首页 > 最新文献

Cancer research最新文献

英文 中文
Resistance Management for Cancer: Lessons from Farmers. 癌症的抗药性管理:农民的经验教训。
IF 2.9 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-15 DOI: 10.1158/0008-5472.CAN-23-3374
Sareh Seyedi, Valerie K Harris, Stefania E Kapsetaki, Shrinath Narayanan, Daniel Saha, Zachary Compton, Rezvan Yousefi, Alexander May, Efe Fakir, Amy M Boddy, Marco Gerlinger, Christina Wu, Lida Mina, Silvie Huijben, Dawn H Gouge, Luis Cisneros, Peter C Ellsworth, Carlo C Maley

One of the main reasons we have not been able to cure cancers is that treatments select for drug-resistant cells. Pest managers face similar challenges with pesticides selecting for pesticide-resistant insects, resulting in similar mechanisms of resistance. Pest managers have developed 10 principles that could be translated to controlling cancers: (i) prevent onset, (ii) monitor continuously, (iii) identify thresholds below which there will be no intervention, (iv) change interventions in response to burden, (v) preferentially select nonchemical control methods, (vi) use target-specific drugs, (vii) use the lowest effective dose, (viii) reduce cross-resistance, (ix) evaluate success based on long-term management, and (x) forecast growth and response. These principles are general to all cancers and cancer drugs and so could be employed broadly to improve oncology. Here, we review the parallel difficulties in controlling drug resistance in pests and cancer cells. We show how the principles of resistance management in pests might be applied to cancer. Integrated pest management inspired the development of adaptive therapy in oncology to increase progression-free survival and quality of life in patients with cancers where cures are unlikely. These pest management principles have the potential to inform clinical trial design.

我们无法治愈癌症的主要原因之一是,治疗方法会选择出抗药性细胞。害虫管理者也面临着类似的挑战,杀虫剂会选择出抗药性昆虫,从而产生类似的抗药性机制。害虫管理者已经制定了十项原则,这些原则可以转化为控制癌症的原则:(1)预防发病;(2)持续监测;(3)确定阈值,低于阈值将不予干预;(4)根据负担改变干预措施;(5)优先选择非化学控制方法;(6)使用靶向药物;(7)使用最低有效剂量;(8)减少交叉抗药性;(9)根据长期管理评估成功与否;(10)预测生长和反应。这些原则适用于所有癌症和抗癌药物,因此可广泛用于改善肿瘤学。在此,我们回顾了在控制害虫和癌细胞抗药性方面遇到的并行困难。我们展示了如何将害虫抗药性管理原则应用于癌症。害虫综合防治启发了肿瘤适应性疗法的发展,以提高癌症患者的无进展生存期和生活质量,而这是不可能治愈的。这些害虫管理原则有可能为临床试验设计提供参考。
{"title":"Resistance Management for Cancer: Lessons from Farmers.","authors":"Sareh Seyedi, Valerie K Harris, Stefania E Kapsetaki, Shrinath Narayanan, Daniel Saha, Zachary Compton, Rezvan Yousefi, Alexander May, Efe Fakir, Amy M Boddy, Marco Gerlinger, Christina Wu, Lida Mina, Silvie Huijben, Dawn H Gouge, Luis Cisneros, Peter C Ellsworth, Carlo C Maley","doi":"10.1158/0008-5472.CAN-23-3374","DOIUrl":"10.1158/0008-5472.CAN-23-3374","url":null,"abstract":"<p><p>One of the main reasons we have not been able to cure cancers is that treatments select for drug-resistant cells. Pest managers face similar challenges with pesticides selecting for pesticide-resistant insects, resulting in similar mechanisms of resistance. Pest managers have developed 10 principles that could be translated to controlling cancers: (i) prevent onset, (ii) monitor continuously, (iii) identify thresholds below which there will be no intervention, (iv) change interventions in response to burden, (v) preferentially select nonchemical control methods, (vi) use target-specific drugs, (vii) use the lowest effective dose, (viii) reduce cross-resistance, (ix) evaluate success based on long-term management, and (x) forecast growth and response. These principles are general to all cancers and cancer drugs and so could be employed broadly to improve oncology. Here, we review the parallel difficulties in controlling drug resistance in pests and cancer cells. We show how the principles of resistance management in pests might be applied to cancer. Integrated pest management inspired the development of adaptive therapy in oncology to increase progression-free survival and quality of life in patients with cancers where cures are unlikely. These pest management principles have the potential to inform clinical trial design.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":"3715-3727"},"PeriodicalIF":2.9,"publicationDate":"2024-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11565176/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142364508","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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 Dual-Payload Antibody-Drug Conjugate Targeting CD276/B7-H3 Elicits Cytotoxicity and Immune Activation in Triple-Negative Breast Cancer. 靶向 CD276/B7-H3 的双负载抗体-药物共轭物在三阴性乳腺癌中激发细胞毒性和免疫激活作用
IF 2.9 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-15 DOI: 10.1158/0008-5472.CAN-23-4099
Zhuoxin Zora Zhou, Yingnan Si, Jiashuai Zhang, Kai Chen, Ashley George, Seulhee Kim, Lufang Zhou, Xiaoguang Margaret Liu

Triple-negative breast cancer (TNBC) is a highly aggressive and heterogeneous disease that often relapses following treatment with standard radiotherapies and cytotoxic chemotherapies. Combination therapies have potential for treating refractory metastatic TNBC. In this study, we aimed to develop an antibody-drug conjugate with dual payloads (DualADC) as a chemoimmunotherapy for TNBC. The overexpression of an immune checkpoint transmembrane CD276 (also known as B7-H3) was associated with angiogenesis, metastasis, and immune tolerance in more than 60% of patients with TNBC. Development of a mAb capable of targeting the extracellular domain of surface CD276 enabled delivery of payloads to tumors, and a platform was established for concurrent conjugation of a traditional cytotoxic payload and an immunoregulating Toll-like receptor 7/8 agonist to the CD276 mAb. The DualADC effectively killed multiple TNBC subtypes, significantly enhanced immune functions in the tumor microenvironment, and reduced tumor burden by up to 90% to 100% in animal studies. Single-cell RNA sequencing, multiplex cytokine analysis, and histology elucidated the impact of treatment on tumor cells and the immune landscape. This study suggests that the developed DualADC could represent a promising targeted chemoimmunotherapy for TNBC. Significance: An anti-CD276 monoclonal antibody conjugated with both a cytotoxic drug and an immune boosting reagent effectively targets triple-negative breast cancer by inducing tumor cell death and stimulating immune cell infiltration.

三阴性乳腺癌(TNBC)是一种侵袭性极强的异质性疾病,在接受标准放射治疗和细胞毒性化疗后往往会复发。联合疗法具有治疗难治性转移性 TNBC 的潜力。在此,我们旨在开发一种具有双重有效载荷的抗体药物共轭物(DualADC),作为 TNBC 的化疗免疫疗法。在超过60%的TNBC患者中,免疫检查点跨膜CD276(又称B7-H3)的过度表达与血管生成、转移和免疫耐受有关。开发出一种能够靶向表面 CD276 细胞外结构域的单克隆抗体 (mAb),从而能够向肿瘤输送有效载荷,并建立了一个平台,将传统的细胞毒性有效载荷和免疫调节收费样受体 7/8 激动剂同时与 CD276 mAb 连接。DualADC 能有效杀死多种 TNBC 亚型,显著增强肿瘤微环境中的免疫功能,并在动物实验中将肿瘤负荷降低了 90-100%。单细胞 RNA 序列测定、多重细胞因子分析和组织学研究阐明了治疗对肿瘤细胞和免疫环境的影响。这项研究表明,所开发的 DualADC 是一种治疗 TNBC 的前景广阔的靶向化疗免疫疗法。
{"title":"A Dual-Payload Antibody-Drug Conjugate Targeting CD276/B7-H3 Elicits Cytotoxicity and Immune Activation in Triple-Negative Breast Cancer.","authors":"Zhuoxin Zora Zhou, Yingnan Si, Jiashuai Zhang, Kai Chen, Ashley George, Seulhee Kim, Lufang Zhou, Xiaoguang Margaret Liu","doi":"10.1158/0008-5472.CAN-23-4099","DOIUrl":"10.1158/0008-5472.CAN-23-4099","url":null,"abstract":"<p><p>Triple-negative breast cancer (TNBC) is a highly aggressive and heterogeneous disease that often relapses following treatment with standard radiotherapies and cytotoxic chemotherapies. Combination therapies have potential for treating refractory metastatic TNBC. In this study, we aimed to develop an antibody-drug conjugate with dual payloads (DualADC) as a chemoimmunotherapy for TNBC. The overexpression of an immune checkpoint transmembrane CD276 (also known as B7-H3) was associated with angiogenesis, metastasis, and immune tolerance in more than 60% of patients with TNBC. Development of a mAb capable of targeting the extracellular domain of surface CD276 enabled delivery of payloads to tumors, and a platform was established for concurrent conjugation of a traditional cytotoxic payload and an immunoregulating Toll-like receptor 7/8 agonist to the CD276 mAb. The DualADC effectively killed multiple TNBC subtypes, significantly enhanced immune functions in the tumor microenvironment, and reduced tumor burden by up to 90% to 100% in animal studies. Single-cell RNA sequencing, multiplex cytokine analysis, and histology elucidated the impact of treatment on tumor cells and the immune landscape. This study suggests that the developed DualADC could represent a promising targeted chemoimmunotherapy for TNBC. Significance: An anti-CD276 monoclonal antibody conjugated with both a cytotoxic drug and an immune boosting reagent effectively targets triple-negative breast cancer by inducing tumor cell death and stimulating immune cell infiltration.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":"3848-3863"},"PeriodicalIF":2.9,"publicationDate":"2024-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11565169/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142072067","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Low-Molecular Weight Cyclin E Confers a Vulnerability to PKMYT1 Inhibition in Triple-Negative Breast Cancer. 低分子量细胞周期蛋白 E 使三阴性乳腺癌易受 PKMYT1 抑制作用的影响
IF 2.9 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-15 DOI: 10.1158/0008-5472.CAN-23-4130
Mi Li, Amriti R Lulla, Yan Wang, Spyros Tsavaschidis, Fuchenchu Wang, Cansu Karakas, Tuyen D T Nguyen, Tuyen N Bui, Marc A Pina, Mei-Kuang Chen, Sofia Mastoraki, Asha S Multani, Natalie W Fowlkes, Aysegul Sahin, C Gary Marshall, Kelly K Hunt, Khandan Keyomarsi

Cyclin E is a regulatory subunit of CDK2 that mediates S phase entry and progression. The cleavage of full-length cyclin E (FL-cycE) to low-molecular weight isoforms (LMW-E) dramatically alters substrate specificity, promoting G1-S cell cycle transition and accelerating mitotic exit. Approximately 70% of triple-negative breast cancers (TNBC) express LMW-E, which correlates with poor prognosis. PKMYT1 also plays an important role in mitosis by inhibiting CDK1 to block premature mitotic entry, suggesting it could be a therapeutic target in TNBC expressing LMW-E. In this study, analysis of tumor samples of patients with TNBC revealed that coexpression of LMW-E and PKMYT1-catalyzed CDK1 phosphorylation predicted poor response to neoadjuvant chemotherapy. Compared with FL-cycE, LMW-E specifically upregulates PKMYT1 expression and protein stability, thereby increasing CDK1 phosphorylation. Inhibiting PKMYT1 with the selective inhibitor RP-6306 (lunresertib) elicited LMW-E-dependent antitumor effects, accelerating premature mitotic entry, inhibiting replication fork restart, and enhancing DNA damage, chromosomal breakage, apoptosis, and replication stress. Importantly, TNBC cell line xenografts expressing LMW-E showed greater sensitivity to RP-6306 than tumors with empty vector or FL-cycE. Furthermore, RP-6306 exerted tumor suppressive effects in LMW-E transgenic murine mammary tumors and patient-derived xenografts of LMW-E-high TNBC but not in the LMW-E null models examined in parallel. Lastly, transcriptomic and immune profiling demonstrated that RP-6306 treatment induced interferon responses and T-cell infiltration in the LMW-E-high tumor microenvironment, enhancing the antitumor immune response. These findings highlight the LMW-E/PKMYT1/CDK1 regulatory axis as a promising therapeutic target in TNBC, providing the rationale for further clinical development of PKMYT1 inhibitors in this aggressive breast cancer subtype. Significance: PKMYT1 upregulation and CDK1 phosphorylation in triple-negative breast cancer expressing low-molecular weight cyclin E leads to suboptimal responses to chemotherapy but sensitizes tumors to PKMYT1 inhibitors, proposing a personalized treatment strategy.

细胞周期蛋白 E 是 CDK2 的调节亚基,介导 S 期的进入和进展。将全长细胞周期蛋白 E(FL-cycE)裂解为低分子量异构体(LMW-E)可显著改变底物的特异性,促进 G1/S 细胞周期转变并加速有丝分裂的退出。约 70% 的三阴性乳腺癌(TNBC)表达 LMW-E,这与预后不良有关。PKMYT1还通过抑制CDK1阻止有丝分裂过早进入而在有丝分裂中发挥重要作用,这表明它可能是表达LMW-E的TNBC的治疗靶点。对TNBC患者肿瘤样本的分析表明,LMW-E和PKMYT1催化的CDK1磷酸化的共同表达预示着对新辅助化疗的不良反应。与FL-cycE相比,LMW-E能特异性地上调PKMYT1的表达和蛋白稳定性,从而提高CDK1的磷酸化。用选择性抑制剂RP-6306(lunresertib)抑制PKMYT1可产生依赖于LMW-E的抗肿瘤效应,加速有丝分裂过早进入,抑制复制叉重启,增强DNA损伤、染色体断裂、细胞凋亡和复制应激。重要的是,表达 LMW-E 的 TNBC 细胞系异种移植对 RP-6306 的敏感性高于空载体或 FL-cycE 的肿瘤。此外,RP-6306 在 LMW-E 转基因小鼠乳腺肿瘤和 LMW-E 高的 TNBC 患者衍生异种移植物中发挥了抑制肿瘤的作用,但在同时检测的 LMW-E 空模型中却没有这种作用。最后,转录组和免疫分析表明,RP-6306 治疗可诱导 LMW-E 高肿瘤微环境中的干扰素反应和 T 细胞浸润,从而增强抗肿瘤免疫反应。这些发现强调了LMW-E/PKMYT1/CDK1调控轴是TNBC的一个很有前景的治疗靶点,为在这一侵袭性乳腺癌亚型中进一步临床开发PKMYT1抑制剂提供了依据。
{"title":"Low-Molecular Weight Cyclin E Confers a Vulnerability to PKMYT1 Inhibition in Triple-Negative Breast Cancer.","authors":"Mi Li, Amriti R Lulla, Yan Wang, Spyros Tsavaschidis, Fuchenchu Wang, Cansu Karakas, Tuyen D T Nguyen, Tuyen N Bui, Marc A Pina, Mei-Kuang Chen, Sofia Mastoraki, Asha S Multani, Natalie W Fowlkes, Aysegul Sahin, C Gary Marshall, Kelly K Hunt, Khandan Keyomarsi","doi":"10.1158/0008-5472.CAN-23-4130","DOIUrl":"10.1158/0008-5472.CAN-23-4130","url":null,"abstract":"<p><p>Cyclin E is a regulatory subunit of CDK2 that mediates S phase entry and progression. The cleavage of full-length cyclin E (FL-cycE) to low-molecular weight isoforms (LMW-E) dramatically alters substrate specificity, promoting G1-S cell cycle transition and accelerating mitotic exit. Approximately 70% of triple-negative breast cancers (TNBC) express LMW-E, which correlates with poor prognosis. PKMYT1 also plays an important role in mitosis by inhibiting CDK1 to block premature mitotic entry, suggesting it could be a therapeutic target in TNBC expressing LMW-E. In this study, analysis of tumor samples of patients with TNBC revealed that coexpression of LMW-E and PKMYT1-catalyzed CDK1 phosphorylation predicted poor response to neoadjuvant chemotherapy. Compared with FL-cycE, LMW-E specifically upregulates PKMYT1 expression and protein stability, thereby increasing CDK1 phosphorylation. Inhibiting PKMYT1 with the selective inhibitor RP-6306 (lunresertib) elicited LMW-E-dependent antitumor effects, accelerating premature mitotic entry, inhibiting replication fork restart, and enhancing DNA damage, chromosomal breakage, apoptosis, and replication stress. Importantly, TNBC cell line xenografts expressing LMW-E showed greater sensitivity to RP-6306 than tumors with empty vector or FL-cycE. Furthermore, RP-6306 exerted tumor suppressive effects in LMW-E transgenic murine mammary tumors and patient-derived xenografts of LMW-E-high TNBC but not in the LMW-E null models examined in parallel. Lastly, transcriptomic and immune profiling demonstrated that RP-6306 treatment induced interferon responses and T-cell infiltration in the LMW-E-high tumor microenvironment, enhancing the antitumor immune response. These findings highlight the LMW-E/PKMYT1/CDK1 regulatory axis as a promising therapeutic target in TNBC, providing the rationale for further clinical development of PKMYT1 inhibitors in this aggressive breast cancer subtype. Significance: PKMYT1 upregulation and CDK1 phosphorylation in triple-negative breast cancer expressing low-molecular weight cyclin E leads to suboptimal responses to chemotherapy but sensitizes tumors to PKMYT1 inhibitors, proposing a personalized treatment strategy.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":"3864-3880"},"PeriodicalIF":2.9,"publicationDate":"2024-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11567801/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142072098","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
PTEN Loss Shapes Macrophage Dynamics in High-Grade Serous Ovarian Carcinoma. PTEN缺失会影响高级别浆液性卵巢癌中巨噬细胞的动态变化。
IF 12.5 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-15 DOI: 10.1158/0008-5472.CAN-23-3890
Sarah Spear, Olivia Le Saux, Hasan B Mirza, Nayana Iyer, Katie Tyson, Fabio Grundland Freile, Josephine B Walton, Chloé Woodman, Sheba Jarvis, Darren P Ennis, Carmen Aguirre Hernandez, Yuewei Xu, Pavlina Spiliopoulou, James D Brenton, Ana P Costa-Pereira, David P Cook, Barbara C Vanderhyden, Hector C Keun, Evangelos Triantafyllou, James N Arnold, Iain A McNeish

High-grade serous ovarian carcinoma (HGSC) remains a disease with poor prognosis that is unresponsive to current immune checkpoint inhibitors. Although PI3K pathway alterations, such as PTEN loss, are common in HGSC, attempts to target this pathway have been unsuccessful. We hypothesized that aberrant PI3K pathway activation may alter the HGSC immune microenvironment and present a targeting opportunity. Single-cell RNA sequencing identified populations of resident macrophages specific to Pten-null omental tumors in murine models, which were confirmed by flow cytometry. These macrophages were derived from peritoneal fluid macrophages and exhibited a unique gene expression program, marked by high expression of the enzyme heme oxygenase-1 (HMOX1). Targeting resident peritoneal macrophages prevented the appearance of HMOX1hi macrophages and reduced tumor growth. In addition, direct inhibition of HMOX1 extended survival in vivo. RNA sequencing identified IL33 in Pten-null tumor cells as a likely candidate driver, leading to the appearance of HMOX1hi macrophages. Human HGSC tumors also contained HMOX1hi macrophages with a corresponding gene expression program. Moreover, the presence of these macrophages was correlated with activated tumoral PI3K/mTOR signaling and poor overall survival in patients with HGSC. In contrast, tumors with low numbers of HMOX1hi macrophages were marked by increased adaptive immune response gene expression. These data suggest targeting HMOX1hi macrophages as a potential therapeutic strategy for treating poor prognosis HGSC. Significance: Macrophages with elevated HMOX1 expression are enriched in PTEN-deficient high-grade serous ovarian carcinoma, promote tumor growth, and represent a potential therapeutic target.

高分化浆液性卵巢癌(HGSC)仍然是一种预后不良的疾病,对目前的免疫检查点抑制剂无反应。虽然PI3K通路的改变(如PTEN缺失)在HGSC中很常见,但针对这一通路的尝试一直没有成功。我们推测,PI3K通路的异常激活可能会改变HGSC的免疫微环境,并带来靶向机会。单细胞 RNA 测序确定了小鼠模型中 Pten 缺失网膜肿瘤特异性的常驻巨噬细胞群,流式细胞术证实了这一点。这些巨噬细胞来自腹腔液巨噬细胞,具有独特的基因表达程序,其特征是高表达血红素加氧酶-1(HMOX1)。靶向腹腔巨噬细胞可阻止 HMOX1hi 巨噬细胞的出现,并减少肿瘤的生长。此外,直接抑制 HMOX1 还能延长体内存活时间。RNA测序发现,Pten-null肿瘤细胞中的IL33可能是导致HMOX1hi巨噬细胞出现的候选驱动因子。人类 HGSC 肿瘤也含有 HMOX1hi 巨噬细胞和相应的基因表达程序。此外,这些巨噬细胞的存在与肿瘤 PI3K/mTOR 信号的激活和 HGSC 患者的总生存率低有关。相比之下,HMOX1hi 巨噬细胞数量少的肿瘤适应性免疫反应基因表达增加。这些数据表明,靶向 HMOX1hi 巨噬细胞是治疗预后不良的 HGSC 的一种潜在治疗策略。
{"title":"PTEN Loss Shapes Macrophage Dynamics in High-Grade Serous Ovarian Carcinoma.","authors":"Sarah Spear, Olivia Le Saux, Hasan B Mirza, Nayana Iyer, Katie Tyson, Fabio Grundland Freile, Josephine B Walton, Chloé Woodman, Sheba Jarvis, Darren P Ennis, Carmen Aguirre Hernandez, Yuewei Xu, Pavlina Spiliopoulou, James D Brenton, Ana P Costa-Pereira, David P Cook, Barbara C Vanderhyden, Hector C Keun, Evangelos Triantafyllou, James N Arnold, Iain A McNeish","doi":"10.1158/0008-5472.CAN-23-3890","DOIUrl":"10.1158/0008-5472.CAN-23-3890","url":null,"abstract":"<p><p>High-grade serous ovarian carcinoma (HGSC) remains a disease with poor prognosis that is unresponsive to current immune checkpoint inhibitors. Although PI3K pathway alterations, such as PTEN loss, are common in HGSC, attempts to target this pathway have been unsuccessful. We hypothesized that aberrant PI3K pathway activation may alter the HGSC immune microenvironment and present a targeting opportunity. Single-cell RNA sequencing identified populations of resident macrophages specific to Pten-null omental tumors in murine models, which were confirmed by flow cytometry. These macrophages were derived from peritoneal fluid macrophages and exhibited a unique gene expression program, marked by high expression of the enzyme heme oxygenase-1 (HMOX1). Targeting resident peritoneal macrophages prevented the appearance of HMOX1hi macrophages and reduced tumor growth. In addition, direct inhibition of HMOX1 extended survival in vivo. RNA sequencing identified IL33 in Pten-null tumor cells as a likely candidate driver, leading to the appearance of HMOX1hi macrophages. Human HGSC tumors also contained HMOX1hi macrophages with a corresponding gene expression program. Moreover, the presence of these macrophages was correlated with activated tumoral PI3K/mTOR signaling and poor overall survival in patients with HGSC. In contrast, tumors with low numbers of HMOX1hi macrophages were marked by increased adaptive immune response gene expression. These data suggest targeting HMOX1hi macrophages as a potential therapeutic strategy for treating poor prognosis HGSC. Significance: Macrophages with elevated HMOX1 expression are enriched in PTEN-deficient high-grade serous ovarian carcinoma, promote tumor growth, and represent a potential therapeutic target.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":" ","pages":"3772-3787"},"PeriodicalIF":12.5,"publicationDate":"2024-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7616669/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142072100","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Conditional Activation of c-MYC in Distinct Catecholaminergic Cells Drives Development of Neuroblastoma or Somatostatinoma c-MYC在不同儿茶酚胺能细胞中的条件性激活驱动神经母细胞瘤或体细胞脂肪瘤的发育
IF 11.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-12 DOI: 10.1158/0008-5472.can-24-1142
Tingting Wang, Lingling Liu, Jie Fang, Hongjian Jin, Sivaraman Natarajan, Heather Sheppard, Meifen Lu, Gregory Turner, Thomas Confer, Melissa Johnson, Jeffrey Steinberg, Larry Ha, Nour Yadak, Richa Jain, David J. Picketts, Xiaotu Ma, Andrew Murphy, Andrew M. Davidoff, Evan S. Glazer, John Easton, Xiang Chen, Ruoning Wang, Jun Yang
c-MYC is an important driver of high-risk neuroblastoma. A lack of c-MYC–driven genetically engineered mouse models (GEMM) has hampered the ability to better understand mechanisms of neuroblastoma oncogenesis and to develop effective therapies. Here, we showed that conditional c-MYC induction via Cre recombinase driven by a tyrosine hydroxylase (Th) promoter led to a preponderance of PDX1+ somatostatinoma, a type of pancreatic neuroendocrine tumor (PNET). However, c-MYC activation via an improved Cre recombinase driven by a dopamine β-hydroxylase (Dbh) promoter resulted in neuroblastoma development. The c-MYC murine neuroblastoma tumors recapitulated the pathologic and genetic features of human neuroblastoma and responded to anti-GD2 immunotherapy and DFMO, an FDA-approved inhibitor targeting the MYC transcriptional target ODC1. Thus, c-MYC overexpression results in different but related tumor types depending on the targeted cell. The GEMMs represent valuable tools for testing immunotherapies and targeted therapies for these diseases.
c-MYC是高危神经母细胞瘤的重要驱动因素。由于缺乏c-MYC驱动的基因工程小鼠模型(GEMM),人们无法更好地了解神经母细胞瘤的致癌机制,也无法开发出有效的疗法。在这里,我们发现,通过酪氨酸羟化酶(Th)启动子驱动的Cre重组酶进行条件性c-MYC诱导,会导致PDX1+体节细胞瘤(一种胰腺神经内分泌肿瘤(PNET))的发生。然而,通过多巴胺β-羟化酶(Dbh)启动子驱动的改良Cre重组酶激活c-MYC,则会导致神经母细胞瘤的发生。c-MYC小鼠神经母细胞瘤肿瘤再现了人类神经母细胞瘤的病理和遗传特征,并对抗GD2免疫疗法和DFMO(一种FDA批准的针对MYC转录靶标ODC1的抑制剂)有反应。因此,c-MYC 过表达会导致不同但相关的肿瘤类型,具体取决于靶细胞。GEMMs是测试针对这些疾病的免疫疗法和靶向疗法的宝贵工具。
{"title":"Conditional Activation of c-MYC in Distinct Catecholaminergic Cells Drives Development of Neuroblastoma or Somatostatinoma","authors":"Tingting Wang, Lingling Liu, Jie Fang, Hongjian Jin, Sivaraman Natarajan, Heather Sheppard, Meifen Lu, Gregory Turner, Thomas Confer, Melissa Johnson, Jeffrey Steinberg, Larry Ha, Nour Yadak, Richa Jain, David J. Picketts, Xiaotu Ma, Andrew Murphy, Andrew M. Davidoff, Evan S. Glazer, John Easton, Xiang Chen, Ruoning Wang, Jun Yang","doi":"10.1158/0008-5472.can-24-1142","DOIUrl":"https://doi.org/10.1158/0008-5472.can-24-1142","url":null,"abstract":"c-MYC is an important driver of high-risk neuroblastoma. A lack of c-MYC–driven genetically engineered mouse models (GEMM) has hampered the ability to better understand mechanisms of neuroblastoma oncogenesis and to develop effective therapies. Here, we showed that conditional c-MYC induction via Cre recombinase driven by a tyrosine hydroxylase (Th) promoter led to a preponderance of PDX1+ somatostatinoma, a type of pancreatic neuroendocrine tumor (PNET). However, c-MYC activation via an improved Cre recombinase driven by a dopamine β-hydroxylase (Dbh) promoter resulted in neuroblastoma development. The c-MYC murine neuroblastoma tumors recapitulated the pathologic and genetic features of human neuroblastoma and responded to anti-GD2 immunotherapy and DFMO, an FDA-approved inhibitor targeting the MYC transcriptional target ODC1. Thus, c-MYC overexpression results in different but related tumor types depending on the targeted cell. The GEMMs represent valuable tools for testing immunotherapies and targeted therapies for these diseases.","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":"35 1","pages":""},"PeriodicalIF":11.2,"publicationDate":"2024-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142601095","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ferroptotic Neutrophils Induce Immunosuppression and Chemoresistance in Breast Cancer 铁性中性粒细胞诱导乳腺癌的免疫抑制和耐药性
IF 11.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-12 DOI: 10.1158/0008-5472.can-24-1941
Wenfeng Zeng, Ruihua Zhang, Penghan Huang, Minxia Chen, Houying Chen, Xin Zeng, Jiang Liu, Jiahui Zhang, Di Huang, Liyan Lao
Inducing ferroptosis in tumor cells is emerging as a strategy for treating malignancies that are refractory to traditional treatment modalities. However, the consequences of ferroptosis of immune cells in the tumor microenvironment (TME) need to be better understood in order to realize the potential of this approach. In this study, we discovered that neutrophils in chemoresistant breast cancer are highly sensitive to ferroptosis. Reduction of the acyltransferase MBOAT1 in chemoresistance-associated neutrophils induced phospholipid reprogramming, switching the preference from monounsaturated fatty acids to polyunsaturated fatty acids, which increased their susceptibility to ferroptosis. Ferroptotic neutrophils secreted PGE2, IDO and oxidized lipids that suppressed the proliferation and cytotoxicity of antitumor CD8+ T cells. Furthermore, neutrophil ferroptosis was closely related to a distinct subset of IL-1beta+CXCL3+CD4+ (Fer-CD4) T lymphocytes, which were enriched in chemoresistant tumors. Fer-CD4 T cells orchestrated neutrophil ferroptosis by modulating MBOAT1 expression via IL-1beta/IL-1R1/NF-kappaB signaling. Moreover, Fer-CD4 T cells secreted CXCL3, IL-8 and S100A9 to replenish the neutrophil pool in the TME. Ferroptotic neutrophils in turn fostered Fer-CD4 T cell differentiation. In spontaneous tumorigenesis mouse models, targeting IL-1beta+ CD4+ T cells or IL-1R1+ neutrophils broke the crosstalk, restraining neutrophil ferroptosis, enhancing antitumor immunity, and overcoming chemoresistance. Overall, these findings uncover the role of neutrophil ferroptosis in shaping the immune landscape and propose appealing targets for restoring immunosurveillance and chemosensitivity in breast cancer.
诱导肿瘤细胞的铁变态反应正在成为治疗对传统治疗方法难治的恶性肿瘤的一种策略。然而,为了实现这种方法的潜力,我们需要更好地了解肿瘤微环境(TME)中免疫细胞铁突变的后果。在这项研究中,我们发现化疗耐药乳腺癌中的中性粒细胞对嗜铁细胞高度敏感。化疗耐药性相关中性粒细胞中酰基转移酶 MBOAT1 的减少诱导了磷脂的重编程,使其从偏好单不饱和脂肪酸转向多不饱和脂肪酸,从而增加了它们对铁凋亡的敏感性。铁变态中性粒细胞分泌 PGE2、IDO 和氧化脂质,抑制了抗肿瘤 CD8+ T 细胞的增殖和细胞毒性。此外,中性粒细胞铁嗜性与IL-1β+CXCL3+CD4+(Fer-CD4)T淋巴细胞的独特亚群密切相关,后者在化疗耐药肿瘤中富集。Fer-CD4 T细胞通过IL-1beta/IL-1R1/NF-kappaB信号调节MBOAT1的表达,从而协调中性粒细胞的铁突变。此外,Fer-CD4 T 细胞分泌 CXCL3、IL-8 和 S100A9 来补充 TME 中的中性粒细胞池。铁嗜中性粒细胞反过来又促进了 Fer-CD4 T 细胞的分化。在自发性肿瘤发生小鼠模型中,以IL-1β+ CD4+ T细胞或IL-1R1+中性粒细胞为靶点打破了串联,抑制了中性粒细胞铁嗜性,增强了抗肿瘤免疫力,克服了化疗耐药性。总之,这些发现揭示了中性粒细胞铁嗜性在形成免疫格局中的作用,并为恢复乳腺癌的免疫监视和化疗敏感性提出了有吸引力的靶点。
{"title":"Ferroptotic Neutrophils Induce Immunosuppression and Chemoresistance in Breast Cancer","authors":"Wenfeng Zeng, Ruihua Zhang, Penghan Huang, Minxia Chen, Houying Chen, Xin Zeng, Jiang Liu, Jiahui Zhang, Di Huang, Liyan Lao","doi":"10.1158/0008-5472.can-24-1941","DOIUrl":"https://doi.org/10.1158/0008-5472.can-24-1941","url":null,"abstract":"Inducing ferroptosis in tumor cells is emerging as a strategy for treating malignancies that are refractory to traditional treatment modalities. However, the consequences of ferroptosis of immune cells in the tumor microenvironment (TME) need to be better understood in order to realize the potential of this approach. In this study, we discovered that neutrophils in chemoresistant breast cancer are highly sensitive to ferroptosis. Reduction of the acyltransferase MBOAT1 in chemoresistance-associated neutrophils induced phospholipid reprogramming, switching the preference from monounsaturated fatty acids to polyunsaturated fatty acids, which increased their susceptibility to ferroptosis. Ferroptotic neutrophils secreted PGE2, IDO and oxidized lipids that suppressed the proliferation and cytotoxicity of antitumor CD8+ T cells. Furthermore, neutrophil ferroptosis was closely related to a distinct subset of IL-1beta+CXCL3+CD4+ (Fer-CD4) T lymphocytes, which were enriched in chemoresistant tumors. Fer-CD4 T cells orchestrated neutrophil ferroptosis by modulating MBOAT1 expression via IL-1beta/IL-1R1/NF-kappaB signaling. Moreover, Fer-CD4 T cells secreted CXCL3, IL-8 and S100A9 to replenish the neutrophil pool in the TME. Ferroptotic neutrophils in turn fostered Fer-CD4 T cell differentiation. In spontaneous tumorigenesis mouse models, targeting IL-1beta+ CD4+ T cells or IL-1R1+ neutrophils broke the crosstalk, restraining neutrophil ferroptosis, enhancing antitumor immunity, and overcoming chemoresistance. Overall, these findings uncover the role of neutrophil ferroptosis in shaping the immune landscape and propose appealing targets for restoring immunosurveillance and chemosensitivity in breast cancer.","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":"13 1","pages":""},"PeriodicalIF":11.2,"publicationDate":"2024-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142601097","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The circRNA cEMSY Induces Immunogenic Cell Death and Boosts Immunotherapy Efficacy in Lung Adenocarcinoma circRNA cEMSY能诱导免疫原性细胞死亡并提高肺腺癌的免疫治疗效果
IF 11.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-12 DOI: 10.1158/0008-5472.can-24-1484
Yijian Zhang, Xuming Song, Yipeng Feng, Yuxian Qian, Bing Chen, Te Zhang, Hui Wang, Yuzhong Chen, Xinnian Yu, Hanlin Ding, Rutao Li, Pengfe Ge, Lin Xu, Gaochao Dong, Feng Jiang
Immunogenic cell death (ICD) induces an active immune response. Activating ICD represents a potential approach to boost the anti-tumor activity of immunotherapy, highlighting the need to identify effective and safe ICD inducers. In this study, we identified a conserved, ICD-related circular RNA cEMSY by systematically screening ICD models induced by multiple cell stressors in lung adenocarcinoma (LUAD). cEMSY triggered ICD in LUAD both in vitro and in vivo, leading to the release of damage-associated molecular patterns and promoting T cell cross-priming by dendritic cells (DCs). Notably, the intratumoral delivery of lipid nanoparticle-encapsulated cEMSY induced a potent antitumor immune response in an immunosuppressed tumor model, which synergized with PD-1 blockade to facilitate long-term anti-tumor immunity with no apparent toxicities. Mechanistically, cEMSY mediated mitochondrial aggregation of the RNA-binding protein TDP-43 that enabled leakage of mitochondrial DNA to stimulate the cGAS–STING pathway, activating the antiviral immune response. Clinically, elevated expression of cEMSY correlated with enhanced infiltration of DCs and CD8+ T cells and favorable immunotherapy response in LUAD. Together, these findings support the dual potential of cEMSY as a target and biomarker for improving immune checkpoint inhibitor responses in LUAD.
免疫原性细胞死亡(ICD)诱导活跃的免疫反应。激活 ICD 是提高免疫疗法抗肿瘤活性的一种潜在方法,这就凸显了识别有效、安全的 ICD 诱导因子的必要性。在这项研究中,我们通过系统筛选肺腺癌(LUAD)中多种细胞应激源诱导的ICD模型,发现了一种保守的、与ICD相关的环状RNA cEMSY。cEMSY可在体外和体内触发LUAD中的ICD,导致损伤相关分子模式的释放,并促进树突状细胞(DC)对T细胞的交叉刺激。值得注意的是,在免疫抑制的肿瘤模型中,脂质纳米粒子包裹的cEMSY瘤内给药诱导了有效的抗肿瘤免疫反应,与PD-1阻断协同促进了长期抗肿瘤免疫,且无明显毒性。从机理上讲,cEMSY介导了RNA结合蛋白TDP-43的线粒体聚集,使线粒体DNA泄漏,从而刺激cGAS-STING通路,激活抗病毒免疫反应。在临床上,cEMSY表达的升高与DCs和CD8+ T细胞浸润的增强以及LUAD患者良好的免疫治疗反应相关。这些发现共同支持了 cEMSY 作为改善 LUAD 免疫检查点抑制剂反应的靶点和生物标记物的双重潜力。
{"title":"The circRNA cEMSY Induces Immunogenic Cell Death and Boosts Immunotherapy Efficacy in Lung Adenocarcinoma","authors":"Yijian Zhang, Xuming Song, Yipeng Feng, Yuxian Qian, Bing Chen, Te Zhang, Hui Wang, Yuzhong Chen, Xinnian Yu, Hanlin Ding, Rutao Li, Pengfe Ge, Lin Xu, Gaochao Dong, Feng Jiang","doi":"10.1158/0008-5472.can-24-1484","DOIUrl":"https://doi.org/10.1158/0008-5472.can-24-1484","url":null,"abstract":"Immunogenic cell death (ICD) induces an active immune response. Activating ICD represents a potential approach to boost the anti-tumor activity of immunotherapy, highlighting the need to identify effective and safe ICD inducers. In this study, we identified a conserved, ICD-related circular RNA cEMSY by systematically screening ICD models induced by multiple cell stressors in lung adenocarcinoma (LUAD). cEMSY triggered ICD in LUAD both in vitro and in vivo, leading to the release of damage-associated molecular patterns and promoting T cell cross-priming by dendritic cells (DCs). Notably, the intratumoral delivery of lipid nanoparticle-encapsulated cEMSY induced a potent antitumor immune response in an immunosuppressed tumor model, which synergized with PD-1 blockade to facilitate long-term anti-tumor immunity with no apparent toxicities. Mechanistically, cEMSY mediated mitochondrial aggregation of the RNA-binding protein TDP-43 that enabled leakage of mitochondrial DNA to stimulate the cGAS–STING pathway, activating the antiviral immune response. Clinically, elevated expression of cEMSY correlated with enhanced infiltration of DCs and CD8+ T cells and favorable immunotherapy response in LUAD. Together, these findings support the dual potential of cEMSY as a target and biomarker for improving immune checkpoint inhibitor responses in LUAD.","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":"3 1","pages":""},"PeriodicalIF":11.2,"publicationDate":"2024-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142601143","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mutant PP2A Induces IGFBP2 Secretion to Promote Development of High-Grade Uterine Cancer 突变 PP2A 诱导 IGFBP2 分泌,促进高级别子宫癌的发展
IF 11.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-12 DOI: 10.1158/0008-5472.can-24-1263
Terrance J. Haanen, Sophie Boock, Catherine G. Callahan, Irene Peris, Kaitlin P. Zawacki, Brynne Raines, Charles A. Nino, Brian Tran, Alexis Harold, Gabrielle Hodges Onishi, Matthew Hinderman, Amanda Dowdican, Wei Huang, Derek J. Taylor, Sarah E. Taylor, Mark W. Jackson, Analisa DiFeo, Caitlin M. O'Connor, Goutham Narla
Uterine serous carcinoma (USC) and uterine carcinosarcoma (UCS) tumors are uniquely aggressive, suggesting that the primary tumor is intrinsically equipped to disseminate and metastasize. Previous work identified mutational hotspots within PPP2R1A, which encodes the Aα scaffolding subunit of protein phosphatase 2A (PP2A), a heterotrimeric serine/threonine phosphatase. Two recurrent heterozygous PPP2R1A mutations, P179R and S256F, occur exclusively within high-grade subtypes of uterine cancer and can drive tumorigenesis and metastasis. Elucidation of the mechanisms by which PP2A-Aα mutants promote tumor development and progression could help identify therapeutic opportunities. Here, we showed that expression of these mutants in USC/UCS cell-lines enhanced tumor-initiating capacity, drove a hybrid epithelial-to-mesenchymal (EM) plasticity phenotype, and elevated secretion of the tumorigenic cytokine IGFBP2. Therapeutic targeting of the IGFBP2/IGF1R signaling axis using small molecules and genetic approaches resulted in marked tumor growth inhibition. Mechanistically, PP2A regulated IGFBP2 expression through the transcription factor, NF-κB, which harbors a B56 recognition motif. Collectively, these results identify a role for PP2A in regulating paracrine cancer cell signaling that can be targeted to block the initiation and metastasis of high-grade uterine cancer.
子宫浆液性癌(USC)和子宫癌肉瘤(UCS)具有独特的侵袭性,这表明原发肿瘤具有扩散和转移的内在条件。先前的研究发现了 PPP2R1A 的突变热点,该基因编码蛋白磷酸酶 2A(PP2A)的 Aα 支架亚基,PP2A 是一种异三聚丝氨酸/苏氨酸磷酸酶。PPP2R1A 的两个复发性杂合突变(P179R 和 S256F)仅发生在高级别亚型子宫癌中,可导致肿瘤发生和转移。阐明 PP2A-Aα 突变体促进肿瘤发生和发展的机制有助于发现治疗机会。在这里,我们发现这些突变体在USC/UCS细胞系中的表达增强了肿瘤诱发能力,驱动了上皮-间质(EM)混合可塑性表型,并提高了致瘤细胞因子IGFBP2的分泌。利用小分子和基因方法对IGFBP2/IGF1R信号轴进行靶向治疗可明显抑制肿瘤生长。从机理上讲,PP2A通过转录因子NF-κB调节IGFBP2的表达,而NF-κB含有一个B56识别基团。总之,这些研究结果确定了 PP2A 在调节癌细胞旁分泌信号中的作用,可以以此为靶点阻断高级别子宫癌的发生和转移。
{"title":"Mutant PP2A Induces IGFBP2 Secretion to Promote Development of High-Grade Uterine Cancer","authors":"Terrance J. Haanen, Sophie Boock, Catherine G. Callahan, Irene Peris, Kaitlin P. Zawacki, Brynne Raines, Charles A. Nino, Brian Tran, Alexis Harold, Gabrielle Hodges Onishi, Matthew Hinderman, Amanda Dowdican, Wei Huang, Derek J. Taylor, Sarah E. Taylor, Mark W. Jackson, Analisa DiFeo, Caitlin M. O'Connor, Goutham Narla","doi":"10.1158/0008-5472.can-24-1263","DOIUrl":"https://doi.org/10.1158/0008-5472.can-24-1263","url":null,"abstract":"Uterine serous carcinoma (USC) and uterine carcinosarcoma (UCS) tumors are uniquely aggressive, suggesting that the primary tumor is intrinsically equipped to disseminate and metastasize. Previous work identified mutational hotspots within PPP2R1A, which encodes the Aα scaffolding subunit of protein phosphatase 2A (PP2A), a heterotrimeric serine/threonine phosphatase. Two recurrent heterozygous PPP2R1A mutations, P179R and S256F, occur exclusively within high-grade subtypes of uterine cancer and can drive tumorigenesis and metastasis. Elucidation of the mechanisms by which PP2A-Aα mutants promote tumor development and progression could help identify therapeutic opportunities. Here, we showed that expression of these mutants in USC/UCS cell-lines enhanced tumor-initiating capacity, drove a hybrid epithelial-to-mesenchymal (EM) plasticity phenotype, and elevated secretion of the tumorigenic cytokine IGFBP2. Therapeutic targeting of the IGFBP2/IGF1R signaling axis using small molecules and genetic approaches resulted in marked tumor growth inhibition. Mechanistically, PP2A regulated IGFBP2 expression through the transcription factor, NF-κB, which harbors a B56 recognition motif. Collectively, these results identify a role for PP2A in regulating paracrine cancer cell signaling that can be targeted to block the initiation and metastasis of high-grade uterine cancer.","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":"127 1","pages":""},"PeriodicalIF":11.2,"publicationDate":"2024-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142601096","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
An Autophagy-Targeting Chimera Induces Degradation of Androgen Receptor Mutants and AR-v7 in Castration-Resistant Prostate Cancer 自噬靶向嵌合体可诱导阉割耐药前列腺癌中雄激素受体突变体和 AR-v7 的降解
IF 11.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-12 DOI: 10.1158/0008-5472.can-24-0591
Tae Hyun Bae, Ki Woon Sung, Tri M. Pham, Abdo J. Najy, Alaleh Zamiri, Hyejeong Jang, Su Ran Mun, Seongho Kim, Ha Kyoung Kwon, Yeon Sung Son, Dongping Shi, Steven Kregel, Elisabeth I. Heath, Michael L. Cher, Yong Tae Kwon, Hyeong-Reh C. Kim
Genetic alterations play a pivotal role in various human diseases, particularly cancer. The androgen receptor (AR) is a crucial transcription factor driving prostate cancer (PCa) progression across all stages. Current AR-targeting therapies utilize competitive AR antagonists or pathway suppressors. However, therapy resistance often emerges due to AR mutations and AR splice variants, such as AR-v7. To overcome this, we developed ATC-324, an AR degrader using the innovative protein degradation technology platform AUTOphagy-TArgeting Chimera (AUTOTAC). ATC-324 was designed to comprise enzalutamide, an AR inhibitor, as a target-binding ligand and YT 6-2, a ligand of the autophagy receptor p62/SQSTM1, as an autophagy-targeting ligand. ATC-324 induces the formation of the AR/p62 complex, leading to autophagy-lysosomal degradation of AR. Importantly, ATC-324 effectively degrades AR mutants frequently detected in PCa and co-degrades AR-v7 as a heterodimer with full-length AR. ATC-324 reduces nuclear AR levels and downregulates the target gene expression of AR and AR-v7, leading to cytotoxicity in AR-positive PCa cells. We also provide evidence of the therapeutic potential of ATC-324 in vivo as well as ex vivo bone organ culture. Moreover, ATC-324 remains potent in enzalutamide-resistant PCa cells. These results demonstrate the potential of the AUTOTAC platform to target previously considered undruggable proteins and overcome certain drug resistance mechanisms.
基因改变在各种人类疾病,尤其是癌症中起着举足轻重的作用。雄激素受体(AR)是驱动前列腺癌(PCa)各阶段进展的关键转录因子。目前的 AR 靶向疗法利用竞争性 AR 拮抗剂或通路抑制剂。然而,由于AR突变和AR剪接变体(如AR-v7),常常会出现耐药性。为了克服这一问题,我们利用创新性蛋白质降解技术平台 AUTOphagy-TArgeting Chimera(AUTOTAC)开发了一种 AR 降解剂 ATC-324。ATC-324 由 AR 抑制剂恩杂鲁胺和自噬受体 p62/SQSTM1 的配体 YT 6-2 组成,前者是目标结合配体,后者是自噬靶向配体。ATC-324 可诱导 AR/p62 复合物的形成,从而导致 AR 的自噬-溶酶体降解。重要的是,ATC-324 能有效降解 PCa 中经常检测到的 AR 突变体,并以异源二聚体的形式与全长 AR 共同降解 AR-v7。ATC-324 可降低核 AR 水平,下调 AR 和 AR-v7 的靶基因表达,从而对 AR 阳性 PCa 细胞产生细胞毒性。我们还提供了 ATC-324 在体内和体外骨器官培养中的治疗潜力证据。此外,ATC-324 对恩扎鲁胺耐药的 PCa 细胞仍然有效。这些结果证明了 AUTOTAC 平台的潜力,它可以靶向以前被认为是不可药用的蛋白质,并克服某些耐药机制。
{"title":"An Autophagy-Targeting Chimera Induces Degradation of Androgen Receptor Mutants and AR-v7 in Castration-Resistant Prostate Cancer","authors":"Tae Hyun Bae, Ki Woon Sung, Tri M. Pham, Abdo J. Najy, Alaleh Zamiri, Hyejeong Jang, Su Ran Mun, Seongho Kim, Ha Kyoung Kwon, Yeon Sung Son, Dongping Shi, Steven Kregel, Elisabeth I. Heath, Michael L. Cher, Yong Tae Kwon, Hyeong-Reh C. Kim","doi":"10.1158/0008-5472.can-24-0591","DOIUrl":"https://doi.org/10.1158/0008-5472.can-24-0591","url":null,"abstract":"Genetic alterations play a pivotal role in various human diseases, particularly cancer. The androgen receptor (AR) is a crucial transcription factor driving prostate cancer (PCa) progression across all stages. Current AR-targeting therapies utilize competitive AR antagonists or pathway suppressors. However, therapy resistance often emerges due to AR mutations and AR splice variants, such as AR-v7. To overcome this, we developed ATC-324, an AR degrader using the innovative protein degradation technology platform AUTOphagy-TArgeting Chimera (AUTOTAC). ATC-324 was designed to comprise enzalutamide, an AR inhibitor, as a target-binding ligand and YT 6-2, a ligand of the autophagy receptor p62/SQSTM1, as an autophagy-targeting ligand. ATC-324 induces the formation of the AR/p62 complex, leading to autophagy-lysosomal degradation of AR. Importantly, ATC-324 effectively degrades AR mutants frequently detected in PCa and co-degrades AR-v7 as a heterodimer with full-length AR. ATC-324 reduces nuclear AR levels and downregulates the target gene expression of AR and AR-v7, leading to cytotoxicity in AR-positive PCa cells. We also provide evidence of the therapeutic potential of ATC-324 in vivo as well as ex vivo bone organ culture. Moreover, ATC-324 remains potent in enzalutamide-resistant PCa cells. These results demonstrate the potential of the AUTOTAC platform to target previously considered undruggable proteins and overcome certain drug resistance mechanisms.","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":"33 1","pages":""},"PeriodicalIF":11.2,"publicationDate":"2024-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142601098","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Loss of CDKN2A Enhances the Efficacy of Immunotherapy in EGFR Mutant Non-Small Cell Lung Cancer CDKN2A 缺失可增强表皮生长因子受体突变非小细胞肺癌免疫疗法的疗效
IF 11.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-11-08 DOI: 10.1158/0008-5472.can-24-1817
Simeng Wang, Jia-Cheng Lai, Yu Li, Chengfang Tang, Jiajia Lu, Min Han, Xianjiang Ye, Lina Jia, Wei Cui, Jingyu Yang, Chunfu Wu, Lihui Wang
Mutant epidermal growth factor receptor (EGFR) is a common driver of non-small cell lung cancer (NSCLC). While mutant EGFR has been reported to limit the efficacy of immunotherapy, a subset of EGFR mutant NSCLC patients benefit from treatment with immune checkpoint inhibitors. A better understanding of how co-occurring genomic alterations in oncogenic driver genes impact immunotherapy efficacy may provide a more complete understanding of cancer heterogeneity and identify biomarkers of response. Here, we investigated the effects of frequent EGFR co-mutations in EGFR mutant lung cancer models and identified loss-of-function mutation of CDKN2A as a potential sensitizer to anti-PD-1 treatment in vitro and in vivo. Mechanistically, CDKN2A loss impacted the composition of the tumor immune microenvironment (TIME) by promoting the expression of PD-L2 through reduced ubiquitination of c-Myc, and mutant EGFR cooperated to upregulate c-Myc and PD-L2 by activating the MAPK pathway. Blocking PD-L2 induced anti-tumor immune responses mediated by CD8+ T cells in EGFR/CDKN2A co-mutated lung cancer. Importantly, a small-molecule PD-L2 inhibitor, zinc undecylenate, remodeled the TIME of EGFR/CDKN2A co-mutant tumors and enhanced the anti-tumor efficacy of EGFR-tyrosine kinase inhibitors. Collectively, these results identify EGFR/CDKN2A co-mutation as a distinct subtype of NSCLC that shows superior sensitivity to immune checkpoint blockade and reveals a potential combined therapeutic strategy for treating this NSCLC subtype.
表皮生长因子受体(EGFR)突变是非小细胞肺癌(NSCLC)的常见诱因。据报道,突变表皮生长因子受体限制了免疫疗法的疗效,但也有一部分表皮生长因子受体突变的非小细胞肺癌患者受益于免疫检查点抑制剂的治疗。如果能更好地了解致癌驱动基因中同时出现的基因组改变如何影响免疫疗法的疗效,就能更全面地了解癌症的异质性并确定反应的生物标志物。在这里,我们研究了表皮生长因子受体(EGFR)突变肺癌模型中频繁发生的EGFR共突变的影响,并发现CDKN2A功能缺失突变是体外和体内抗PD-1治疗的潜在增敏剂。从机理上讲,CDKN2A的缺失通过减少c-Myc的泛素化促进PD-L2的表达,从而影响肿瘤免疫微环境(TIME)的组成,而突变的表皮生长因子受体则通过激活MAPK通路协同上调c-Myc和PD-L2。在表皮生长因子受体/CDKN2A共同突变的肺癌中,阻断PD-L2可诱导CD8+ T细胞介导的抗肿瘤免疫反应。重要的是,小分子 PD-L2 抑制剂十一烯酸锌能重塑表皮生长因子受体/CDKN2A 共突变肿瘤的 TIME,增强表皮生长因子受体酪氨酸激酶抑制剂的抗肿瘤疗效。总之,这些结果确定了表皮生长因子受体/CDKN2A共突变是一种独特的NSCLC亚型,它对免疫检查点阻断剂显示出更高的敏感性,并揭示了治疗这种NSCLC亚型的潜在联合治疗策略。
{"title":"Loss of CDKN2A Enhances the Efficacy of Immunotherapy in EGFR Mutant Non-Small Cell Lung Cancer","authors":"Simeng Wang, Jia-Cheng Lai, Yu Li, Chengfang Tang, Jiajia Lu, Min Han, Xianjiang Ye, Lina Jia, Wei Cui, Jingyu Yang, Chunfu Wu, Lihui Wang","doi":"10.1158/0008-5472.can-24-1817","DOIUrl":"https://doi.org/10.1158/0008-5472.can-24-1817","url":null,"abstract":"Mutant epidermal growth factor receptor (EGFR) is a common driver of non-small cell lung cancer (NSCLC). While mutant EGFR has been reported to limit the efficacy of immunotherapy, a subset of EGFR mutant NSCLC patients benefit from treatment with immune checkpoint inhibitors. A better understanding of how co-occurring genomic alterations in oncogenic driver genes impact immunotherapy efficacy may provide a more complete understanding of cancer heterogeneity and identify biomarkers of response. Here, we investigated the effects of frequent EGFR co-mutations in EGFR mutant lung cancer models and identified loss-of-function mutation of CDKN2A as a potential sensitizer to anti-PD-1 treatment in vitro and in vivo. Mechanistically, CDKN2A loss impacted the composition of the tumor immune microenvironment (TIME) by promoting the expression of PD-L2 through reduced ubiquitination of c-Myc, and mutant EGFR cooperated to upregulate c-Myc and PD-L2 by activating the MAPK pathway. Blocking PD-L2 induced anti-tumor immune responses mediated by CD8+ T cells in EGFR/CDKN2A co-mutated lung cancer. Importantly, a small-molecule PD-L2 inhibitor, zinc undecylenate, remodeled the TIME of EGFR/CDKN2A co-mutant tumors and enhanced the anti-tumor efficacy of EGFR-tyrosine kinase inhibitors. Collectively, these results identify EGFR/CDKN2A co-mutation as a distinct subtype of NSCLC that shows superior sensitivity to immune checkpoint blockade and reveals a potential combined therapeutic strategy for treating this NSCLC subtype.","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":"84 1","pages":""},"PeriodicalIF":11.2,"publicationDate":"2024-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142597192","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cancer research
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