocacy efforts which will be critical in expanding our resources of biologically relevant Ewing sarcoma models to enable translation of preclinical findings into effective therapeutic strategies for patients with Ewing sarcoma.

尤文氏肉瘤(EwS)是一种侵袭性骨和软组织癌症,影响青少年和年轻人。体外和体内EwS模型有助于提高我们对EwS生物学的理解,对于评估潜在的治疗方法至关重要,特别是对于有效治疗选择仍然有限的转移性或复发性疾病。通过儿童肿瘤学组(COG)骨肿瘤委员会和欧洲尤因联盟(EEC)之间的国际合作,我们回顾了目前用于EwS研究的临床前建模的现状,包括体外(细胞系和肿瘤类器官)和体内(小鼠和非哺乳动物异种移植)模型系统。我们讨论了可能影响实验结果的因素,提供了体外和体内研究的测试考虑因素,并描述了现有的临床前数据库。我们强调了EwS建模的当前需求,以及加强国际合作研究和患者倡导努力的重要性,这对于扩大我们的生物相关EwS模型资源至关重要,从而能够将临床前研究结果转化为EwS患者的有效治疗策略。
{"title":"Advancing Preclinical Biology for Ewing Sarcoma: An International Effort.","authors":"Filemon S Dela Cruz, Elizabeth A Stewart, Didier Surdez, Jessica D Daley, Alice Soragni, Eleni M Tomazou, Jaime Alvarez-Perez, Tamar Y Feinberg, James F Amatruda, Shireen S Ganapathi, Joyce E Ohm, Christine M Heske, Sarah Cohen-Gogo, Dusan Pesic, Joshua O Nash, Adam Shlien, Elizabeth A Roundhill, Susan A Burchill, Brian D Crompton, Elizabeth R Lawlor, David M Loeb, Olivier Delattre, Jaume Mora, Katia Scotlandi, Damon R Reed, Patrick J Grohar, Thomas G P Grünewald, Heinrich Kovar, Kelly M Bailey","doi":"10.1158/1535-7163.MCT-25-0428","DOIUrl":"10.1158/1535-7163.MCT-25-0428","url":null,"abstract":"<p><p>Ewing sarcoma is an aggressive bone and soft-tissue cancer affecting adolescents and young adults. In vitro and in vivo models of Ewing sarcoma have been instrumental in advancing our understanding of Ewing sarcoma biology and essential in evaluating potential therapies, particularly for metastatic or relapsed disease for which effective treatment options remain limited. Through an international collaborative effort between the Children's Oncology Group Bone Tumor Committee and the Euro Ewing Consortium, we review the current landscape of preclinical modeling used in Ewing sarcoma research encompassing both in vitro (cell lines and tumor organoids) and in vivo (mouse and nonmammalian xenografts) model systems. We discuss factors that can influence experimental results, provide testing considerations for both in vitro and in vivo studies, and descriptions of existing preclinical data repositories. We highlight current needs in Ewing sarcoma modeling and the importance of enhanced international cooperative research and patient advocacy efforts which will be critical in expanding our resources of biologically relevant Ewing sarcoma models to enable translation of preclinical findings into effective therapeutic strategies for patients with Ewing sarcoma.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"48-70"},"PeriodicalIF":5.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12757721/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145006328","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
Antitumor Activity of Trastuzumab Deruxtecan in Pediatric Solid Tumors with Variable HER2 Expression. 曲妥珠单抗deruxtecan在可变HER2表达的儿童实体瘤中的抗肿瘤活性
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-02 DOI: 10.1158/1535-7163.MCT-25-0437
Chelsey M Burke, Tamar Y Feinberg, Samantha Brosius, Umeshkumar K Bhanot, Mala Jain, Irina Linkov, Armaan Siddiquee, Kristina Guillan, Glorymar Ibáñez, Andoyo A Ndengu, Paul Calder, Nestor Rosales, Diego F Coutinho, Matthew M Long, Raina Fishkin, Sheeno Thyparambil, Julia L Glade Bender, Damon R Reed, Daoqi You, Michael V Ortiz, Emily K Slotkin, Andrew L Kung, Filemon S Dela Cruz

Trastuzumab deruxtecan (T-DXd) is an ERBB2/HER2-targeting antibody-drug conjugate (ADC) with efficacy across adult cancers exhibiting variable HER2 expression. Prior studies demonstrating HER2 expression in osteosarcoma motivated a clinical trial of T-DXd in pediatric and adolescent/young adults with osteosarcoma, but the trial was terminated early because of inactivity. We evaluated the activity of T-DXd using osteosarcoma patient-derived xenograft (PDX) models and found a 22% objective response rate despite no detectable HER2 expression across PDXs tested. To further assess non-HER2-mediated activity, we evaluated the activity of T-DXd across 31 pediatric cancer cell lines and found osteosarcoma to be amongst the most resistant to T-DXd, as well as unconjugated deruxtecan, providing a potential explanation for the negative results observed in the clinical trial of T-DXd in osteosarcoma. T-DXd evaluation in PDX models representing pediatric histologies with greater intrinsic sensitivity to deruxtecan, including pediatric renal tumors and desmoplastic small round cell tumor, revealed both HER2-enhanced activity as well as substantial non-HER2-mediated activity, as evidenced by equipotent activity using an isotype-matched control ADC. Together, these results underscore translational opportunities for ADC therapeutics in tumor histologies with high sensitivity to the payload and in which enhanced tumor delivery may be mediated by antibody-targeted mechanisms as well as macromolecular characteristics of ADCs (e.g., enhanced permeability and retention effect) and tumor microenvironmental factors (e.g., proteolytic payload release). Our findings challenge the role of HER2 as a biomarker predictive of T-DXd response in pediatric cancers and support further biomarker-agnostic clinical development of T-DXd in desmoplastic small round cell tumor and pediatric renal tumors.

曲妥珠单抗德uxtecan (T-DXd)是一种靶向HER2的抗体-药物偶联物(ADC),对具有可变HER2表达的成人癌症有效。先前的研究表明,HER2在骨肉瘤(OS)中表达,促使T-DXd在患有骨肉瘤的儿童和青少年/年轻成人中进行临床试验,但由于缺乏活动而提前终止。我们使用OS患者来源的异种移植(PDX)模型评估T-DXd的活性,发现尽管在测试的PDX中没有检测到HER2表达,但客观有效率为22%。为了进一步评估非her2介导的活性,我们评估了T-DXd在31个儿科癌细胞系中的活性,发现OS是对T-DXd和未结合的德鲁替康最耐药的细胞之一,这为T-DXd在OS临床试验中观察到的阴性结果提供了可能的解释。在PDX模型中,T-DXd评估代表了对德鲁替康具有更大内在敏感性的儿童组织学,包括儿童肾肿瘤和促结性小圆细胞肿瘤(DSRCT),显示her2增强的活性,以及大量非her2介导的活性,如使用同型匹配对照ADC的等效活性所证明的那样。总之,这些结果强调了ADC治疗在对有效载荷高度敏感的肿瘤组织学中的转化机会,其中增强的肿瘤递送可能由抗体靶向机制、ADC的大分子特性(例如,增强的渗透性和保留效应)和肿瘤微环境因素(例如,蛋白水解有效载荷释放)介导。我们的研究结果挑战了HER2作为预测儿童癌症T-DXd反应的生物标志物的作用,并支持T-DXd在DSRCT和儿童肾肿瘤中进一步的生物标志物不可知的临床发展。
{"title":"Antitumor Activity of Trastuzumab Deruxtecan in Pediatric Solid Tumors with Variable HER2 Expression.","authors":"Chelsey M Burke, Tamar Y Feinberg, Samantha Brosius, Umeshkumar K Bhanot, Mala Jain, Irina Linkov, Armaan Siddiquee, Kristina Guillan, Glorymar Ibáñez, Andoyo A Ndengu, Paul Calder, Nestor Rosales, Diego F Coutinho, Matthew M Long, Raina Fishkin, Sheeno Thyparambil, Julia L Glade Bender, Damon R Reed, Daoqi You, Michael V Ortiz, Emily K Slotkin, Andrew L Kung, Filemon S Dela Cruz","doi":"10.1158/1535-7163.MCT-25-0437","DOIUrl":"10.1158/1535-7163.MCT-25-0437","url":null,"abstract":"<p><p>Trastuzumab deruxtecan (T-DXd) is an ERBB2/HER2-targeting antibody-drug conjugate (ADC) with efficacy across adult cancers exhibiting variable HER2 expression. Prior studies demonstrating HER2 expression in osteosarcoma motivated a clinical trial of T-DXd in pediatric and adolescent/young adults with osteosarcoma, but the trial was terminated early because of inactivity. We evaluated the activity of T-DXd using osteosarcoma patient-derived xenograft (PDX) models and found a 22% objective response rate despite no detectable HER2 expression across PDXs tested. To further assess non-HER2-mediated activity, we evaluated the activity of T-DXd across 31 pediatric cancer cell lines and found osteosarcoma to be amongst the most resistant to T-DXd, as well as unconjugated deruxtecan, providing a potential explanation for the negative results observed in the clinical trial of T-DXd in osteosarcoma. T-DXd evaluation in PDX models representing pediatric histologies with greater intrinsic sensitivity to deruxtecan, including pediatric renal tumors and desmoplastic small round cell tumor, revealed both HER2-enhanced activity as well as substantial non-HER2-mediated activity, as evidenced by equipotent activity using an isotype-matched control ADC. Together, these results underscore translational opportunities for ADC therapeutics in tumor histologies with high sensitivity to the payload and in which enhanced tumor delivery may be mediated by antibody-targeted mechanisms as well as macromolecular characteristics of ADCs (e.g., enhanced permeability and retention effect) and tumor microenvironmental factors (e.g., proteolytic payload release). Our findings challenge the role of HER2 as a biomarker predictive of T-DXd response in pediatric cancers and support further biomarker-agnostic clinical development of T-DXd in desmoplastic small round cell tumor and pediatric renal tumors.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"156-167"},"PeriodicalIF":5.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12490493/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145006385","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Recycling Collagen Receptor uPARAP Is a Unique Mediator of Stromal Drug Delivery to Carcinoma Cells. 再生胶原受体uPARAP是一个独特的介质间质药物输送到癌细胞。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-02 DOI: 10.1158/1535-7163.MCT-25-0051
Kirstine S Nørregaard, Ida M E Larsen, Henrik J Jürgensen, Michaela Hansen Blomquist, Pinar Çakilkaya, Virginia Metrangolo, Alba Martinez Perlado, Oliver Krigslund, Henrik Gårdsvoll, Thomas T Poulsen, Dominik Mumberg, Eric Santoni-Rugiu, Lars H Engelholm, Niels Behrendt

The genetic instability of cancer cells leads to cellular resistance against most targeted cancer drugs. Cancer-associated fibroblasts (CAF) infiltrate all carcinomas and are genetically stable. Using antibody-drug conjugates (ADC), we exploit the unique properties of a rapidly recycling endocytic receptor, uPARAP, to achieve highly efficient CAF-mediated drug delivery and killing of carcinomas. This receptor is generally not present on carcinoma cells and is only expressed in a restricted group of mesenchymal cancer cell types, which are sensitive to uPARAP-directed ADCs. However, we show that uPARAP is highly expressed in CAFs in all carcinoma types examined. This property is recapitulated in mouse xenograft carcinoma models. In these models, despite the absence of uPARAP on the carcinoma cells, uPARAP-targeting ADCs with clinically validated payloads, monomethyl auristatin E and deruxtecan, eradicated tumors with remarkable efficiency. Systemic treatment with anti-uPARAP ADC led to the permanent eradication of tumors in mice carrying subcutaneous xenografts with human EBC-1 lung carcinoma cells. A pronounced repression of tumor growth and a strongly increased mouse survival rate were also obtained with human HT29 colon adenocarcinoma cells, both when these tumors were growing subcutaneously and after the homing of tumor cells to bone from the circulation. CAFs were largely refractory to ADC treatment and retained a high expression of uPARAP. uPARAP-expressing fibroblasts could also process an anti-uPARAP ADC in vitro and deliver the cytotoxic component to carcinoma cells. The current bystander mechanism may be exploited in the majority of the most prevalent solid cancers, thus making uPARAP an extraordinarily versatile target for ADC-based cancer treatment.

癌细胞的遗传不稳定性导致细胞对大多数靶向癌症药物产生耐药性。癌症相关成纤维细胞(CAFs)浸润所有癌症,并且在遗传上是稳定的。利用抗体-药物偶联物(adc),我们利用快速循环的内吞受体uPARAP的独特特性,实现了高效的caff介导的药物传递和肿瘤杀伤。这种受体通常不存在于癌细胞中,只在对uparap导向的adc敏感的有限组间充质癌细胞类型中表达。然而,我们发现uPARAP在所有癌症类型的cas中都高度表达。这种特性在小鼠异种移植物癌模型中得到了再现。在这些模型中,尽管癌细胞上没有uPARAP,但uPARAP靶向adc与临床验证的有效载荷MMAE和Dxd根除肿瘤的效率显著。用抗uparap ADC进行全身治疗,可使携带人EBC-1肺癌细胞皮下异种移植物的小鼠的肿瘤永久根除。人类HT29结肠腺癌细胞也显著抑制肿瘤生长,并显著提高小鼠存活率,无论是当这些肿瘤在皮下生长时,还是肿瘤细胞从循环中归巢到骨后。CAFs对ADC治疗有很大的难治性,并且保持了uPARAP的高表达。表达uparap的成纤维细胞也可以在体外加工抗uparap ADC,并将细胞毒性成分传递给癌细胞。目前的旁观者机制可能在大多数最常见的实体癌症中被利用,从而使uPARAP成为基于adc的癌症治疗的一个非常通用的靶点。
{"title":"The Recycling Collagen Receptor uPARAP Is a Unique Mediator of Stromal Drug Delivery to Carcinoma Cells.","authors":"Kirstine S Nørregaard, Ida M E Larsen, Henrik J Jürgensen, Michaela Hansen Blomquist, Pinar Çakilkaya, Virginia Metrangolo, Alba Martinez Perlado, Oliver Krigslund, Henrik Gårdsvoll, Thomas T Poulsen, Dominik Mumberg, Eric Santoni-Rugiu, Lars H Engelholm, Niels Behrendt","doi":"10.1158/1535-7163.MCT-25-0051","DOIUrl":"10.1158/1535-7163.MCT-25-0051","url":null,"abstract":"<p><p>The genetic instability of cancer cells leads to cellular resistance against most targeted cancer drugs. Cancer-associated fibroblasts (CAF) infiltrate all carcinomas and are genetically stable. Using antibody-drug conjugates (ADC), we exploit the unique properties of a rapidly recycling endocytic receptor, uPARAP, to achieve highly efficient CAF-mediated drug delivery and killing of carcinomas. This receptor is generally not present on carcinoma cells and is only expressed in a restricted group of mesenchymal cancer cell types, which are sensitive to uPARAP-directed ADCs. However, we show that uPARAP is highly expressed in CAFs in all carcinoma types examined. This property is recapitulated in mouse xenograft carcinoma models. In these models, despite the absence of uPARAP on the carcinoma cells, uPARAP-targeting ADCs with clinically validated payloads, monomethyl auristatin E and deruxtecan, eradicated tumors with remarkable efficiency. Systemic treatment with anti-uPARAP ADC led to the permanent eradication of tumors in mice carrying subcutaneous xenografts with human EBC-1 lung carcinoma cells. A pronounced repression of tumor growth and a strongly increased mouse survival rate were also obtained with human HT29 colon adenocarcinoma cells, both when these tumors were growing subcutaneously and after the homing of tumor cells to bone from the circulation. CAFs were largely refractory to ADC treatment and retained a high expression of uPARAP. uPARAP-expressing fibroblasts could also process an anti-uPARAP ADC in vitro and deliver the cytotoxic component to carcinoma cells. The current bystander mechanism may be exploited in the majority of the most prevalent solid cancers, thus making uPARAP an extraordinarily versatile target for ADC-based cancer treatment.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"140-155"},"PeriodicalIF":5.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12757718/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144642885","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
IL15/IL15Rα Complex Induces an Antitumor Immune Response following Radiotherapy only in the Absence of Tregs and Fails to Expand Progenitor TCF1+ CD8 T Cells. IL15/IL15Ra复合物仅在缺乏Tregs的情况下诱导放射治疗后的抗肿瘤免疫反应,并且不能扩增祖细胞TCF1+ CD8 T细胞。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-02 DOI: 10.1158/1535-7163.MCT-24-0900
Miles Piper, Chloe A Hodgson, Jacob Gadwa, Maureen Hoen, Michael W Knitz, Justin Yu, Sophia Corbo, Nicholas A Olimpo, Elliott J Yee, Yuwen Zhu, Keira Y Larson, Christian Klein, Maria Amann, Anthony J Saviola, Sana D Karam

In this work, we show that the combination of radiotherapy (RT) and an IL15/IL15Rα fusion complex (IL15c) fails to confer antitumor efficacy; however, a CD8-driven antitumor immune response can be elicited with the concurrent administration of an aCD25 regulatory T cell-depleting antibody. Using IL15-/- and Rag1-/- knockout mouse models, we show that the response to RT + IL15c + aCD25 is dependent on both IL15 and cytotoxic T lymphocytes. Furthermore, despite an equivalent survival benefit following treatment with RT + IL15c + aCD25 and combination RT and PD1-IL2v, a novel immunocytokine with PD1- and IL2Rβγ-binding domains, cytotoxic T lymphocyte immunophenotyping and phosphoproteomics analysis of intracellular metabolites showed a significant upregulation of activation and functionality in CD8 T cells in the RT + PD1-IL2v regimen. Finally, we show that in the absence of functional IL15 signaling, the immunostimulatory response to RT + PD1-IL2v is significantly diminished with a concurrent lack of TCF+ CD8 T-cell generation, suggesting a necessity of IL15 for CD8 stem cells in mediating a durable response to treatment. Together, our results are illustrative of a mechanism wherein unimpeded effector T-cell activation through IL2Rβ signaling and regulatory T-cell inhibition are necessary in mediating an antitumor immune response.

在这项工作中,我们表明放射治疗(RT)和IL15/IL15Rα融合复合物(标记为IL15c)的组合不能赋予抗肿瘤功效;然而,cd8驱动的抗肿瘤免疫反应可以通过同时给药aCD25 treg消耗抗体引起。使用IL15-/-和Rag1-/-敲除小鼠模型,我们发现对RT + IL15c + aCD25的反应依赖于IL15和ctl。此外,尽管RT + IL15c + aCD25以及RT和PD1-IL2v联合治疗具有同等的生存益处,但一种具有PD-1和IL2Rbg结合域的新型免疫细胞因子、CTL免疫表型和细胞内代谢物的磷酸化蛋白质组学分析显示,在RT + PD1-IL2v治疗方案中,CD8 T细胞的活化和功能显著上调。最后,我们发现,在缺乏功能性IL15信号的情况下,对RT + PD1-IL2v的免疫刺激反应显著减弱,同时缺乏TCF+ CD8 T细胞的生成,这表明CD8干细胞需要IL15来介导对治疗的持久反应。总之,我们的结果说明了一种机制,其中通过IL2Rb信号传导和Treg抑制的无阻碍效应T细胞激活是介导抗肿瘤免疫反应所必需的。
{"title":"IL15/IL15Rα Complex Induces an Antitumor Immune Response following Radiotherapy only in the Absence of Tregs and Fails to Expand Progenitor TCF1+ CD8 T Cells.","authors":"Miles Piper, Chloe A Hodgson, Jacob Gadwa, Maureen Hoen, Michael W Knitz, Justin Yu, Sophia Corbo, Nicholas A Olimpo, Elliott J Yee, Yuwen Zhu, Keira Y Larson, Christian Klein, Maria Amann, Anthony J Saviola, Sana D Karam","doi":"10.1158/1535-7163.MCT-24-0900","DOIUrl":"10.1158/1535-7163.MCT-24-0900","url":null,"abstract":"<p><p>In this work, we show that the combination of radiotherapy (RT) and an IL15/IL15Rα fusion complex (IL15c) fails to confer antitumor efficacy; however, a CD8-driven antitumor immune response can be elicited with the concurrent administration of an aCD25 regulatory T cell-depleting antibody. Using IL15-/- and Rag1-/- knockout mouse models, we show that the response to RT + IL15c + aCD25 is dependent on both IL15 and cytotoxic T lymphocytes. Furthermore, despite an equivalent survival benefit following treatment with RT + IL15c + aCD25 and combination RT and PD1-IL2v, a novel immunocytokine with PD1- and IL2Rβγ-binding domains, cytotoxic T lymphocyte immunophenotyping and phosphoproteomics analysis of intracellular metabolites showed a significant upregulation of activation and functionality in CD8 T cells in the RT + PD1-IL2v regimen. Finally, we show that in the absence of functional IL15 signaling, the immunostimulatory response to RT + PD1-IL2v is significantly diminished with a concurrent lack of TCF+ CD8 T-cell generation, suggesting a necessity of IL15 for CD8 stem cells in mediating a durable response to treatment. Together, our results are illustrative of a mechanism wherein unimpeded effector T-cell activation through IL2Rβ signaling and regulatory T-cell inhibition are necessary in mediating an antitumor immune response.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"168-182"},"PeriodicalIF":5.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144961723","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
In Vivo Tumor Growth Control by General Control Nonderepressible 2-Targeting Agents Results from Kinase Activation. 激酶激活的一般控制非抑制2靶向药物对体内肿瘤生长的控制
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2026-01-02 DOI: 10.1158/1535-7163.MCT-24-0960
Feven Tameire, Paulina Wojnarowicz, Crissy Dudgeon, Kathryn T Bieging-Rolett, Sho Fujisawa, Savi Ramurthy, Owen Reilly, Christopher G Thomson, Bradley S Sherborne, Simon J Taylor, Fang He, Pengwei Pan, Baozhong Li, Earl May, Alan C Rigby, Mark J Mulvihill, Nandita Bose, David Surguladze, Eric S Lightcap

General control nonderepressible 2 (GCN2; EIF2AK4) is a serine-threonine kinase in the integrated stress response signaling pathway that initiates adaptive responses during nutrient stress conditions. Although pharmacologic inhibition of GCN2 under nutrient stress conditions induces apoptosis and inhibits tumor growth, GCN2 inhibition without nutrient stress has been reported to have no effect on tumor growth. By exploring an array of GCN2 inhibitors, we demonstrate that multiple agents in fact activate GCN2 in biochemical and cell-based assays at low concentrations and inhibit GCN2 at higher concentrations. Unexpectedly, it is this activation, and not inhibition, of the GCN2 pathway that is associated with decreased viability in vitro and tumor growth inhibition in vivo across multiple models. Knockdown and knockout experiments show that activation of the integrated stress response by GCN2-targeting agents is dependent on GCN2. ISRIB, a modulator of eIF2B, ablates the viability effect, demonstrating the dependence on translation initiation. Activating doses result in the induction of cleaved caspase 3 and cleaved PARP. In contrast, a nonactivating GCN2-targeting agent does not affect viability. These results provide a clearer understanding of the challenges and opportunities for the clinical development of compounds targeting GCN2.

General Control nonderexsible 2 (GCN2; EIF2AK4)是综合应激反应(ISR)信号通路中的一种丝氨酸-苏氨酸激酶,在营养胁迫条件下启动适应性反应。在营养胁迫条件下,药理抑制GCN2可诱导细胞凋亡并抑制肿瘤生长,而在没有营养胁迫的情况下抑制GCN2对肿瘤生长没有影响。通过探索一系列GCN2抑制剂,我们证明了多种药物实际上在低浓度的生化和细胞基础实验中激活GCN2,而在高浓度时抑制GCN2。出乎意料的是,正是这种GCN2途径的激活,而不是抑制,在多种模型中与体外生存能力下降和体内肿瘤生长抑制相关。敲除和敲除实验表明,GCN2靶向药物对ISR的激活依赖于GCN2。作为eIF2B的调制剂,ISRIB消除了生存力效应,表明其依赖于翻译起始。激活剂量可诱导裂解caspase 3和裂解PARP。相反,非激活gcn2靶向剂不会影响生存能力。这些结果为临床开发靶向GCN2的化合物提供了更清晰的挑战和机遇。
{"title":"In Vivo Tumor Growth Control by General Control Nonderepressible 2-Targeting Agents Results from Kinase Activation.","authors":"Feven Tameire, Paulina Wojnarowicz, Crissy Dudgeon, Kathryn T Bieging-Rolett, Sho Fujisawa, Savi Ramurthy, Owen Reilly, Christopher G Thomson, Bradley S Sherborne, Simon J Taylor, Fang He, Pengwei Pan, Baozhong Li, Earl May, Alan C Rigby, Mark J Mulvihill, Nandita Bose, David Surguladze, Eric S Lightcap","doi":"10.1158/1535-7163.MCT-24-0960","DOIUrl":"10.1158/1535-7163.MCT-24-0960","url":null,"abstract":"<p><p>General control nonderepressible 2 (GCN2; EIF2AK4) is a serine-threonine kinase in the integrated stress response signaling pathway that initiates adaptive responses during nutrient stress conditions. Although pharmacologic inhibition of GCN2 under nutrient stress conditions induces apoptosis and inhibits tumor growth, GCN2 inhibition without nutrient stress has been reported to have no effect on tumor growth. By exploring an array of GCN2 inhibitors, we demonstrate that multiple agents in fact activate GCN2 in biochemical and cell-based assays at low concentrations and inhibit GCN2 at higher concentrations. Unexpectedly, it is this activation, and not inhibition, of the GCN2 pathway that is associated with decreased viability in vitro and tumor growth inhibition in vivo across multiple models. Knockdown and knockout experiments show that activation of the integrated stress response by GCN2-targeting agents is dependent on GCN2. ISRIB, a modulator of eIF2B, ablates the viability effect, demonstrating the dependence on translation initiation. Activating doses result in the induction of cleaved caspase 3 and cleaved PARP. In contrast, a nonactivating GCN2-targeting agent does not affect viability. These results provide a clearer understanding of the challenges and opportunities for the clinical development of compounds targeting GCN2.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"71-83"},"PeriodicalIF":5.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144961755","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
Non-Glycosylated, Legumain-Cleavable ISACs Drive Potent Anti-Tumor Immunotherapy via a Bystander Effect. 非糖基化,豆科蛋白可切割的ISACs通过旁观者效应驱动有效的抗肿瘤免疫治疗。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-31 DOI: 10.1158/1535-7163.MCT-25-1153
Anqi Zhang, Mohan Reddy Mullapudi, Lina Wu, Michele Yi, Mohammad Asikur Rahman, Aiman A Yaseen, H Kayla Zhong, Tracy A Brooks, L Nathan Tumey

Pancreatic ductal adenocarcinoma (PDAC) is the most lethal form of pancreatic cancer, with poor prognosis driven by late diagnosis, therapeutic resistance, and an immunosuppressive tumor microenvironment. Interactions between tumor cells and immune cells promote immune evasion and tumor progression, limiting the efficacy of immune checkpoint blockade and other immunotherapies. Given the high expression of TROP2 in PDAC, we developed a TROP2-targeted TLR7 agonist (E104) designed for selective accumulation within the tumor microenvironment to activate antitumor immunity. Although antibody bioactivity is traditionally linked to Fcγ receptor (FcγR) engagement and recruitment of effector cells, our legumain-cleavable, non-glycosylated immune-stimulating antibody conjugates (NG-ISACs) induce robust myeloid activation, cytokine release, and tumor regression without FcγR-mediated functions of natural killer cells or macrophages. Rather, NG-anti-TROP2-E104-ISACs depend on tumor antigen recognition and TLR7 activation, not FcγR-driven antibody-dependent cellular cytotoxicity (ADCC) or antibody-dependent cellular phagocytosis (ADCP), to elicit tumor regression and adaptive immunity, as evidenced by anti-TROP2 antibody generation in a syngeneic model. By incorporating the cell-permeable E104 payload with bystander activity, NG-ISACs can activate immune responses independently of FcγR binding. In vitro, NG-anti-TROP2-E104-ISACs bypass FcγRIIa-mediated ADCC and FcγRIIIa-mediated ADCP while maintaining potency in co-cultures of TROP2-positive tumor and effector cells. Moreover, NG-anti-TROP2-E104-ISACs display reduced acute toxicity compared to glycosylated counterparts. Together, these findings delineate the bystander mechanism underlying FcγR-independent immune stimulation and establish a framework for designing ISACs with improved safety.

胰腺导管腺癌(Pancreatic ductal adencarcinoma, PDAC)是最致命的胰腺癌,由于诊断较晚、治疗耐药和肿瘤微环境免疫抑制,预后较差。肿瘤细胞和免疫细胞之间的相互作用促进免疫逃避和肿瘤进展,限制了免疫检查点封锁和其他免疫疗法的疗效。鉴于TROP2在PDAC中的高表达,我们开发了一种靶向TROP2的TLR7激动剂(E104),设计用于在肿瘤微环境中选择性积累以激活抗肿瘤免疫。虽然抗体的生物活性传统上与Fcγ受体(Fcγ r)的结合和效应细胞的募集有关,但我们的非糖基化免疫刺激抗体偶联物(NG-ISACs)可诱导强大的髓细胞活化、细胞因子释放和肿瘤消退,而不需要Fcγ r介导的自然杀伤细胞或巨噬细胞的功能。相反,NG-anti-TROP2-E104-ISACs依赖于肿瘤抗原识别和TLR7激活,而不是fc - γ r驱动的抗体依赖性细胞毒性(ADCC)或抗体依赖性细胞吞噬(ADCP),从而引发肿瘤消退和适应性免疫,这一点在同基因模型中的抗trop2抗体产生中得到了证明。通过将具有细胞渗透性的E104有效载荷与旁观者活性结合,NG-ISACs可以独立于FcγR结合激活免疫反应。在体外,NG-anti-TROP2-E104-ISACs绕过fc γ riia介导的ADCC和fc γ riia介导的ADCP,同时在trop2阳性肿瘤和效应细胞共培养中保持效力。此外,NG-anti-TROP2-E104-ISACs与糖基化的对应物相比,表现出更低的急性毒性。总之,这些发现描述了fc γ r非依赖性免疫刺激的旁观者机制,并为设计具有更高安全性的ISACs建立了框架。
{"title":"Non-Glycosylated, Legumain-Cleavable ISACs Drive Potent Anti-Tumor Immunotherapy via a Bystander Effect.","authors":"Anqi Zhang, Mohan Reddy Mullapudi, Lina Wu, Michele Yi, Mohammad Asikur Rahman, Aiman A Yaseen, H Kayla Zhong, Tracy A Brooks, L Nathan Tumey","doi":"10.1158/1535-7163.MCT-25-1153","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-1153","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) is the most lethal form of pancreatic cancer, with poor prognosis driven by late diagnosis, therapeutic resistance, and an immunosuppressive tumor microenvironment. Interactions between tumor cells and immune cells promote immune evasion and tumor progression, limiting the efficacy of immune checkpoint blockade and other immunotherapies. Given the high expression of TROP2 in PDAC, we developed a TROP2-targeted TLR7 agonist (E104) designed for selective accumulation within the tumor microenvironment to activate antitumor immunity. Although antibody bioactivity is traditionally linked to Fcγ receptor (FcγR) engagement and recruitment of effector cells, our legumain-cleavable, non-glycosylated immune-stimulating antibody conjugates (NG-ISACs) induce robust myeloid activation, cytokine release, and tumor regression without FcγR-mediated functions of natural killer cells or macrophages. Rather, NG-anti-TROP2-E104-ISACs depend on tumor antigen recognition and TLR7 activation, not FcγR-driven antibody-dependent cellular cytotoxicity (ADCC) or antibody-dependent cellular phagocytosis (ADCP), to elicit tumor regression and adaptive immunity, as evidenced by anti-TROP2 antibody generation in a syngeneic model. By incorporating the cell-permeable E104 payload with bystander activity, NG-ISACs can activate immune responses independently of FcγR binding. In vitro, NG-anti-TROP2-E104-ISACs bypass FcγRIIa-mediated ADCC and FcγRIIIa-mediated ADCP while maintaining potency in co-cultures of TROP2-positive tumor and effector cells. Moreover, NG-anti-TROP2-E104-ISACs display reduced acute toxicity compared to glycosylated counterparts. Together, these findings delineate the bystander mechanism underlying FcγR-independent immune stimulation and establish a framework for designing ISACs with improved safety.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145864164","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
PM534, a Novel Colchicine‑Site Tubulin Inhibitor with Broad‑Spectrum and Resistance‑Overcoming Antitumor Activity. PM534,一种具有广谱抗肿瘤活性的新型秋水仙碱微管蛋白抑制剂。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-31 DOI: 10.1158/1535-7163.MCT-25-0559
Pablo Aviles, Marcelo Lima Ribeiro, Maria Jose Guillén, Marta Martinez-Diez, Maria Jose Muñoz-Alonso, Gema Santamaria-Nuñez, Daniel Torralba, Patricia Alamo, Alberto Gallardo, María A Oliva, Ramon Mangues, J Fernando Diaz, Carmen Cuevas

This study evaluates PM534, a novel colchicine-binding domain inhibitor, for its potential in cancer therapy. PM534 exhibited potent in vitro efficacy against a panel of fourteen human cancer cell lines, including breast, ovarian, and prostate cancers, with GI50 values in the low nanomolar range. Both continuous (72 hours) and short-term (1-24 hours) exposure led to irreversible effects, inducing G2/M cell cycle arrest and multinucleation. Additionally, PM534 also impaired angiogenic process. It effectively inhibited HUVEC cell functions, including adhesion with an IC50 of 2.3 nM, markedly more potent than colchicine (IC50 = 1,800 nM). At concentrations as low as 1.6 nM, PM534 delayed wound closure in migration assays, completely inhibiting migration above 4 nM. Additionally, PM534 abrogated invasion and disrupted capillary-like network formation at concentrations starting from 0.5 nM, without inducing cytotoxicity. In vivo PM534 demonstrated robust antitumor efficacy across six xenograft models, including ovarian (A2780, ES-2), triple-negative breast (MDA-MB-231, HCC-1937), and prostate (VCaP, 22Rv1) tumors. This treatment also led to statistically significant increases in median survival times across all models, without inducing signs of systemic toxicity. Mechanistically, PM534 induced apoptosis, mitotic catastrophe, and necrosis in tumor tissues. Importantly, PM534 retained efficacy in models overexpressing multidrug resistance proteins P-glycoprotein or β-III tubulin, overcoming common resistance mechanisms that limit the effectiveness of other tubulin-binding agents. Collectively, these findings highlight PM534 as a promising antitumor agent with potent activity against diverse and treatment-resistant malignancies. A Phase I clinical trial (NCT05835609) is underway to assess the therapeutic potential of PM534 in patients with advanced solid tumors.

本研究评估了新型秋水仙碱结合域抑制剂PM534在癌症治疗中的潜力。PM534在体外对14种人类癌细胞系(包括乳腺癌、卵巢癌和前列腺癌)表现出强有力的疗效,其GI50值在低纳摩尔范围内。连续(72小时)和短期(1-24小时)暴露均导致不可逆效应,诱导G2/M细胞周期阻滞和多核。此外,PM534还会损害血管生成过程。有效抑制HUVEC细胞功能,包括粘附,IC50为2.3 nM,明显高于秋水仙碱(IC50 = 1,800 nM)。在低至1.6 nM的浓度下,PM534在迁移实验中延迟伤口闭合,完全抑制4 nM以上的迁移。此外,PM534在0.5 nM的浓度下消除了入侵并破坏了毛细血管样网络的形成,而不会引起细胞毒性。体内PM534在六种异种移植模型中显示出强大的抗肿瘤功效,包括卵巢(A2780, ES-2),三阴性乳腺(MDA-MB-231, HCC-1937)和前列腺(VCaP, 22Rv1)肿瘤。这种治疗还导致所有模型的中位生存时间在统计学上显著增加,没有引起全身毒性的迹象。机制上,PM534诱导肿瘤组织凋亡、有丝分裂突变和坏死。重要的是,PM534在过表达多药耐药蛋白p -糖蛋白或β-III微管蛋白的模型中保持了疗效,克服了限制其他微管蛋白结合剂有效性的常见耐药机制。总的来说,这些发现突出了PM534作为一种有希望的抗肿瘤药物,对多种恶性肿瘤和治疗抵抗具有有效活性。一项I期临床试验(NCT05835609)正在进行中,以评估PM534在晚期实体瘤患者中的治疗潜力。
{"title":"PM534, a Novel Colchicine‑Site Tubulin Inhibitor with Broad‑Spectrum and Resistance‑Overcoming Antitumor Activity.","authors":"Pablo Aviles, Marcelo Lima Ribeiro, Maria Jose Guillén, Marta Martinez-Diez, Maria Jose Muñoz-Alonso, Gema Santamaria-Nuñez, Daniel Torralba, Patricia Alamo, Alberto Gallardo, María A Oliva, Ramon Mangues, J Fernando Diaz, Carmen Cuevas","doi":"10.1158/1535-7163.MCT-25-0559","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0559","url":null,"abstract":"<p><p>This study evaluates PM534, a novel colchicine-binding domain inhibitor, for its potential in cancer therapy. PM534 exhibited potent in vitro efficacy against a panel of fourteen human cancer cell lines, including breast, ovarian, and prostate cancers, with GI50 values in the low nanomolar range. Both continuous (72 hours) and short-term (1-24 hours) exposure led to irreversible effects, inducing G2/M cell cycle arrest and multinucleation. Additionally, PM534 also impaired angiogenic process. It effectively inhibited HUVEC cell functions, including adhesion with an IC50 of 2.3 nM, markedly more potent than colchicine (IC50 = 1,800 nM). At concentrations as low as 1.6 nM, PM534 delayed wound closure in migration assays, completely inhibiting migration above 4 nM. Additionally, PM534 abrogated invasion and disrupted capillary-like network formation at concentrations starting from 0.5 nM, without inducing cytotoxicity. In vivo PM534 demonstrated robust antitumor efficacy across six xenograft models, including ovarian (A2780, ES-2), triple-negative breast (MDA-MB-231, HCC-1937), and prostate (VCaP, 22Rv1) tumors. This treatment also led to statistically significant increases in median survival times across all models, without inducing signs of systemic toxicity. Mechanistically, PM534 induced apoptosis, mitotic catastrophe, and necrosis in tumor tissues. Importantly, PM534 retained efficacy in models overexpressing multidrug resistance proteins P-glycoprotein or β-III tubulin, overcoming common resistance mechanisms that limit the effectiveness of other tubulin-binding agents. Collectively, these findings highlight PM534 as a promising antitumor agent with potent activity against diverse and treatment-resistant malignancies. A Phase I clinical trial (NCT05835609) is underway to assess the therapeutic potential of PM534 in patients with advanced solid tumors.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145892759","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Molecular Cancer Therapeutics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1