首页 > 最新文献

Molecular Cancer Therapeutics最新文献

英文 中文
A small molecule BCL6 inhibitor effectively suppresses diffuse large B cell lymphoma cells growth. 一种小分子 BCL6 抑制剂能有效抑制弥漫大 B 细胞淋巴瘤细胞的生长。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-10-10 DOI: 10.1158/1535-7163.MCT-23-0830
Yajing Xing, Weikai Guo, Min Wu, Jiuqing Xie, Dongxia Huang, Pan Hu, Miaoran Zhou, Lin Zhang, Yadong Zhong, Mingyao Liu, Yihua Chen, Zhengfang Yi

The B-cell lymphoma 6 (BCL6) transcription factor plays a key role in establishment of germinal center (GC) formation. Diffuse large B cell lymphoma (DLBCL) originates from the GC reaction due to dysregulation of BCL6. Disrupting BCL6 and its corepressors interaction has become the foundation for rationally designing lymphoma therapies. However, BCL6 inhibitors with good activities in vitro and in vivo are rare and there are no clinically approved BCL6 inhibitors. Here, we discovered and developed a novel range of [1,2,4] triazolo[1,5-a] pyrimidine derivatives targeting BCL6/SMRT interaction. The analogue WK692 directly bound BCL6BTB, disrupted BCL6BTB/SMRT interaction and activated the expression of BCL6 downstream genes inside cells, inhibited DLBCL growth and induced apoptosis in vitro, inhibited GC formation, decreased proportion of follicular helper T (Tfh) cells and impaired immunoglobulin affinity maturation. Further studies showed that WK692 inhibited the DLBCL growth without toxic effects in vivo and synergizes with the EZH2 and PRMT5 inhibitors. Our results demonstrated that WK692 as a BCL6 inhibitor may be developed as a novel potential anticancer agent against DLBCL.

B 细胞淋巴瘤 6(BCL6)转录因子在生殖中心(GC)的形成过程中起着关键作用。弥漫大B细胞淋巴瘤(DLBCL)起源于BCL6失调导致的生殖中心反应。破坏 BCL6 及其核心抑制因子的相互作用已成为合理设计淋巴瘤疗法的基础。然而,具有良好体外和体内活性的BCL6抑制剂并不多见,目前也没有临床批准的BCL6抑制剂。在此,我们发现并开发了一系列新型[1,2,4]三唑并[1,5-a]嘧啶衍生物,靶向BCL6/SMRT相互作用。类似物 WK692 可直接与 BCL6BTB 结合,破坏 BCL6BTB/SMRT 相互作用,激活细胞内 BCL6 下游基因的表达,在体外抑制 DLBCL 的生长并诱导其凋亡,抑制 GC 的形成,降低滤泡辅助 T(Tfh)细胞的比例,损害免疫球蛋白的亲和性成熟。进一步的研究表明,WK692能抑制DLBCL在体内的生长,且无毒性作用,并能与EZH2和PRMT5抑制剂协同作用。我们的研究结果表明,作为一种 BCL6 抑制剂,WK692 可被开发为一种潜在的新型 DLBCL 抗癌药物。
{"title":"A small molecule BCL6 inhibitor effectively suppresses diffuse large B cell lymphoma cells growth.","authors":"Yajing Xing, Weikai Guo, Min Wu, Jiuqing Xie, Dongxia Huang, Pan Hu, Miaoran Zhou, Lin Zhang, Yadong Zhong, Mingyao Liu, Yihua Chen, Zhengfang Yi","doi":"10.1158/1535-7163.MCT-23-0830","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-23-0830","url":null,"abstract":"<p><p>The B-cell lymphoma 6 (BCL6) transcription factor plays a key role in establishment of germinal center (GC) formation. Diffuse large B cell lymphoma (DLBCL) originates from the GC reaction due to dysregulation of BCL6. Disrupting BCL6 and its corepressors interaction has become the foundation for rationally designing lymphoma therapies. However, BCL6 inhibitors with good activities in vitro and in vivo are rare and there are no clinically approved BCL6 inhibitors. Here, we discovered and developed a novel range of [1,2,4] triazolo[1,5-a] pyrimidine derivatives targeting BCL6/SMRT interaction. The analogue WK692 directly bound BCL6BTB, disrupted BCL6BTB/SMRT interaction and activated the expression of BCL6 downstream genes inside cells, inhibited DLBCL growth and induced apoptosis in vitro, inhibited GC formation, decreased proportion of follicular helper T (Tfh) cells and impaired immunoglobulin affinity maturation. Further studies showed that WK692 inhibited the DLBCL growth without toxic effects in vivo and synergizes with the EZH2 and PRMT5 inhibitors. Our results demonstrated that WK692 as a BCL6 inhibitor may be developed as a novel potential anticancer agent against DLBCL.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142391892","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
Tumor integrin-targeted glucose oxidase enzyme promotes ROS-mediated cell death that combines with interferon alpha therapy for tumor control. 肿瘤整合素靶向葡萄糖氧化酶可促进 ROS 介导的细胞死亡,与干扰素 alpha疗法相结合可控制肿瘤。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-10-09 DOI: 10.1158/1535-7163.MCT-24-0163
Jordan A Stinson, Allison Sheen, Brianna M Lax, Grace N Yang, Lauren Duhamel, Luciano Santollani, Elizabeth Fink, Joseph Palmeri, K Dane Wittrup

While heightened intratumoral levels of reactive oxygen species (ROS) are typically associated with a suppressive tumor microenvironment, under certain conditions ROS contribute to tumor elimination. Treatment approaches, including some chemotherapy and radiation protocols, increase cancer cell ROS levels that influence their mechanism of cell death and subsequent recognition by the immune system. Furthermore, activated myeloid cells rapidly generate ROS upon encounter with pathogens or infected cells to eliminate disease, and recently, this effector function has been noted in cancer contexts as well. Collectively, ROS-induced cancer cell death may help initiate adaptive anti-tumor immune responses that could synergize with current approved immunotherapies, for improved control of solid tumors. In this work, we explore the use of glucose oxidase, an enzyme which produces hydrogen peroxide, a type of ROS, to therapeutically mimic the endogenous oxidative burst from myeloid cells to promote antigen generation within the tumor microenvironment. We engineer the enzyme to target pan-tumor expressed integrins both as a tumor-agnostic therapeutic approach, but also as a strategy to prolong local enzyme activity following intratumoral administration. We found the targeted enzyme potently induced cancer cell death and enhanced cross-presentation by dendritic cells in vitro, and further combined with interferon alpha for long-term tumor control in murine MC38 tumors in vivo. Optimizing the single-dose administration of this enzyme overcomes limitations with immunogenicity noted for other pro-oxidant enzyme approaches. Overall, our results suggest ROS-induced cell death can be harnessed for tumor control, and highlight the potential use of designed enzyme therapies alongside immunotherapy against cancer.

虽然肿瘤内活性氧(ROS)水平的升高通常与抑制性肿瘤微环境有关,但在某些情况下,ROS 也有助于肿瘤的消除。包括某些化疗和放疗方案在内的治疗方法会增加癌细胞的活性氧水平,从而影响其细胞死亡机制和免疫系统的识别。此外,活化的骨髓细胞在遇到病原体或感染细胞时会迅速产生 ROS,以消除疾病。总而言之,ROS 诱导的癌细胞死亡可能有助于启动适应性抗肿瘤免疫反应,从而与目前已获批准的免疫疗法协同作用,改善对实体瘤的控制。在这项研究中,我们探索利用葡萄糖氧化酶(一种产生过氧化氢(ROS 的一种)的酶)来治疗性地模拟髓系细胞的内源性氧化爆发,从而促进肿瘤微环境中抗原的生成。我们对这种酶进行了工程化改造,以靶向泛肿瘤表达的整合素,既作为一种肿瘤诊断治疗方法,也作为一种在瘤内给药后延长局部酶活性的策略。我们发现,这种靶向酶在体外能有效诱导癌细胞死亡并增强树突状细胞的交叉呈递,在体内与α干扰素结合还能长期控制小鼠MC38肿瘤。优化这种酶的单剂量给药克服了其他促氧化酶方法的免疫原性限制。总之,我们的研究结果表明,可以利用 ROS 诱导的细胞死亡来控制肿瘤,并强调了设计的酶疗法与抗癌免疫疗法的潜在用途。
{"title":"Tumor integrin-targeted glucose oxidase enzyme promotes ROS-mediated cell death that combines with interferon alpha therapy for tumor control.","authors":"Jordan A Stinson, Allison Sheen, Brianna M Lax, Grace N Yang, Lauren Duhamel, Luciano Santollani, Elizabeth Fink, Joseph Palmeri, K Dane Wittrup","doi":"10.1158/1535-7163.MCT-24-0163","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0163","url":null,"abstract":"<p><p>While heightened intratumoral levels of reactive oxygen species (ROS) are typically associated with a suppressive tumor microenvironment, under certain conditions ROS contribute to tumor elimination. Treatment approaches, including some chemotherapy and radiation protocols, increase cancer cell ROS levels that influence their mechanism of cell death and subsequent recognition by the immune system. Furthermore, activated myeloid cells rapidly generate ROS upon encounter with pathogens or infected cells to eliminate disease, and recently, this effector function has been noted in cancer contexts as well. Collectively, ROS-induced cancer cell death may help initiate adaptive anti-tumor immune responses that could synergize with current approved immunotherapies, for improved control of solid tumors. In this work, we explore the use of glucose oxidase, an enzyme which produces hydrogen peroxide, a type of ROS, to therapeutically mimic the endogenous oxidative burst from myeloid cells to promote antigen generation within the tumor microenvironment. We engineer the enzyme to target pan-tumor expressed integrins both as a tumor-agnostic therapeutic approach, but also as a strategy to prolong local enzyme activity following intratumoral administration. We found the targeted enzyme potently induced cancer cell death and enhanced cross-presentation by dendritic cells in vitro, and further combined with interferon alpha for long-term tumor control in murine MC38 tumors in vivo. Optimizing the single-dose administration of this enzyme overcomes limitations with immunogenicity noted for other pro-oxidant enzyme approaches. Overall, our results suggest ROS-induced cell death can be harnessed for tumor control, and highlight the potential use of designed enzyme therapies alongside immunotherapy against cancer.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142391894","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
Zelenirstat Inhibits N-Myristoyltransferases to Disrupt Src Family Kinase Signalling and Oxidative Phosphorylation Killing Acute Myeloid Leukemia Cells. Zelenirstat 可抑制 N-肉豆蔻酰转移酶,从而破坏 Src 家族激酶信号和氧化磷酸化,杀死急性髓性白血病细胞。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-10-09 DOI: 10.1158/1535-7163.MCT-24-0307
Jay M Gamma, Qiang Liu, Erwan Beauchamp, Aishwarya Iyer, Megan C Yap, Zoulika Zak, Cassidy Ekstrom, Rony Pain, Morris A Kostiuk, John R Mackey, Joseph Brandwein, Jean Cy Wang, Luc G Berthiaume

Acute myeloid leukemia (AML) is a hematological malignancy with limited treatment options and a high likelihood of recurrence after chemotherapy. We studied N-myristoylation, the myristate modification of proteins linked to survival signaling and metabolism, as a potential therapeutic target for AML. N-myristoylation is catalyzed by two N-myristoyltransferases (NMTs), NMT1 and NMT2, with varying expressions in AML cell lines and patient samples. We identified NMT2 expression as a marker for AML patient survival, and low NMT2 expression was associated with poor outcomes. We used the first-in-class pan-NMT inhibitor, zelenirstat, to investigate the role of N-myristoylation in AML. Zelenirstat effectively inhibits myristoylation in AML cell lines and patient samples, leading to degradation of Src family kinases (SFKs), induction of endoplasmic reticulum (ER) stress, apoptosis, and cell death. Zelenirstat was well tolerated in vivo and reduced the leukemic burden in an ectopic AML cell line and in multiple orthotopic AML patient-derived xenograft models. The leukemia stem cell (LSC)-enriched fractions of the hierarchical OCI-AML22 model were highly sensitive to myristoylation inhibition. Zelenirstat also impairs mitochondrial complex I and oxidative phosphorylation, which are critical for LSC survival. These findings suggest that targeting N-myristoylation with zelenirstat represents a novel therapeutic approach for AML, with promise in patients with currently poor outcomes.

急性髓性白血病(AML)是一种血液恶性肿瘤,其治疗方案有限,化疗后复发的可能性很高。我们研究了N-肉豆蔻酰化(N-myristoylation),这是一种与生存信号转导和新陈代谢相关的蛋白质肉豆蔻酸酯修饰,是治疗急性髓性白血病的潜在靶点。N-肉豆蔻酰化由两种N-肉豆蔻酰转移酶(NMTs)催化,即NMT1和NMT2,它们在AML细胞系和患者样本中的表达各不相同。我们发现 NMT2 的表达是急性髓细胞性白血病患者存活的标志物,NMT2 的低表达与不良预后有关。我们使用第一类泛NMT抑制剂泽仑司特来研究N-肉豆蔻酰化在急性髓细胞白血病中的作用。Zelenirstat能有效抑制AML细胞系和患者样本中的肉豆蔻酰化,导致Src家族激酶(SFKs)降解、诱导内质网(ER)应激、细胞凋亡和细胞死亡。Zelenirstat在体内耐受性良好,在异位急性髓细胞白血病细胞系和多个正位急性髓细胞白血病患者异种移植模型中可减少白血病负担。分层 OCI-AML22 模型中的白血病干细胞(LSC)富集部分对肉豆蔻酰化抑制高度敏感。Zelenirstat也会损害线粒体复合体I和氧化磷酸化,而线粒体复合体I和氧化磷酸化对LSC的存活至关重要。这些研究结果表明,泽仑司特以N-肉豆蔻酰化为靶点是治疗急性髓细胞性白血病的一种新方法,有望用于目前疗效不佳的患者。
{"title":"Zelenirstat Inhibits N-Myristoyltransferases to Disrupt Src Family Kinase Signalling and Oxidative Phosphorylation Killing Acute Myeloid Leukemia Cells.","authors":"Jay M Gamma, Qiang Liu, Erwan Beauchamp, Aishwarya Iyer, Megan C Yap, Zoulika Zak, Cassidy Ekstrom, Rony Pain, Morris A Kostiuk, John R Mackey, Joseph Brandwein, Jean Cy Wang, Luc G Berthiaume","doi":"10.1158/1535-7163.MCT-24-0307","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0307","url":null,"abstract":"<p><p>Acute myeloid leukemia (AML) is a hematological malignancy with limited treatment options and a high likelihood of recurrence after chemotherapy. We studied N-myristoylation, the myristate modification of proteins linked to survival signaling and metabolism, as a potential therapeutic target for AML. N-myristoylation is catalyzed by two N-myristoyltransferases (NMTs), NMT1 and NMT2, with varying expressions in AML cell lines and patient samples. We identified NMT2 expression as a marker for AML patient survival, and low NMT2 expression was associated with poor outcomes. We used the first-in-class pan-NMT inhibitor, zelenirstat, to investigate the role of N-myristoylation in AML. Zelenirstat effectively inhibits myristoylation in AML cell lines and patient samples, leading to degradation of Src family kinases (SFKs), induction of endoplasmic reticulum (ER) stress, apoptosis, and cell death. Zelenirstat was well tolerated in vivo and reduced the leukemic burden in an ectopic AML cell line and in multiple orthotopic AML patient-derived xenograft models. The leukemia stem cell (LSC)-enriched fractions of the hierarchical OCI-AML22 model were highly sensitive to myristoylation inhibition. Zelenirstat also impairs mitochondrial complex I and oxidative phosphorylation, which are critical for LSC survival. These findings suggest that targeting N-myristoylation with zelenirstat represents a novel therapeutic approach for AML, with promise in patients with currently poor outcomes.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142391896","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
Tumor-specific antigen delivery for T-cell therapy via a pH-sensitive peptide conjugate. 通过 pH 值敏感的多肽共轭物输送肿瘤特异性抗原,用于 T 细胞疗法。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-10-09 DOI: 10.1158/1535-7163.MCT-23-0809
Annali M Yurkevicz, Yanfeng Liu, Samuel G Katz, Peter M Glazer

Identifying an optimal antigen for targeted cancer therapy is challenging as the antigen landscape on cancerous tissues mimics that of healthy tissues, with few unique tumor-specific antigens identified in individual patients. pH low insertion peptides (pHLIPs) act as a unique delivery platform that can specifically target the acidic microenvironment of tumors, sparing healthy tissue in the process. We developed a pHLIP-peptide conjugate to deliver the SIINFEKL peptide, an immunogenic fragment of ovalbumin, to tumor cells in vivo. When processed intracellularly, SIINFEKL is presented for immune recognition through the major histocompatibility complex (MHC) class I pathway. We observed selective delivery of pHLIP-SIINFEKL both in vitro and in vivo using fluorescently labeled constructs. In vitro, treatment of melanoma tumor cells with pHLIP-SIINFEKL resulted in recognition by SIINFEKL-specific T cells (OT1), leading to T cell activation and effector function. Mechanistically, we show that this recognition by OT1 cells was abrogated by siRNA/shRNA knockdown of multiple components within the MHC class I pathway in the target tumor cells, indicating that an intact antigen processing pathway in the cancer cells is necessary to mediate the effect of pHLIP-directed SIINFEKL delivery. In vivo, pHLIP-SIINFEKL treatment of tumor-bearing mice resulted in recruitment of OT1 T cells and suppression of tumor growth in two syngeneic tumor models in immunocompetent mice, with no effect when mutating either the pHLIP or SIINFEKL components of the conjugate. These results suggest that pHLIP-mediated peptide delivery can be used to deliver novel artificial antigens that can be targeted by cell-based therapies.

为癌症靶向治疗确定最佳抗原具有挑战性,因为癌症组织上的抗原结构与健康组织的抗原结构相似,在患者个体中几乎没有发现独特的肿瘤特异性抗原。pH低插入肽(pHLIPs)是一种独特的递送平台,可特异性地靶向肿瘤的酸性微环境,在此过程中不会损伤健康组织。我们开发了一种pHLIP-肽共轭物,用于在体内向肿瘤细胞递送卵清蛋白的免疫原性片段SIINFEKL肽。SIINFEKL在细胞内经过处理后,可通过主要组织相容性复合体(MHC)I类途径进行免疫识别。我们使用荧光标记的构建体观察到了 pHLIP-SIINFEKL 在体外和体内的选择性递送。在体外,用pHLIP-SIINFEKL处理黑色素瘤肿瘤细胞会导致SIINFEKL特异性T细胞(OT1)的识别,从而导致T细胞的活化和效应功能。从机理上讲,我们的研究表明,靶肿瘤细胞中 MHC I 类通路中的多种成分被 siRNA/shRNA 敲除后,OT1 细胞的这种识别作用就会减弱,这表明癌细胞中完整的抗原处理通路是 pHLIP 引导的 SIINFEKL 递送产生作用的必要条件。在体内,pHLIP-SIINFEKL 对携带肿瘤的小鼠进行处理后,可招募 OT1 T 细胞,并抑制免疫功能正常小鼠的两种合成肿瘤模型中的肿瘤生长。这些结果表明,pHLIP 介导的多肽递送可用于递送新型人工抗原,细胞疗法可将其作为靶标。
{"title":"Tumor-specific antigen delivery for T-cell therapy via a pH-sensitive peptide conjugate.","authors":"Annali M Yurkevicz, Yanfeng Liu, Samuel G Katz, Peter M Glazer","doi":"10.1158/1535-7163.MCT-23-0809","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-23-0809","url":null,"abstract":"<p><p>Identifying an optimal antigen for targeted cancer therapy is challenging as the antigen landscape on cancerous tissues mimics that of healthy tissues, with few unique tumor-specific antigens identified in individual patients. pH low insertion peptides (pHLIPs) act as a unique delivery platform that can specifically target the acidic microenvironment of tumors, sparing healthy tissue in the process. We developed a pHLIP-peptide conjugate to deliver the SIINFEKL peptide, an immunogenic fragment of ovalbumin, to tumor cells in vivo. When processed intracellularly, SIINFEKL is presented for immune recognition through the major histocompatibility complex (MHC) class I pathway. We observed selective delivery of pHLIP-SIINFEKL both in vitro and in vivo using fluorescently labeled constructs. In vitro, treatment of melanoma tumor cells with pHLIP-SIINFEKL resulted in recognition by SIINFEKL-specific T cells (OT1), leading to T cell activation and effector function. Mechanistically, we show that this recognition by OT1 cells was abrogated by siRNA/shRNA knockdown of multiple components within the MHC class I pathway in the target tumor cells, indicating that an intact antigen processing pathway in the cancer cells is necessary to mediate the effect of pHLIP-directed SIINFEKL delivery. In vivo, pHLIP-SIINFEKL treatment of tumor-bearing mice resulted in recruitment of OT1 T cells and suppression of tumor growth in two syngeneic tumor models in immunocompetent mice, with no effect when mutating either the pHLIP or SIINFEKL components of the conjugate. These results suggest that pHLIP-mediated peptide delivery can be used to deliver novel artificial antigens that can be targeted by cell-based therapies.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142391895","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
Riluzole Enhancing anti-PD-1 Efficacy by Activating cGAS/STING Signaling in Colorectal Cancer. 利鲁唑通过激活结直肠癌中的cGAS/STING信号增强抗PD-1疗效
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-10-09 DOI: 10.1158/1535-7163.MCT-24-0289
Beiyuan Liang, Misbah Khan, Hayden Storts, Evan H Zhang, Xinru Zheng, Xuanxuan Xing, Hazel Claybon, Jenna Wilson, Chunjie Li, Ning Jin, Richard Fishel, Wayne O Miles, Jing J Wang

Colorectal cancer is the second leading cause of cancer mortality in the US. Although immune checkpoint blockade therapies including anti-PD-1/PD-L1 have been successful in treating a subset of colorectal cancer patients, response rates remain low. We have found that riluzole, a well-tolerated FDA-approved oral medicine for treating amyotrophic lateral sclerosis, increased intratumoral CD8+ T cells and suppressed tumor growth of colon cancer cells in syngeneic immune competent mice. Riluzole-mediated tumor suppression was dependent on the presence of CD8+ T cells. Riluzole activates the cytosolic DNA sensing cGAS/STING pathway in colon cancer cells, resulting in increased expression of interferon β (IFNβ) and IFNβ-regulated genes including CXCL10. Inhibition of ATM, but not ATR, resulted in a synergistic increase in IFNβ expression, suggesting that riluzole induces ATM-mediated damage response that contribute to cGAS/STING activation. Depletion of cGAS or STING significantly attenuated riluzole-induced expression of IFNβ and CXCL10 as well as increase of intratumoral CD8+ T cells and suppression of tumor growth. These results indicate that riluzole-mediated tumor infiltration of CD8+ T cells and attenuation of tumor growth is dependent on tumor cell intrinsic STING activation. To determine whether riluzole treatment primes the tumor microenvironment for immune checkpoint modulation, riluzole was combined with anti-PD-1 treatment. This combination showed greater efficacy than either single agent, and strongly suppressed tumor growth in vivo. Taken together, our studies indicate that riluzole activates cGAS/STING-mediated innate immune responses, which might be exploited to sensitize colorectal tumors to anti-PD-1/PD-L1 therapies. .

结直肠癌是美国癌症死亡的第二大原因。尽管包括抗PD-1/PD-L1在内的免疫检查点阻断疗法已成功治疗了一部分结直肠癌患者,但反应率仍然很低。我们发现,经 FDA 批准用于治疗肌萎缩侧索硬化症的利鲁唑是一种耐受性良好的口服药物,它能增加瘤内 CD8+ T 细胞,抑制合成免疫小鼠结肠癌细胞的肿瘤生长。利鲁唑介导的肿瘤抑制依赖于 CD8+ T 细胞的存在。利鲁唑可激活结肠癌细胞中的细胞DNA感应cGAS/STING通路,导致干扰素β(IFNβ)和IFNβ调控基因(包括CXCL10)的表达增加。抑制ATM(而非ATR)会导致IFNβ表达的协同增加,这表明利鲁唑诱导了ATM介导的损伤反应,从而促进了cGAS/STING的激活。cGAS 或 STING 的耗竭可显著降低利鲁唑诱导的 IFNβ 和 CXCL10 的表达以及瘤内 CD8+ T 细胞的增加和肿瘤生长的抑制。这些结果表明,利鲁唑介导的 CD8+ T 细胞肿瘤浸润和肿瘤生长抑制依赖于肿瘤细胞内在的 STING 激活。为了确定利鲁唑治疗是否为免疫检查点调控提供了肿瘤微环境,利鲁唑与抗PD-1治疗相结合。这种联合疗法的疗效优于任何一种单药,并能强烈抑制体内肿瘤的生长。总之,我们的研究表明,利鲁唑能激活 cGAS/STING 介导的先天性免疫反应,可以利用这种反应使结直肠肿瘤对抗 PD-1/PD-L1 疗法敏感。.
{"title":"Riluzole Enhancing anti-PD-1 Efficacy by Activating cGAS/STING Signaling in Colorectal Cancer.","authors":"Beiyuan Liang, Misbah Khan, Hayden Storts, Evan H Zhang, Xinru Zheng, Xuanxuan Xing, Hazel Claybon, Jenna Wilson, Chunjie Li, Ning Jin, Richard Fishel, Wayne O Miles, Jing J Wang","doi":"10.1158/1535-7163.MCT-24-0289","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0289","url":null,"abstract":"<p><p>Colorectal cancer is the second leading cause of cancer mortality in the US. Although immune checkpoint blockade therapies including anti-PD-1/PD-L1 have been successful in treating a subset of colorectal cancer patients, response rates remain low. We have found that riluzole, a well-tolerated FDA-approved oral medicine for treating amyotrophic lateral sclerosis, increased intratumoral CD8+ T cells and suppressed tumor growth of colon cancer cells in syngeneic immune competent mice. Riluzole-mediated tumor suppression was dependent on the presence of CD8+ T cells. Riluzole activates the cytosolic DNA sensing cGAS/STING pathway in colon cancer cells, resulting in increased expression of interferon β (IFNβ) and IFNβ-regulated genes including CXCL10. Inhibition of ATM, but not ATR, resulted in a synergistic increase in IFNβ expression, suggesting that riluzole induces ATM-mediated damage response that contribute to cGAS/STING activation. Depletion of cGAS or STING significantly attenuated riluzole-induced expression of IFNβ and CXCL10 as well as increase of intratumoral CD8+ T cells and suppression of tumor growth. These results indicate that riluzole-mediated tumor infiltration of CD8+ T cells and attenuation of tumor growth is dependent on tumor cell intrinsic STING activation. To determine whether riluzole treatment primes the tumor microenvironment for immune checkpoint modulation, riluzole was combined with anti-PD-1 treatment. This combination showed greater efficacy than either single agent, and strongly suppressed tumor growth in vivo. Taken together, our studies indicate that riluzole activates cGAS/STING-mediated innate immune responses, which might be exploited to sensitize colorectal tumors to anti-PD-1/PD-L1 therapies. .</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142391893","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
Inflammation and Immunity in Liver Neoplasms: Implications for Future Therapeutic Strategies. 肝肿瘤中的炎症和免疫:未来治疗策略的意义》。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-10-04 DOI: 10.1158/1535-7163.MCT-23-0726
Jieun Kim, Ekihiro Seki

Over the past two decades, the "hallmarks of cancer" have revolutionized cancer research and highlighted the crucial roles of inflammation and immunity. Pro-tumorigenic inflammation promotes cancer development along with inhibition of anti-tumor immunity, shaping the tumor microenvironment (TME) towards a tumor-permissive state and further enhancing the malignant potential of cancer cells. This immunosuppressive TME allows tumors to evade immunosurveillance. Thus, understanding the complex interplay between tumors and the immune system within the TME has become pivotal, especially with the advent of immunotherapy. Although immunotherapy has achieved notable success in many malignancies, primary liver cancer, particularly hepatocellular carcinoma (HCC), presents unique challenges. The hepatic immunosuppressive environment poses obstacles to the effectiveness of immunotherapy, along with high mortality rates and limited treatment options for patients with liver cancer. In this review, we discuss current understanding of the complex immune-mediated mechanisms underlying liver neoplasms, focusing on HCC and liver metastases. We describe the molecular and cellular heterogeneity within the TME, highlighting how this presents unique challenges and opportunities for immunotherapy in liver cancers. By unraveling the immune landscape of liver neoplasms, this review aims to contribute to the development of more effective therapeutic interventions, ultimately improving clinical outcomes for patients with liver cancer.

在过去二十年里,"癌症标志 "彻底改变了癌症研究,凸显了炎症和免疫的关键作用。致癌炎症促进了癌症的发展,同时抑制了抗肿瘤免疫,使肿瘤微环境(TME)趋于肿瘤容许状态,进一步提高了癌细胞的恶性潜能。这种具有免疫抑制作用的肿瘤微环境可使肿瘤逃避免疫监视。因此,了解肿瘤和免疫系统在 TME 内的复杂相互作用已变得至关重要,尤其是随着免疫疗法的出现。尽管免疫疗法在许多恶性肿瘤中都取得了显著的成功,但原发性肝癌,尤其是肝细胞癌(HCC),却面临着独特的挑战。肝脏免疫抑制环境对免疫疗法的有效性构成了障碍,而且肝癌患者的死亡率高、治疗方案有限。在这篇综述中,我们将讨论目前对肝脏肿瘤背后复杂的免疫介导机制的理解,重点关注 HCC 和肝转移。我们描述了肝转移灶内的分子和细胞异质性,强调这为肝癌的免疫疗法带来了独特的挑战和机遇。通过揭示肝脏肿瘤的免疫环境,本综述旨在促进开发更有效的治疗干预措施,最终改善肝癌患者的临床预后。
{"title":"Inflammation and Immunity in Liver Neoplasms: Implications for Future Therapeutic Strategies.","authors":"Jieun Kim, Ekihiro Seki","doi":"10.1158/1535-7163.MCT-23-0726","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-23-0726","url":null,"abstract":"<p><p>Over the past two decades, the \"hallmarks of cancer\" have revolutionized cancer research and highlighted the crucial roles of inflammation and immunity. Pro-tumorigenic inflammation promotes cancer development along with inhibition of anti-tumor immunity, shaping the tumor microenvironment (TME) towards a tumor-permissive state and further enhancing the malignant potential of cancer cells. This immunosuppressive TME allows tumors to evade immunosurveillance. Thus, understanding the complex interplay between tumors and the immune system within the TME has become pivotal, especially with the advent of immunotherapy. Although immunotherapy has achieved notable success in many malignancies, primary liver cancer, particularly hepatocellular carcinoma (HCC), presents unique challenges. The hepatic immunosuppressive environment poses obstacles to the effectiveness of immunotherapy, along with high mortality rates and limited treatment options for patients with liver cancer. In this review, we discuss current understanding of the complex immune-mediated mechanisms underlying liver neoplasms, focusing on HCC and liver metastases. We describe the molecular and cellular heterogeneity within the TME, highlighting how this presents unique challenges and opportunities for immunotherapy in liver cancers. By unraveling the immune landscape of liver neoplasms, this review aims to contribute to the development of more effective therapeutic interventions, ultimately improving clinical outcomes for patients with liver cancer.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142375651","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Vitamin A Metabolism and Resistance of Hepatic Metastases to Immunotherapy. 维生素 A 代谢与肝转移瘤对免疫疗法的抵抗力
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-10-04 DOI: 10.1158/1535-7163.MCT-24-0367
Peter C Jones, Daniel D Von Hoff

The liver is an immune tolerant organ, allowing for organ transplantation with less immune suppression compared to other organs. It also provides fertile soil for tumor metastases, which tend to be more resistant to checkpoint blockade immunotherapy than metastases in other organs. This resistance may result from the sum of incremental evolutionary adaptions in various cell types to prevent overaction to antigens absorbed from the gut into the portal circulation or it might involve a central mechanism. Here we propose that metabolism of vitamin A, which is highly concentrated in the liver, is a root source of tolerance and resistance of hepatic metastases to checkpoint blockade. Suppression of retinoic acid synthesis from vitamin A with disulfiram may mitigate tolerance and produce enhanced immunotherapy treatment results for patients with liver metastases.

肝脏是一个免疫耐受器官,与其他器官相比,它可以在较少免疫抑制的情况下进行器官移植。肝脏也为肿瘤转移提供了肥沃的土壤,与其他器官的肿瘤转移相比,肝脏往往对检查点阻断免疫疗法更具抵抗力。这种抗药性可能是各类细胞为防止对从肠道吸收到门静脉循环的抗原产生过度反应而逐步进化适应的结果,也可能涉及一种中心机制。在此,我们提出,高度集中于肝脏的维生素 A 代谢是肝转移瘤对检查点阻断耐受性和抗性的根源。用双硫仑抑制维生素 A 合成维甲酸可能会减轻肝转移患者的耐受性,并提高免疫疗法的治疗效果。
{"title":"Vitamin A Metabolism and Resistance of Hepatic Metastases to Immunotherapy.","authors":"Peter C Jones, Daniel D Von Hoff","doi":"10.1158/1535-7163.MCT-24-0367","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0367","url":null,"abstract":"<p><p>The liver is an immune tolerant organ, allowing for organ transplantation with less immune suppression compared to other organs. It also provides fertile soil for tumor metastases, which tend to be more resistant to checkpoint blockade immunotherapy than metastases in other organs. This resistance may result from the sum of incremental evolutionary adaptions in various cell types to prevent overaction to antigens absorbed from the gut into the portal circulation or it might involve a central mechanism. Here we propose that metabolism of vitamin A, which is highly concentrated in the liver, is a root source of tolerance and resistance of hepatic metastases to checkpoint blockade. Suppression of retinoic acid synthesis from vitamin A with disulfiram may mitigate tolerance and produce enhanced immunotherapy treatment results for patients with liver metastases.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142372256","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
PKMYT1 Is a Marker of Treatment Response and a Therapeutic Target for CDK4/6 Inhibitor-Resistance in ER+ Breast Cancer. PKMYT1是ER+乳腺癌治疗反应的标记物和CDK4/6抑制剂耐药性的治疗靶点
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-10-01 DOI: 10.1158/1535-7163.MCT-23-0564
Anran Chen, Beom-Jun Kim, Aparna Mitra, Craig T Vollert, Jonathan T Lei, Diana Fandino, Meenakshi Anurag, Matthew V Holt, Xuxu Gou, Jacob B Pilcher, Matthew P Goetz, Donald W Northfelt, Susan G Hilsenbeck, C Gary Marshall, Marc L Hyer, Robert Papp, Shou-Yun Yin, Carmine De Angelis, Rachel Schiff, Suzanne A W Fuqua, Cynthia X Ma, Charles E Foulds, Matthew J Ellis

Endocrine therapies (ET) with cyclin-dependent kinase 4/6 (CDK4/6) inhibition are the standard treatment for estrogen receptor-α-positive (ER+) breast cancer, however drug resistance is common. In this study, proteogenomic analyses of patient-derived xenografts (PDXs) from patients with 22 ER+ breast cancer demonstrated that protein kinase, membrane-associated tyrosine/threonine one (PKMYT1), a WEE1 homolog, is estradiol (E2) regulated in E2-dependent PDXs and constitutively expressed when growth is E2-independent. In clinical samples, high PKMYT1 mRNA levels associated with resistance to both ET and CDK4/6 inhibition. The PKMYT1 inhibitor lunresertib (RP-6306) with gemcitabine selectively and synergistically reduced the viability of ET and palbociclib-resistant ER+ breast cancer cells without functional p53. In vitro the combination increased DNA damage and apoptosis. In palbociclib-resistant, TP53 mutant PDX-derived organoids and PDXs, RP-6306 with low-dose gemcitabine induced greater tumor volume reduction compared to treatment with either single agent. Our study demonstrates the clinical potential of RP-6306 in combination with gemcitabine for ET and CDK4/6 inhibitor resistant TP53 mutant ER+ breast cancer.

抑制CDK4/6的内分泌疗法(ET)是治疗雌激素受体α阳性(ER+)乳腺癌的标准疗法,但耐药性很常见。在这项研究中,对22个ER+乳腺癌患者衍生异种移植物(PDXs)进行的蛋白质基因组学分析表明,WEE1同源物PKMYT1在依赖雌激素(E2)的PDXs中受雌二醇(E2)调控,而在不依赖E2生长的PDXs中则呈组成型表达。在临床样本中,高水平的 PKMYT1 mRNA 与对 ET 和 CDK4/6 抑制的耐受性有关。PKMYT1 抑制剂 lunresertib(RP-6306)与吉西他滨可选择性地协同降低对 ET 和帕博西尼耐药的无功能 p53 的 ER+ 乳腺癌细胞的活力。在体外,该组合可增加 DNA 损伤和细胞凋亡。在palbociclib耐药、TP53突变的PDX器官组织和异种移植物中,RP-6306与低剂量吉西他滨联合用药与单药治疗相比,能诱导更大的肿瘤体积缩小。我们的研究证明了RP-6306与吉西他滨联合治疗ET和CDK4/6抑制剂耐药的TP53突变ER+乳腺癌的临床潜力。
{"title":"PKMYT1 Is a Marker of Treatment Response and a Therapeutic Target for CDK4/6 Inhibitor-Resistance in ER+ Breast Cancer.","authors":"Anran Chen, Beom-Jun Kim, Aparna Mitra, Craig T Vollert, Jonathan T Lei, Diana Fandino, Meenakshi Anurag, Matthew V Holt, Xuxu Gou, Jacob B Pilcher, Matthew P Goetz, Donald W Northfelt, Susan G Hilsenbeck, C Gary Marshall, Marc L Hyer, Robert Papp, Shou-Yun Yin, Carmine De Angelis, Rachel Schiff, Suzanne A W Fuqua, Cynthia X Ma, Charles E Foulds, Matthew J Ellis","doi":"10.1158/1535-7163.MCT-23-0564","DOIUrl":"10.1158/1535-7163.MCT-23-0564","url":null,"abstract":"<p><p>Endocrine therapies (ET) with cyclin-dependent kinase 4/6 (CDK4/6) inhibition are the standard treatment for estrogen receptor-α-positive (ER+) breast cancer, however drug resistance is common. In this study, proteogenomic analyses of patient-derived xenografts (PDXs) from patients with 22 ER+ breast cancer demonstrated that protein kinase, membrane-associated tyrosine/threonine one (PKMYT1), a WEE1 homolog, is estradiol (E2) regulated in E2-dependent PDXs and constitutively expressed when growth is E2-independent. In clinical samples, high PKMYT1 mRNA levels associated with resistance to both ET and CDK4/6 inhibition. The PKMYT1 inhibitor lunresertib (RP-6306) with gemcitabine selectively and synergistically reduced the viability of ET and palbociclib-resistant ER+ breast cancer cells without functional p53. In vitro the combination increased DNA damage and apoptosis. In palbociclib-resistant, TP53 mutant PDX-derived organoids and PDXs, RP-6306 with low-dose gemcitabine induced greater tumor volume reduction compared to treatment with either single agent. Our study demonstrates the clinical potential of RP-6306 in combination with gemcitabine for ET and CDK4/6 inhibitor resistant TP53 mutant ER+ breast cancer.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11443213/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141087789","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
Blocking M2-Like Macrophage Polarization Using Decoy Oligodeoxynucleotide-Based Gene Therapy Prevents Immune Evasion for Pancreatic Cancer Treatment. 利用基于诱饵寡聚脱氧核苷酸的基因疗法阻止 M2 样巨噬细胞极化,防止胰腺癌治疗中的免疫逃避。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-10-01 DOI: 10.1158/1535-7163.MCT-23-0767
Chang-Jung Chen, Hao-Chen Wang, Ya-Chin Hou, Yi-Ying Wu, Chi-Chang Shieh, Yan-Shen Shan

M2-like macrophages exhibit immunosuppressive activity and promote pancreatic cancer progression. Reactive oxygen species (ROS) affect macrophage polarization; however, the mechanism remains unclear. This study aimed to elucidate the underlying molecular basis and design a gene therapy to inhibit M2-like polarization. Microarray analysis and immunofluorescence staining were performed in M1-like and M2-like macrophages to ascertain the expression of CYBB, a major intracellular ROS source. Coculture assay and syngeneic orthotopic pancreatic cancer mice models were used to study the mechanism of M2-like skewing. Decoy oligodeoxynucleotides (ODNs) were designed to manipulate CYBB transcription to inhibit M2-like polarization and control tumor growth. Lipopolysaccharide treatment polarized U937 cells to M1-like macrophages in which CYBB expression was increased. In contrast, coculture with PANC-1 cells induced M2-like polarization in U937 cells with CYBB downregulation. High CD204 M2-like expression in combination with low CYBB expression was associated with the worst prognosis in patients with pancreatic cancer. STAT6 and HDAC2 in U937 cells were activated by cancer cell-derived IL4 after coculture and then bound to the CYBB promoter to repress CYBB expression, resulting in M2-like polarization. Diphenyleneiodonium, 8λ³-iodatricyclo[7.4.0.02,⁷]trideca-1(13),2,4,6,9,11-hexaen-8-ylium chloride that inhibits ROS production could block this action. Knockdown of STAT6 and HDAC2 also inhibited M2-like polarization and maintained the M1-like phenotype of U937 cells after coculture. Decoy ODNs interrupting the binding of STAT6 to the CYBB promoter counteracted M2-like polarization and tumor growth and triggered antitumor immunity in vivo. Gene therapy using STAT6-CYBB decoy ODNs can inhibit M2-like polarization, representing a potential therapeutic tool for pancreatic cancer.

M2 样巨噬细胞具有免疫抑制活性并促进胰腺癌的进展。活性氧(ROS)会影响巨噬细胞的极化,但其机制仍不清楚。本研究旨在阐明其分子基础,并设计一种基因疗法来抑制M2样极化。研究人员对M1样和M2样巨噬细胞进行了微阵列分析和IF染色,以确定细胞内ROS的主要来源CYBB的表达。共培养试验和同种异体胰腺癌小鼠模型被用来研究 M2 样性倾斜的机制。设计了诱饵寡脱氧核苷酸(ODN)来操纵CYBB转录,从而抑制M2样极化并控制肿瘤生长。脂多糖(LPS)处理可将 U937 细胞极化为 M1 样巨噬细胞,其中 CYBB 的表达增加。相反,与PANC-1细胞共培养可诱导U937细胞M2样极化,CYBB下调。CD204 M2样高表达结合CYBB低表达与胰腺癌患者最差的预后有关。U937细胞中的STAT6和HDAC2在共培养后被癌细胞衍生的IL-4激活,然后与CYBB启动子结合,抑制CYBB的表达,导致M2样极化。抑制 ROS 生成的 DPI 可阻断这一作用。敲除 STAT6 和 HDAC2 也能抑制 M2 样极化,并在共培养后维持 U937 细胞的 M1 样表型。干扰STAT6与CYBB启动子结合的诱饵ODNs可抑制M2样极化和肿瘤生长,并在体内引发抗肿瘤免疫。使用STAT6-CYBB诱饵ODNs进行基因治疗可抑制M2样极化,是一种潜在的胰腺癌治疗工具。
{"title":"Blocking M2-Like Macrophage Polarization Using Decoy Oligodeoxynucleotide-Based Gene Therapy Prevents Immune Evasion for Pancreatic Cancer Treatment.","authors":"Chang-Jung Chen, Hao-Chen Wang, Ya-Chin Hou, Yi-Ying Wu, Chi-Chang Shieh, Yan-Shen Shan","doi":"10.1158/1535-7163.MCT-23-0767","DOIUrl":"10.1158/1535-7163.MCT-23-0767","url":null,"abstract":"<p><p>M2-like macrophages exhibit immunosuppressive activity and promote pancreatic cancer progression. Reactive oxygen species (ROS) affect macrophage polarization; however, the mechanism remains unclear. This study aimed to elucidate the underlying molecular basis and design a gene therapy to inhibit M2-like polarization. Microarray analysis and immunofluorescence staining were performed in M1-like and M2-like macrophages to ascertain the expression of CYBB, a major intracellular ROS source. Coculture assay and syngeneic orthotopic pancreatic cancer mice models were used to study the mechanism of M2-like skewing. Decoy oligodeoxynucleotides (ODNs) were designed to manipulate CYBB transcription to inhibit M2-like polarization and control tumor growth. Lipopolysaccharide treatment polarized U937 cells to M1-like macrophages in which CYBB expression was increased. In contrast, coculture with PANC-1 cells induced M2-like polarization in U937 cells with CYBB downregulation. High CD204 M2-like expression in combination with low CYBB expression was associated with the worst prognosis in patients with pancreatic cancer. STAT6 and HDAC2 in U937 cells were activated by cancer cell-derived IL4 after coculture and then bound to the CYBB promoter to repress CYBB expression, resulting in M2-like polarization. Diphenyleneiodonium, 8λ³-iodatricyclo[7.4.0.02,⁷]trideca-1(13),2,4,6,9,11-hexaen-8-ylium chloride that inhibits ROS production could block this action. Knockdown of STAT6 and HDAC2 also inhibited M2-like polarization and maintained the M1-like phenotype of U937 cells after coculture. Decoy ODNs interrupting the binding of STAT6 to the CYBB promoter counteracted M2-like polarization and tumor growth and triggered antitumor immunity in vivo. Gene therapy using STAT6-CYBB decoy ODNs can inhibit M2-like polarization, representing a potential therapeutic tool for pancreatic cancer.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11443249/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141440688","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
KRAS-Driven Tumorigenesis and KRAS-Driven Therapy in Pancreatic Adenocarcinoma. 胰腺腺癌中 KRAS 驱动的肿瘤发生和 KRAS 驱动的治疗。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-10-01 DOI: 10.1158/1535-7163.MCT-23-0519
Minh T Than, Mark O'Hara, Ben Z Stanger, Kim A Reiss

Pancreatic ductal adenocarcinoma (PDAC) is associated with significant morbidity and mortality and is projected to be the second leading cause of cancer-related deaths by 2030. Mutations in KRAS are found in the vast majority of PDAC cases and plays an important role in the development of the disease. KRAS drives tumor cell proliferation and survival through activating the MAPK pathway to drive cell cycle progression and to lead to MYC-driven cellular programs. Moreover, activated KRAS promotes a protumorigenic microenvironment through forming a desmoplastic stroma and by impairing antitumor immunity. Secretion of granulocyte-macrophage colony-stimulating factor and recruitment of myeloid-derived suppressor cells and protumorigenic macrophages results in an immunosuppressive environment while secretion of secrete sonic hedgehog and TGFβ drive fibroblastic features characteristic of PDAC. Recent development of several small molecules to directly target KRAS marks an important milestone in precision medicine. Many molecules show promise in preclinical models of PDAC and in early phase clinical trials. In this review, we discuss the underlying cell intrinsic and extrinsic roles of KRAS in PDAC tumorigenesis, the pharmacologic development of KRAS inhibition, and therapeutic strategies to target KRAS in PDAC.

胰腺导管腺癌(PDAC)与严重的发病率和死亡率有关,预计到 2030 年将成为癌症相关死亡的第二大原因。在绝大多数 PDAC 病例中都发现了 KRAS 基因突变,这种突变在疾病的发展过程中起着重要作用。KRAS 通过激活 MAPK 通路来驱动细胞周期的进展,并导致 MYC 驱动的细胞程序,从而推动肿瘤细胞的增殖和存活。此外,活化的 KRAS 还会通过形成脱鳞基质和损害抗肿瘤免疫力来促进有利于肿瘤生成的微环境。GM-CSF的分泌以及髓源性抑制细胞和亲肿瘤巨噬细胞的招募导致了免疫抑制环境,而SHH和TGF-beta的分泌则推动了PDAC特有的成纤维细胞特征。最近开发出的几种直接靶向 KRAS 的小分子药物标志着精准医疗的一个重要里程碑。许多分子在 PDAC 的临床前模型和早期临床试验中显示出前景。在这篇综述中,我们将讨论 KRAS 在 PDAC 肿瘤发生中的潜在细胞内在和外在作用、KRAS 抑制的药理学发展以及针对 PDAC 中 KRAS 的治疗策略。
{"title":"KRAS-Driven Tumorigenesis and KRAS-Driven Therapy in Pancreatic Adenocarcinoma.","authors":"Minh T Than, Mark O'Hara, Ben Z Stanger, Kim A Reiss","doi":"10.1158/1535-7163.MCT-23-0519","DOIUrl":"10.1158/1535-7163.MCT-23-0519","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) is associated with significant morbidity and mortality and is projected to be the second leading cause of cancer-related deaths by 2030. Mutations in KRAS are found in the vast majority of PDAC cases and plays an important role in the development of the disease. KRAS drives tumor cell proliferation and survival through activating the MAPK pathway to drive cell cycle progression and to lead to MYC-driven cellular programs. Moreover, activated KRAS promotes a protumorigenic microenvironment through forming a desmoplastic stroma and by impairing antitumor immunity. Secretion of granulocyte-macrophage colony-stimulating factor and recruitment of myeloid-derived suppressor cells and protumorigenic macrophages results in an immunosuppressive environment while secretion of secrete sonic hedgehog and TGFβ drive fibroblastic features characteristic of PDAC. Recent development of several small molecules to directly target KRAS marks an important milestone in precision medicine. Many molecules show promise in preclinical models of PDAC and in early phase clinical trials. In this review, we discuss the underlying cell intrinsic and extrinsic roles of KRAS in PDAC tumorigenesis, the pharmacologic development of KRAS inhibition, and therapeutic strategies to target KRAS in PDAC.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11444872/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141907075","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Molecular Cancer Therapeutics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1