首页 > 最新文献

Molecular Cancer Therapeutics最新文献

英文 中文
Pretargeted radioimmunotherapy with the novel anti-oxMIF/HSG bispecific antibody ON105 results in significant tumor regression in murine models of cancer. 新型抗oxMIF/HSG双特异性抗体ON105的预靶向放射免疫疗法可使小鼠癌症模型中的肿瘤显著消退。
IF 5.7 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-05 DOI: 10.1158/1535-7163.MCT-24-0083
Alejandro A Puchol Tarazona, Alexander Schinagl, Irina Mirkina, Gregor Rossmueller, Randolf J Kerschbaumer, Friedmund Bachmann, Michael Thiele

Radioimmunotherapy (RIT) uses mAbs to deliver radionuclides to cancer cells or the tumor microenvironment and has shown promise in treating localized and diffuse tumors. While RIT agents have gained FDA/EMA approval for certain hematological malignancies, effectiveness of RIT in treating solid tumors remains limited. Here we present PreTarg-it®, a novel approach for pretargeted radioimmunotherapy, providing optimized delivery of payloads in a two-step regimen. The effectiveness of PreTarg-it® is demonstrated by a powerful combination of ON105, a novel bispecific antibody against both oxMIF and the histamine-succinyl-glycyl (HSG) hapten, as the first component and the radioactively labeled DOTA-di-HSG peptide as the second component in murine models of cancer. Mice bearing either subcutaneous mouse colorectal CT26 or human pancreatic CFPAC-1 tumors received an intravenous injection of ON105. After ON105 had accumulated in the tumor and cleared from circulation to approximately 1-3% of its peak concentration, 177Lu-DOTA-di-HSG peptide was administered. A single PreTarg-it® treatment cycle resulted in tumor regression when mice bearing CT26 tumors were given the highest treatment dose with a pretargeting delay of three days. Administered with a 5-day interval, the highest dose arrested tumor growth in both CT26 syngrafts and CFPAC-1 xenografts. In all cases, the highest treatment dose resulted in 100% survival at the study endpoint whereas the control cohorts showed 0% and 60% survival in the CT26 and CFPAC-1 models, respectively. Therefore, PreTarg-it® holds potential as a novel and potent therapy for patients with hard-to-treat solid tumors such as pancreatic cancer, as well as those with late-stage malignancies.

放射免疫疗法(RIT)利用 mAbs 向癌细胞或肿瘤微环境释放放射性核素,在治疗局部和弥漫性肿瘤方面前景看好。虽然放射免疫疗法制剂在治疗某些血液恶性肿瘤方面已获得 FDA/EMA 批准,但放射免疫疗法在治疗实体瘤方面的效果仍然有限。在这里,我们介绍一种用于预靶向放射免疫疗法的新方法--PreTarg-it®,它能以两步疗法优化有效载荷的递送。在小鼠癌症模型中,PreTarg-it® 的有效性体现在以新型双特异性抗体 ON105 对抗 oxMIF 和组胺-琥珀酰-甘氨酰(HSG)杂合体作为第一成分,以放射性标记的 DOTA-di-HSG 肽作为第二成分的强大组合上。携带皮下小鼠结直肠癌 CT26 或人胰腺 CFPAC-1 肿瘤的小鼠静脉注射 ON105。ON105在肿瘤中蓄积并从血液循环中清除至其峰值浓度的约1-3%后,再注射177Lu-DOTA-di-HSG肽。对携带 CT26 肿瘤的小鼠施用最高剂量的 PreTarg-it® 治疗,并延迟 3 天进行预靶向治疗,单个 PreTarg-it® 治疗周期可使肿瘤消退。在间隔 5 天的情况下,最高剂量可阻止 CT26 系统移植物和 CFPAC-1 异种移植物的肿瘤生长。在所有情况下,最高治疗剂量都能使研究终点的存活率达到 100%,而对照组在 CT26 和 CFPAC-1 模型中的存活率分别为 0% 和 60%。因此,PreTarg-it® 有潜力成为胰腺癌等难以治疗的实体瘤患者以及晚期恶性肿瘤患者的新型强效疗法。
{"title":"Pretargeted radioimmunotherapy with the novel anti-oxMIF/HSG bispecific antibody ON105 results in significant tumor regression in murine models of cancer.","authors":"Alejandro A Puchol Tarazona, Alexander Schinagl, Irina Mirkina, Gregor Rossmueller, Randolf J Kerschbaumer, Friedmund Bachmann, Michael Thiele","doi":"10.1158/1535-7163.MCT-24-0083","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0083","url":null,"abstract":"<p><p>Radioimmunotherapy (RIT) uses mAbs to deliver radionuclides to cancer cells or the tumor microenvironment and has shown promise in treating localized and diffuse tumors. While RIT agents have gained FDA/EMA approval for certain hematological malignancies, effectiveness of RIT in treating solid tumors remains limited. Here we present PreTarg-it®, a novel approach for pretargeted radioimmunotherapy, providing optimized delivery of payloads in a two-step regimen. The effectiveness of PreTarg-it® is demonstrated by a powerful combination of ON105, a novel bispecific antibody against both oxMIF and the histamine-succinyl-glycyl (HSG) hapten, as the first component and the radioactively labeled DOTA-di-HSG peptide as the second component in murine models of cancer. Mice bearing either subcutaneous mouse colorectal CT26 or human pancreatic CFPAC-1 tumors received an intravenous injection of ON105. After ON105 had accumulated in the tumor and cleared from circulation to approximately 1-3% of its peak concentration, 177Lu-DOTA-di-HSG peptide was administered. A single PreTarg-it® treatment cycle resulted in tumor regression when mice bearing CT26 tumors were given the highest treatment dose with a pretargeting delay of three days. Administered with a 5-day interval, the highest dose arrested tumor growth in both CT26 syngrafts and CFPAC-1 xenografts. In all cases, the highest treatment dose resulted in 100% survival at the study endpoint whereas the control cohorts showed 0% and 60% survival in the CT26 and CFPAC-1 models, respectively. Therefore, PreTarg-it® holds potential as a novel and potent therapy for patients with hard-to-treat solid tumors such as pancreatic cancer, as well as those with late-stage malignancies.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.7,"publicationDate":"2024-06-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141247966","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
89Zr-ImmunoPET for the Specific Detection of EMP2-Positive Tumors. 用于特异性检测 EMP2 阳性肿瘤的 89Zr-ImmunoPET
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-04 DOI: 10.1158/1535-7163.MCT-23-0465
Ann M Chan, Tove Olafsen, Jessica Tsui, Felix B Salazar, Brian Aguirre, Kirstin A Zettlitz, Michael Condro, Anna M Wu, Jonathan Braun, Lynn K Gordon, Negin Ashki, Julian Whitelegge, Shili Xu, Oluwatayo Ikotun, Jason Thanh Lee, Madhuri Wadehra

Epithelial membrane protein-2 (EMP2) is upregulated in a number of tumors and therefore remains a promising target for mAb-based therapy. In the current study, image-guided therapy for an anti-EMP2 mAb was evaluated by PET in both syngeneic and immunodeficient cancer models expressing different levels of EMP2 to enable a better understanding of its tumor uptake and off target accumulation and clearance. The therapeutic efficacy of the anti-EMP2 mAb was initially evaluated in high- and low-expressing tumors, and the mAb reduced tumor load for the high EMP2-expressing 4T1 and HEC-1-A tumors. To create an imaging agent, the anti-EMP2 mAb was conjugated to p-SCN-Bn-deferoxamine (DFO) and radiolabeled with 89Zr. Tumor targeting and tissue biodistribution were evaluated in syngeneic tumor models (4T1, CT26, and Panc02) and human tumor xenograft models (Ramos, HEC-1-A, and U87MG/EMP2). PET imaging revealed radioactive accumulation in EMP2-positive tumors within 24 hours after injection, and the signal was retained for 5 days. High specific uptake was observed in tumors with high EMP2 expression (4T1, CT26, HEC-1-A, and U87MG/EMP2), with less accumulation in tumors with low EMP2 expression (Panc02 and Ramos). Biodistribution at 5 days after injection revealed that the tumor uptake ranged from 2 to approximately 16%ID/cc. The results show that anti-EMP2 mAbs exhibit EMP2-dependent tumor uptake with low off-target accumulation in preclinical cancer models. The development of improved anti-EMP2 Ab fragments may be useful to track EMP2-positive tumors for subsequent therapeutic interventions.

上皮膜蛋白-2(EMP2)在多种肿瘤中上调,因此仍是一种很有前景的单克隆抗体(mAb)治疗靶点。在目前的研究中,通过正电子发射断层扫描(PET)对表达不同水平EMP2的同种异体和免疫缺陷癌症模型中的抗EMP2 mAb进行了图像引导治疗评估,以便更好地了解其肿瘤摄取、靶外蓄积和清除情况。抗 EMP2 mAb 的疗效最初在高表达和低表达肿瘤中进行了评估,结果表明该 mAb 能减少高表达 EMP2 的 4T1 和 HEC-1-A 肿瘤的肿瘤负荷。为了制成成像剂,抗 EMP2 mAb 与对-SCN-Bn-去铁胺(DFO)共轭,并用 89Zr 进行放射性标记。在合成肿瘤模型(4T1、CT26 和 Panc02)和人类肿瘤异种移植模型(Ramos、HEC-1-A 和 U87MG/EMP2)中评估了肿瘤靶向性和组织生物分布。PET 成像显示,EMP2 阳性肿瘤在注射后 24 小时内出现放射性蓄积,信号可持续 5 天。在EMP2高表达的肿瘤(4T1、CT26、HEC-1-A、U87MG/EMP2)中观察到高特异性摄取,而在EMP2低表达的肿瘤(Panc02、Ramos)中积累较少。注射后 5 天的生物分布显示,肿瘤摄取量从 2%ID/cc 到 ~16%ID/cc 不等。结果表明,在临床前癌症模型中,抗EMP2 mAbs表现出依赖于EMP2的肿瘤摄取,且脱靶蓄积较低。开发改进的抗EMP2抗体片段可能有助于追踪EMP2阳性肿瘤,以便进行后续治疗干预。
{"title":"89Zr-ImmunoPET for the Specific Detection of EMP2-Positive Tumors.","authors":"Ann M Chan, Tove Olafsen, Jessica Tsui, Felix B Salazar, Brian Aguirre, Kirstin A Zettlitz, Michael Condro, Anna M Wu, Jonathan Braun, Lynn K Gordon, Negin Ashki, Julian Whitelegge, Shili Xu, Oluwatayo Ikotun, Jason Thanh Lee, Madhuri Wadehra","doi":"10.1158/1535-7163.MCT-23-0465","DOIUrl":"10.1158/1535-7163.MCT-23-0465","url":null,"abstract":"<p><p>Epithelial membrane protein-2 (EMP2) is upregulated in a number of tumors and therefore remains a promising target for mAb-based therapy. In the current study, image-guided therapy for an anti-EMP2 mAb was evaluated by PET in both syngeneic and immunodeficient cancer models expressing different levels of EMP2 to enable a better understanding of its tumor uptake and off target accumulation and clearance. The therapeutic efficacy of the anti-EMP2 mAb was initially evaluated in high- and low-expressing tumors, and the mAb reduced tumor load for the high EMP2-expressing 4T1 and HEC-1-A tumors. To create an imaging agent, the anti-EMP2 mAb was conjugated to p-SCN-Bn-deferoxamine (DFO) and radiolabeled with 89Zr. Tumor targeting and tissue biodistribution were evaluated in syngeneic tumor models (4T1, CT26, and Panc02) and human tumor xenograft models (Ramos, HEC-1-A, and U87MG/EMP2). PET imaging revealed radioactive accumulation in EMP2-positive tumors within 24 hours after injection, and the signal was retained for 5 days. High specific uptake was observed in tumors with high EMP2 expression (4T1, CT26, HEC-1-A, and U87MG/EMP2), with less accumulation in tumors with low EMP2 expression (Panc02 and Ramos). Biodistribution at 5 days after injection revealed that the tumor uptake ranged from 2 to approximately 16%ID/cc. The results show that anti-EMP2 mAbs exhibit EMP2-dependent tumor uptake with low off-target accumulation in preclinical cancer models. The development of improved anti-EMP2 Ab fragments may be useful to track EMP2-positive tumors for subsequent therapeutic interventions.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"890-903"},"PeriodicalIF":5.3,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139990640","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
Patient-Derived Tumor Xenograft Study with CDK4/6 Inhibitor Plus AKT Inhibitor for the Management of Metastatic Castration-Resistant Prostate Cancer. 使用 CDK4/6 抑制剂和 AKT 抑制剂治疗转移性耐受性前列腺癌的患者来源肿瘤异种移植研究。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-04 DOI: 10.1158/1535-7163.MCT-23-0296
Adam M Kase, Justyna Gleba, James L Miller, Erin Miller, Joachim Petit, Michael T Barrett, Yumei Zhou, Ephraim E Parent, Hancheng Cai, Joshua A Knight, Jacob Orme, Jordan Reynolds, William F Durham, Thomas M Metz, Nathalie Meurice, Brandy Edenfield, Aylin Alasonyalilar Demirer, Ahmet Bilgili, Peyton G Hickman, Matthew L Pawlush, Laura Marlow, Daniel P Wickland, Winston Tan, John A Copland

Metastatic castration-resistant prostate cancer (mCRPC) is an aggressive malignancy with poor outcomes. To investigate novel therapeutic strategies, we characterized three new metastatic prostate cancer patient derived-tumor xenograft (PDTX) models and developed 3D spheroids from each to investigate molecular targeted therapy combinations including CDK4/6 inhibitors (CDK4/6i) with AKT inhibitors (ATKi). Metastatic prostate cancer tissue was collected and three PDTX models were established and characterized using whole-exome sequencing. PDTX 3D spheroids were developed from these three PDTXs to show resistance patterns and test novel molecular-targeted therapies. CDK4/6i's were combined with AKTi's to assess synergistic antitumor response to prove our hypothesis that blockade of AKT overcomes drug resistance to CDK4/6i. This combination was evaluated in PDTX three-dimensional (3D) spheroids and in vivo experiments with responses measured by tumor volumes, PSA, and Ga-68 PSMA-11 PET-CT imaging. We demonstrated CDK4/6i's with AKTi's possess synergistic antitumor activity in three mCRPC PDTX models. These models have multiple unique pathogenic and deleterious genomic alterations with resistance to single-agent CDK4/6i's. Despite this, combination therapy with AKTi's was able to overcome resistance mechanisms. The IHC and Western blot analysis confirmed on target effects, whereas tumor volume, serum PSA ELISA, and radionuclide imaging demonstrated response to therapy with statistically significant SUV differences seen with Ga-68 PSMA-11 PET-CT. These preclinical data demonstrating antitumor synergy by overcoming single-agent CDK 4/6i as well as AKTi drug resistance provide the rational for a clinical trial combining a CDK4/6i with an AKTi in patients with mCRPC whose tumor expresses wild-type retinoblastoma 1.

转移性抗性前列腺癌(mCRPC)是一种侵袭性恶性肿瘤,治疗效果不佳。为了研究新的治疗策略,我们对三种新的转移性前列腺癌PDTX模型进行了表征,并从每种模型中培养出三维球体,以研究分子靶向治疗组合,包括CDK4/6抑制剂(CDK4/6i)和AKT抑制剂(ATKi)。收集了转移性前列腺癌组织,建立了三个 PDTX 模型,并使用 WES 进行了表征。从这三种 PDTX 中培养出了 PDTX 3-D 球体,以显示抗药性模式并测试新型分子靶向疗法。我们将 CDK4/6i 与 AKTi 结合使用,以评估协同抗肿瘤反应,从而证明我们的假设,即阻断 AKT 可克服 CDK4/6 抑制剂的耐药性。我们在 PDTX 三维球体和体内实验中评估了这种组合,并通过肿瘤体积、PSA 和 Ga-68 PSMA-11 PET-CT 成像测量了反应。我们证明 CDK4/6i 与 AKTi 在三种 mCRPC PDTX 模型中具有协同抗肿瘤活性。这些模型具有多种独特的致病性和有害基因组改变,对单药 CDK4/6i 具有抗药性。尽管如此,与 AKTi 的联合疗法仍能克服耐药机制。IHC 和 Western 印迹分析证实了靶向效果,而肿瘤体积、血清 PSA 酶联免疫吸附试验和放射性核素成像则显示了对治疗的反应,Ga-68 PSMA-11 PET-CT 显示了具有统计学意义的 SUV 差异。这些临床前数据通过克服单药 CDK 4/6i 和 AKTi 的耐药性而显示出抗肿瘤协同作用,为在肿瘤表达野生型 RB1 的 mCRPC 患者中联合使用 CDK4/6i 和 AKTi 的临床试验提供了依据。
{"title":"Patient-Derived Tumor Xenograft Study with CDK4/6 Inhibitor Plus AKT Inhibitor for the Management of Metastatic Castration-Resistant Prostate Cancer.","authors":"Adam M Kase, Justyna Gleba, James L Miller, Erin Miller, Joachim Petit, Michael T Barrett, Yumei Zhou, Ephraim E Parent, Hancheng Cai, Joshua A Knight, Jacob Orme, Jordan Reynolds, William F Durham, Thomas M Metz, Nathalie Meurice, Brandy Edenfield, Aylin Alasonyalilar Demirer, Ahmet Bilgili, Peyton G Hickman, Matthew L Pawlush, Laura Marlow, Daniel P Wickland, Winston Tan, John A Copland","doi":"10.1158/1535-7163.MCT-23-0296","DOIUrl":"10.1158/1535-7163.MCT-23-0296","url":null,"abstract":"<p><p>Metastatic castration-resistant prostate cancer (mCRPC) is an aggressive malignancy with poor outcomes. To investigate novel therapeutic strategies, we characterized three new metastatic prostate cancer patient derived-tumor xenograft (PDTX) models and developed 3D spheroids from each to investigate molecular targeted therapy combinations including CDK4/6 inhibitors (CDK4/6i) with AKT inhibitors (ATKi). Metastatic prostate cancer tissue was collected and three PDTX models were established and characterized using whole-exome sequencing. PDTX 3D spheroids were developed from these three PDTXs to show resistance patterns and test novel molecular-targeted therapies. CDK4/6i's were combined with AKTi's to assess synergistic antitumor response to prove our hypothesis that blockade of AKT overcomes drug resistance to CDK4/6i. This combination was evaluated in PDTX three-dimensional (3D) spheroids and in vivo experiments with responses measured by tumor volumes, PSA, and Ga-68 PSMA-11 PET-CT imaging. We demonstrated CDK4/6i's with AKTi's possess synergistic antitumor activity in three mCRPC PDTX models. These models have multiple unique pathogenic and deleterious genomic alterations with resistance to single-agent CDK4/6i's. Despite this, combination therapy with AKTi's was able to overcome resistance mechanisms. The IHC and Western blot analysis confirmed on target effects, whereas tumor volume, serum PSA ELISA, and radionuclide imaging demonstrated response to therapy with statistically significant SUV differences seen with Ga-68 PSMA-11 PET-CT. These preclinical data demonstrating antitumor synergy by overcoming single-agent CDK 4/6i as well as AKTi drug resistance provide the rational for a clinical trial combining a CDK4/6i with an AKTi in patients with mCRPC whose tumor expresses wild-type retinoblastoma 1.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"823-835"},"PeriodicalIF":5.3,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140039805","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
Macrophages as Targets in Hepatocellular Carcinoma Therapy. 作为肝癌治疗靶点的巨噬细胞
IF 5.7 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-04 DOI: 10.1158/1535-7163.MCT-23-0660
Yu-Ting Liu, Zheng-Wei Mao, Yuan Ding, Wei-Lin Wang

Hepatocellular carcinoma (HCC) is a malignant tumor with a complex and diverse immunosuppressive microenvironment. Tumor-associated macrophages (TAM) are an essential component of the tumor immune microenvironment. TAMs typically exist in two primary states: anti-tumor M1 macrophages and protumor M2 macrophages. Remarkably, TAMs possess high plasticity, enabling them to switch between different subtypes or alter their biological functions in response to the tumor microenvironment. Based on research into the biological role of TAMs in the occurrence and development of malignant tumors, including HCC, TAMs are emerging as promising targets for novel tumor treatment strategies. In this review, we provide a detailed introduction to the origin and subtypes of TAMs, elucidate their interactions with other cells in the complex tumor microenvironment of HCC, and describe the biological roles, characteristics, and mechanisms of TAMs in the progression of HCC. Furthermore, we furnish an overview of the latest therapeutic strategies targeting TAMs.

肝细胞癌(HCC)是一种恶性肿瘤,具有复杂多样的免疫抑制微环境。肿瘤相关巨噬细胞(TAMs)是肿瘤免疫微环境的重要组成部分。TAMs 通常以两种主要状态存在:抗肿瘤 M1 巨噬细胞和促肿瘤 M2 巨噬细胞。值得注意的是,TAMs 具有高度可塑性,能在不同亚型之间切换,或根据肿瘤微环境改变其生物功能。基于对 TAMs 在包括 HCC 在内的恶性肿瘤的发生和发展中的生物学作用的研究,TAMs 正在成为新型肿瘤治疗策略的有望靶点。在这篇综述中,我们详细介绍了 TAMs 的起源和亚型,阐明了它们在 HCC 复杂的肿瘤微环境中与其他细胞的相互作用,并描述了 TAMs 在 HCC 进展过程中的生物学作用、特征和机制。此外,我们还概述了针对 TAMs 的最新治疗策略。
{"title":"Macrophages as Targets in Hepatocellular Carcinoma Therapy.","authors":"Yu-Ting Liu, Zheng-Wei Mao, Yuan Ding, Wei-Lin Wang","doi":"10.1158/1535-7163.MCT-23-0660","DOIUrl":"10.1158/1535-7163.MCT-23-0660","url":null,"abstract":"<p><p>Hepatocellular carcinoma (HCC) is a malignant tumor with a complex and diverse immunosuppressive microenvironment. Tumor-associated macrophages (TAM) are an essential component of the tumor immune microenvironment. TAMs typically exist in two primary states: anti-tumor M1 macrophages and protumor M2 macrophages. Remarkably, TAMs possess high plasticity, enabling them to switch between different subtypes or alter their biological functions in response to the tumor microenvironment. Based on research into the biological role of TAMs in the occurrence and development of malignant tumors, including HCC, TAMs are emerging as promising targets for novel tumor treatment strategies. In this review, we provide a detailed introduction to the origin and subtypes of TAMs, elucidate their interactions with other cells in the complex tumor microenvironment of HCC, and describe the biological roles, characteristics, and mechanisms of TAMs in the progression of HCC. Furthermore, we furnish an overview of the latest therapeutic strategies targeting TAMs.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"780-790"},"PeriodicalIF":5.7,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139681305","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
Thio-2 Inhibits Key Signaling Pathways Required for the Development and Progression of Castration-resistant Prostate Cancer. Thio-2 可抑制产生和发展抗性前列腺癌所需的关键信号通路。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-04 DOI: 10.1158/1535-7163.MCT-23-0354
Antje Neeb, Ines Figueiredo, Denisa Bogdan, Laura Cato, Jutta Stober, Juan M Jiménez-Vacas, Victor Gourain, Irene I Lee, Rebecca Seeger, Claudia Muhle-Goll, Bora Gurel, Jonathan Welti, Daniel Nava Rodrigues, Jan Rekowski, Xintao Qiu, Yija Jiang, Patrizio Di Micco, Borja Mateos, Stasė Bielskutė, Ruth Riisnaes, Ana Ferreira, Susana Miranda, Mateus Crespo, Lorenzo Buroni, Jian Ning, Suzanne Carreira, Stefan Bräse, Nicole Jung, Simone Gräßle, Amanda Swain, Xavier Salvatella, Stephen R Plymate, Bissan Al-Lazikani, Henry W Long, Wei Yuan, Myles Brown, Andrew C B Cato, Johann S de Bono, Adam Sharp

Therapies that abrogate persistent androgen receptor (AR) signaling in castration-resistant prostate cancer (CRPC) remain an unmet clinical need. The N-terminal domain of the AR that drives transcriptional activity in CRPC remains a challenging therapeutic target. Herein we demonstrate that BCL-2-associated athanogene-1 (BAG-1) mRNA is highly expressed and associates with signaling pathways, including AR signaling, that are implicated in the development and progression of CRPC. In addition, interrogation of geometric and physiochemical properties of the BAG domain of BAG-1 isoforms identifies it to be a tractable but challenging drug target. Furthermore, through BAG-1 isoform mouse knockout studies, we confirm that BAG-1 isoforms regulate hormone physiology and that therapies targeting the BAG domain will be associated with limited "on-target" toxicity. Importantly, the postulated inhibitor of BAG-1 isoforms, Thio-2, suppressed AR signaling and other important pathways implicated in the development and progression of CRPC to reduce the growth of treatment-resistant prostate cancer cell lines and patient-derived models. However, the mechanism by which Thio-2 elicits the observed phenotype needs further elucidation as the genomic abrogation of BAG-1 isoforms was unable to recapitulate the Thio-2-mediated phenotype. Overall, these data support the interrogation of related compounds with improved drug-like properties as a novel therapeutic approach in CRPC, and further highlight the clinical potential of treatments that block persistent AR signaling which are currently undergoing clinical evaluation in CRPC.

在去势抵抗性前列腺癌(CRPC)中消除持续性雄激素受体(AR)信号传导的疗法仍是一项尚未满足的临床需求。在CRPC中,驱动转录活性的AR的N端结构域(NTD)仍然是一个具有挑战性的治疗靶点。在本文中,我们证明了 BAG-1 mRNA 的高表达并与信号通路(包括 AR 信号通路)相关联,而这些通路与 CRPC 的发生和发展有牵连。此外,对 BAG-1 异构体的 BAG 结构域的几何和理化性质进行的研究发现,它是一个具有挑战性的药物靶点。此外,通过 BAG-1 异构体小鼠基因敲除研究,我们证实 BAG-1 异构体可调节激素生理,而且针对 BAG 结构域的疗法将具有有限的 "靶向 "毒性。重要的是,假定的 BAG-1 异构体抑制剂 Thio-2 能抑制 AR 信号传导以及与 CRPC 的发展和进展有关的其他重要通路,从而减少耐药前列腺癌细胞系和患者衍生模型的生长。然而,Thio-2诱导观察到的表型的机制还需要进一步阐明,因为基因组消减BAG-1同工酶无法再现Thio-2介导的表型。总之,这些数据支持研究具有改进的类药物特性的相关化合物,将其作为治疗 CRPC 的一种新方法,并进一步凸显了阻断持续性 AR 信号转导的治疗方法的临床潜力,这些治疗方法目前正在 CRPC 领域进行临床评估。
{"title":"Thio-2 Inhibits Key Signaling Pathways Required for the Development and Progression of Castration-resistant Prostate Cancer.","authors":"Antje Neeb, Ines Figueiredo, Denisa Bogdan, Laura Cato, Jutta Stober, Juan M Jiménez-Vacas, Victor Gourain, Irene I Lee, Rebecca Seeger, Claudia Muhle-Goll, Bora Gurel, Jonathan Welti, Daniel Nava Rodrigues, Jan Rekowski, Xintao Qiu, Yija Jiang, Patrizio Di Micco, Borja Mateos, Stasė Bielskutė, Ruth Riisnaes, Ana Ferreira, Susana Miranda, Mateus Crespo, Lorenzo Buroni, Jian Ning, Suzanne Carreira, Stefan Bräse, Nicole Jung, Simone Gräßle, Amanda Swain, Xavier Salvatella, Stephen R Plymate, Bissan Al-Lazikani, Henry W Long, Wei Yuan, Myles Brown, Andrew C B Cato, Johann S de Bono, Adam Sharp","doi":"10.1158/1535-7163.MCT-23-0354","DOIUrl":"10.1158/1535-7163.MCT-23-0354","url":null,"abstract":"<p><p>Therapies that abrogate persistent androgen receptor (AR) signaling in castration-resistant prostate cancer (CRPC) remain an unmet clinical need. The N-terminal domain of the AR that drives transcriptional activity in CRPC remains a challenging therapeutic target. Herein we demonstrate that BCL-2-associated athanogene-1 (BAG-1) mRNA is highly expressed and associates with signaling pathways, including AR signaling, that are implicated in the development and progression of CRPC. In addition, interrogation of geometric and physiochemical properties of the BAG domain of BAG-1 isoforms identifies it to be a tractable but challenging drug target. Furthermore, through BAG-1 isoform mouse knockout studies, we confirm that BAG-1 isoforms regulate hormone physiology and that therapies targeting the BAG domain will be associated with limited \"on-target\" toxicity. Importantly, the postulated inhibitor of BAG-1 isoforms, Thio-2, suppressed AR signaling and other important pathways implicated in the development and progression of CRPC to reduce the growth of treatment-resistant prostate cancer cell lines and patient-derived models. However, the mechanism by which Thio-2 elicits the observed phenotype needs further elucidation as the genomic abrogation of BAG-1 isoforms was unable to recapitulate the Thio-2-mediated phenotype. Overall, these data support the interrogation of related compounds with improved drug-like properties as a novel therapeutic approach in CRPC, and further highlight the clinical potential of treatments that block persistent AR signaling which are currently undergoing clinical evaluation in CRPC.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"791-808"},"PeriodicalIF":5.3,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11148553/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139983287","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
ATPase Copper Transporting Beta (ATP7B) Is a Novel Target for Improving the Therapeutic Efficacy of Docetaxel by Disulfiram/Copper in Human Prostate Cancer. ATP酶铜转运β(ATP7B)是通过双硫仑/铜改善多西他赛对人类前列腺癌疗效的新靶点。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-04 DOI: 10.1158/1535-7163.MCT-23-0876
Liankun Song, Vyvyan Nguyen, Jun Xie, Shang Jia, Christopher J Chang, Edward Uchio, Xiaolin Zi

Docetaxel has been the standard first-line chemotherapy for lethal metastatic prostate cancer (mPCa) since 2004, but resistance to docetaxel treatment is common. The molecular mechanisms of docetaxel resistance remain largely unknown and could be amenable to interventions that mitigate resistance. We have recently discovered that several docetaxel-resistant mPCa cell lines exhibit lower uptake of cellular copper and uniquely express higher levels of a copper exporter protein ATP7B. Knockdown of ATP7B by silencing RNAs (siRNA) sensitized docetaxel-resistant mPCa cells to the growth-inhibitory and apoptotic effects of docetaxel. Importantly, deletions of ATP7B in human mPCa tissues predict significantly better survival of patients after their first chemotherapy than those with wild-type ATP7B (P = 0.0006). In addition, disulfiram (DSF), an FDA-approved drug for the treatment of alcohol dependence, in combination with copper, significantly enhanced the in vivo antitumor effects of docetaxel in a docetaxel-resistant xenograft tumor model. Our analyses also revealed that DSF and copper engaged with ATP7B to decrease protein levels of COMM domain-containing protein 1 (COMMD1), S-phase kinase-associated protein 2 (Skp2), and clusterin and markedly increase protein expression of cyclin-dependent kinase inhibitor 1 (p21/WAF1). Taken together, our results indicate a copper-dependent nutrient vulnerability through ATP7B exporter in docetaxel-resistant prostate cancer for improving the therapeutic efficacy of docetaxel.

自2004年以来,多西他赛一直是治疗致死性转移性前列腺癌(mPCa)的标准一线化疗药物,但多西他赛治疗耐药的情况却很普遍。多西他赛耐药的分子机制在很大程度上仍不为人所知,但可以通过干预来减轻耐药性。我们最近发现,几种多西他赛耐药的 mPCa 细胞系对细胞铜的吸收率较低,并且独特地表达较高水平的铜输出蛋白 ATP7B。通过沉默 RNA(siRNA)敲除 ATP7B 可使耐多西他赛的 mPCa 细胞对多西他赛的生长抑制和凋亡效应敏感。重要的是,人mPCa组织中ATP7B的缺失预示着患者首次化疗后的生存率明显高于ATP7B野生型患者(P = 0.0006)。此外,美国 FDA 批准用于治疗酒精依赖的药物双硫仑(DSF)与铜联合使用,可在多西他赛耐药异种移植肿瘤模型中显著增强多西他赛的体内抗肿瘤效果。我们的分析还显示,DSF 和铜与 ATP7B 相互作用,降低了 COMM domain-containing protein 1 (COMMD1)、S 期激酶相关蛋白 2 (Skp2) 和 clusterin 的蛋白水平,并明显增加了细胞周期蛋白依赖性激酶抑制剂 1 (p21/WAF1) 的蛋白表达。综上所述,我们的研究结果表明,在多西他赛耐药的PCa中,铜通过ATP7B输出端依赖于营养易损性,从而提高了多西他赛的疗效。
{"title":"ATPase Copper Transporting Beta (ATP7B) Is a Novel Target for Improving the Therapeutic Efficacy of Docetaxel by Disulfiram/Copper in Human Prostate Cancer.","authors":"Liankun Song, Vyvyan Nguyen, Jun Xie, Shang Jia, Christopher J Chang, Edward Uchio, Xiaolin Zi","doi":"10.1158/1535-7163.MCT-23-0876","DOIUrl":"10.1158/1535-7163.MCT-23-0876","url":null,"abstract":"<p><p>Docetaxel has been the standard first-line chemotherapy for lethal metastatic prostate cancer (mPCa) since 2004, but resistance to docetaxel treatment is common. The molecular mechanisms of docetaxel resistance remain largely unknown and could be amenable to interventions that mitigate resistance. We have recently discovered that several docetaxel-resistant mPCa cell lines exhibit lower uptake of cellular copper and uniquely express higher levels of a copper exporter protein ATP7B. Knockdown of ATP7B by silencing RNAs (siRNA) sensitized docetaxel-resistant mPCa cells to the growth-inhibitory and apoptotic effects of docetaxel. Importantly, deletions of ATP7B in human mPCa tissues predict significantly better survival of patients after their first chemotherapy than those with wild-type ATP7B (P = 0.0006). In addition, disulfiram (DSF), an FDA-approved drug for the treatment of alcohol dependence, in combination with copper, significantly enhanced the in vivo antitumor effects of docetaxel in a docetaxel-resistant xenograft tumor model. Our analyses also revealed that DSF and copper engaged with ATP7B to decrease protein levels of COMM domain-containing protein 1 (COMMD1), S-phase kinase-associated protein 2 (Skp2), and clusterin and markedly increase protein expression of cyclin-dependent kinase inhibitor 1 (p21/WAF1). Taken together, our results indicate a copper-dependent nutrient vulnerability through ATP7B exporter in docetaxel-resistant prostate cancer for improving the therapeutic efficacy of docetaxel.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"854-863"},"PeriodicalIF":5.3,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11150099/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139990641","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
Evaluation of the Role of AXL in Fusion-positive Pediatric Rhabdomyosarcoma Identifies the Small-molecule Inhibitor Bemcentinib (BGB324) as Potent Chemosensitizer. 通过评估 AXL 在融合阳性小儿横纹肌肉瘤中的作用,发现小分子抑制剂 bemcentinib (BGB324) 是一种有效的化疗增敏剂。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-04 DOI: 10.1158/1535-7163.MCT-23-0285
Sara G Danielli, Jakob Wurth, Sarah Morice, Samanta Kisele, Didier Surdez, Olivier Delattre, Peter K Bode, Marco Wachtel, Beat W Schäfer

Rhabdomyosarcoma (RMS) is a highly aggressive pediatric cancer with features of skeletal muscle differentiation. More than 80% of the high-risk patients ultimately fail to respond to chemotherapy treatment, leading to limited therapeutic options and dismal prognostic rates. The lack of response and subsequent tumor recurrence is driven in part by stem cell-like cells, the tumor subpopulation that is enriched after treatment, and characterized by expression of the AXL receptor tyrosine kinase (AXL). AXL mediates survival, migration, and therapy resistance in several cancer types; however, its function in RMS remains unclear. In this study, we investigated the role of AXL in RMS tumorigenesis, migration, and chemotherapy response, and whether targeting of AXL with small-molecule inhibitors could potentiate the efficacy of chemotherapy. We show that AXL is expressed in a heterogeneous manner in patient-derived xenografts (PDX), primary cultures and cell line models of RMS, consistent with its stem cell-state selectivity. By generating a CRISPR/Cas9 AXL knock-out and overexpressing models, we show that AXL contributes to the migratory phenotype of RMS, but not to chemotherapy resistance. Instead, pharmacologic blockade with the AXL inhibitors bemcentinib (BGB324), cabozantinib and NPS-1034 rapidly killed RMS cells in an AXL-independent manner and augmented the efficacy of the chemotherapeutics vincristine and cyclophosphamide. In vivo administration of the combination of bemcentinib and vincristine exerted strong antitumoral activity in a rapidly progressing PDX mouse model, significantly reducing tumor burden compared with single-agent treatment. Collectively, our data identify bemcentinib as a promising drug to improve chemotherapy efficacy in patients with RMS.

横纹肌肉瘤(RMS)是一种侵袭性极强的儿科癌症,具有骨骼肌分化的特征。80%以上的高危患者最终对化疗治疗无效,导致治疗方案有限,预后不良。缺乏反应和随后的肿瘤复发部分是由干细胞样细胞驱动的,干细胞样细胞是治疗后富集的肿瘤亚群,其特征是表达AXL受体酪氨酸激酶(AXL)。AXL 在几种癌症类型中介导生存、迁移和耐药性,但它在 RMS 中的功能仍不清楚。在这项研究中,我们探讨了AXL在RMS肿瘤发生、迁移和化疗反应中的作用,以及用小分子抑制剂靶向AXL是否能增强化疗的疗效。我们发现,AXL在RMS患者衍生异种移植物(PDX)、原代培养物和细胞系模型中以不同的方式表达,这与其干细胞状态选择性相一致。通过建立CRISPR/Cas9 AXL基因敲除和过表达模型,我们发现AXL会导致RMS的迁移表型,但不会导致化疗耐药。相反,AXL抑制剂bemcentinib (BGB324)、cabozantinib和NPS-1034的药理阻断以AXL无关的方式迅速杀死了RMS细胞,并增强了化疗药物长春新碱和环磷酰胺的疗效。在快速进展的PDX小鼠模型中,贝仑替尼和长春新碱的联合体内给药发挥了强大的抗肿瘤活性,与单药治疗相比,显著降低了肿瘤的布鲁登氏度。总之,我们的数据表明贝仑替尼是一种有希望改善RMS患者化疗疗效的药物。
{"title":"Evaluation of the Role of AXL in Fusion-positive Pediatric Rhabdomyosarcoma Identifies the Small-molecule Inhibitor Bemcentinib (BGB324) as Potent Chemosensitizer.","authors":"Sara G Danielli, Jakob Wurth, Sarah Morice, Samanta Kisele, Didier Surdez, Olivier Delattre, Peter K Bode, Marco Wachtel, Beat W Schäfer","doi":"10.1158/1535-7163.MCT-23-0285","DOIUrl":"10.1158/1535-7163.MCT-23-0285","url":null,"abstract":"<p><p>Rhabdomyosarcoma (RMS) is a highly aggressive pediatric cancer with features of skeletal muscle differentiation. More than 80% of the high-risk patients ultimately fail to respond to chemotherapy treatment, leading to limited therapeutic options and dismal prognostic rates. The lack of response and subsequent tumor recurrence is driven in part by stem cell-like cells, the tumor subpopulation that is enriched after treatment, and characterized by expression of the AXL receptor tyrosine kinase (AXL). AXL mediates survival, migration, and therapy resistance in several cancer types; however, its function in RMS remains unclear. In this study, we investigated the role of AXL in RMS tumorigenesis, migration, and chemotherapy response, and whether targeting of AXL with small-molecule inhibitors could potentiate the efficacy of chemotherapy. We show that AXL is expressed in a heterogeneous manner in patient-derived xenografts (PDX), primary cultures and cell line models of RMS, consistent with its stem cell-state selectivity. By generating a CRISPR/Cas9 AXL knock-out and overexpressing models, we show that AXL contributes to the migratory phenotype of RMS, but not to chemotherapy resistance. Instead, pharmacologic blockade with the AXL inhibitors bemcentinib (BGB324), cabozantinib and NPS-1034 rapidly killed RMS cells in an AXL-independent manner and augmented the efficacy of the chemotherapeutics vincristine and cyclophosphamide. In vivo administration of the combination of bemcentinib and vincristine exerted strong antitumoral activity in a rapidly progressing PDX mouse model, significantly reducing tumor burden compared with single-agent treatment. Collectively, our data identify bemcentinib as a promising drug to improve chemotherapy efficacy in patients with RMS.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"864-876"},"PeriodicalIF":5.3,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11148551/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140110669","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
Mitochondrial and Cytosolic One-Carbon Metabolism Is a Targetable Metabolic Vulnerability in Cisplatin-Resistant Ovarian Cancer. 线粒体和细胞膜一碳代谢是顺铂耐药卵巢癌的一个可靶向代谢漏洞。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-04 DOI: 10.1158/1535-7163.MCT-23-0550
Adrianne Wallace-Povirk, Carrie O'Connor, Aamod S Dekhne, Xun Bao, Md Junayed Nayeen, Mathew Schneider, Jade M Katinas, Jennifer Wong-Roushar, Seongho Kim, Lisa Polin, Jing Li, Jessica B Back, Charles E Dann, Aleem Gangjee, Zhanjun Hou, Larry H Matherly

One-carbon (C1) metabolism is compartmentalized between the cytosol and mitochondria with the mitochondrial C1 pathway as the major source of glycine and C1 units for cellular biosynthesis. Expression of mitochondrial C1 genes including SLC25A32, serine hydroxymethyl transferase (SHMT) 2, 5,10-methylene tetrahydrofolate dehydrogenase 2, and 5,10-methylene tetrahydrofolate dehydrogenase 1-like was significantly elevated in primary epithelial ovarian cancer (EOC) specimens compared with normal ovaries. 5-Substituted pyrrolo[3,2-d]pyrimidine antifolates (AGF347, AGF359, AGF362) inhibited proliferation of cisplatin-sensitive (A2780, CaOV3, IGROV1) and cisplatin-resistant (A2780-E80, SKOV3) EOC cells. In SKOV3 and A2780-E80 cells, colony formation was inhibited. AGF347 induced apoptosis in SKOV3 cells. In IGROV1 cells, AGF347 was transported by folate receptor (FR) α. AGF347 was also transported into IGROV1 and SKOV3 cells by the proton-coupled folate transporter (SLC46A1) and the reduced folate carrier (SLC19A1). AGF347 accumulated to high levels in the cytosol and mitochondria of SKOV3 cells. By targeted metabolomics with [2,3,3-2H]L-serine, AGF347, AGF359, and AGF362 inhibited SHMT2 in the mitochondria. In the cytosol, SHMT1 and de novo purine biosynthesis (i.e., glycinamide ribonucleotide formyltransferase, 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase) were targeted; AGF359 also inhibited thymidylate synthase. Antifolate treatments of SKOV3 cells depleted cellular glycine, mitochondrial NADH and glutathione, and showed synergistic in vitro inhibition toward SKOV3 and A2780-E80 cells when combined with cisplatin. In vivo studies with subcutaneous SKOV3 EOC xenografts in SCID mice confirmed significant antitumor efficacy of AGF347. Collectively, our studies demonstrate a unique metabolic vulnerability in EOC involving mitochondrial and cytosolic C1 metabolism, which offers a promising new platform for therapy.

一碳(C1)代谢在细胞质和线粒体之间分区,线粒体 C1 途径是细胞生物合成中甘氨酸和 C1 单位的主要来源。与正常卵巢相比,原发性上皮性卵巢癌(EOC)标本中线粒体 C1 基因(包括 SLC25A32、丝氨酸羟甲基转移酶(SHMT)2、5,10-亚甲基四氢叶酸脱氢酶 2 和 5,10-亚甲基四氢叶酸脱氢酶 1-like)的表达明显升高。5-取代吡咯并[3,2-d]嘧啶类抗酚类化合物(AGF347、AGF359、AGF362)可抑制顺铂敏感型(A2780、CaOV3、IGROV1)和耐药型(A2780-E80、SKOV3)EOC细胞的增殖。在 SKOV3 和 A2780-E80 细胞中,集落形成受到抑制。AGF347 可诱导 SKOV3 细胞凋亡。在 IGROV1 细胞中,AGF347 通过叶酸受体(FR)α 转运。AGF347 还通过质子偶联叶酸转运体(SLC46A1)和还原叶酸载体(SLC19A1)转运到 IGROV1 和 SKOV3 细胞中。AGF347 在 SKOV3 细胞的细胞质和线粒体中积累到很高的水平。通过使用[2,3,3-2H]L-丝氨酸进行靶向代谢组学研究,AGF347、AGF359 和 AGF362 抑制了线粒体中的 SHMT2。在细胞质中,SHMT1 和新嘌呤生物合成(即甘氨酰胺核糖核苷酸酰基转移酶、5-氨基咪唑-4-甲酰胺核糖核苷酸酰基转移酶)是靶标;AGF359 还能抑制胸苷酸合成酶。对 SKOV3 细胞进行抗叶酸处理会耗尽细胞中的甘氨酸、线粒体 NADH 和谷胱甘肽,与顺铂联合使用时对 SKOV3 和 A2780-E80 细胞有协同抑制作用。用 SCID 小鼠皮下 SKOV3 EOC 异种移植物进行的体内研究证实,AGF347 具有显著的抗肿瘤疗效。总之,我们的研究证明了 EOC 独特的代谢脆弱性,涉及线粒体和细胞质 C1 代谢,为治疗提供了一个前景广阔的新平台。
{"title":"Mitochondrial and Cytosolic One-Carbon Metabolism Is a Targetable Metabolic Vulnerability in Cisplatin-Resistant Ovarian Cancer.","authors":"Adrianne Wallace-Povirk, Carrie O'Connor, Aamod S Dekhne, Xun Bao, Md Junayed Nayeen, Mathew Schneider, Jade M Katinas, Jennifer Wong-Roushar, Seongho Kim, Lisa Polin, Jing Li, Jessica B Back, Charles E Dann, Aleem Gangjee, Zhanjun Hou, Larry H Matherly","doi":"10.1158/1535-7163.MCT-23-0550","DOIUrl":"10.1158/1535-7163.MCT-23-0550","url":null,"abstract":"<p><p>One-carbon (C1) metabolism is compartmentalized between the cytosol and mitochondria with the mitochondrial C1 pathway as the major source of glycine and C1 units for cellular biosynthesis. Expression of mitochondrial C1 genes including SLC25A32, serine hydroxymethyl transferase (SHMT) 2, 5,10-methylene tetrahydrofolate dehydrogenase 2, and 5,10-methylene tetrahydrofolate dehydrogenase 1-like was significantly elevated in primary epithelial ovarian cancer (EOC) specimens compared with normal ovaries. 5-Substituted pyrrolo[3,2-d]pyrimidine antifolates (AGF347, AGF359, AGF362) inhibited proliferation of cisplatin-sensitive (A2780, CaOV3, IGROV1) and cisplatin-resistant (A2780-E80, SKOV3) EOC cells. In SKOV3 and A2780-E80 cells, colony formation was inhibited. AGF347 induced apoptosis in SKOV3 cells. In IGROV1 cells, AGF347 was transported by folate receptor (FR) α. AGF347 was also transported into IGROV1 and SKOV3 cells by the proton-coupled folate transporter (SLC46A1) and the reduced folate carrier (SLC19A1). AGF347 accumulated to high levels in the cytosol and mitochondria of SKOV3 cells. By targeted metabolomics with [2,3,3-2H]L-serine, AGF347, AGF359, and AGF362 inhibited SHMT2 in the mitochondria. In the cytosol, SHMT1 and de novo purine biosynthesis (i.e., glycinamide ribonucleotide formyltransferase, 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase) were targeted; AGF359 also inhibited thymidylate synthase. Antifolate treatments of SKOV3 cells depleted cellular glycine, mitochondrial NADH and glutathione, and showed synergistic in vitro inhibition toward SKOV3 and A2780-E80 cells when combined with cisplatin. In vivo studies with subcutaneous SKOV3 EOC xenografts in SCID mice confirmed significant antitumor efficacy of AGF347. Collectively, our studies demonstrate a unique metabolic vulnerability in EOC involving mitochondrial and cytosolic C1 metabolism, which offers a promising new platform for therapy.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"809-822"},"PeriodicalIF":5.3,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11150100/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139913011","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
Structural Basis for Multivalent MUC16 Recognition and Robust Anti-Pancreatic Cancer Activity of Humanized Antibody AR9.6. 人源化抗体 AR9.6 多价 MUC16 识别和强大抗胰腺癌活性的结构基础
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-04 DOI: 10.1158/1535-7163.MCT-23-0868
Eric N Aguilar, Satish Sagar, Brandy R Murray, Christabelle Rajesh, Eric K Lei, Sarah A Michaud, David R Goodlett, Thomas C Caffrey, Paul M Grandgenett, Benjamin Swanson, Teresa M Brooks, Adrian R Black, Henk van Faassen, Greg Hussack, Kevin A Henry, Michael A Hollingsworth, Cory L Brooks, Prakash Radhakrishnan

Mucin-16 (MUC16) is a target for antibody-mediated immunotherapy in pancreatic ductal adenocarcinoma (PDAC) among other malignancies. The MUC16-specific monoclonal antibody AR9.6 has shown promise for PDAC immunotherapy and imaging. Here, we report the structural and biological characterization of the humanized AR9.6 antibody (huAR9.6). The structure of huAR9.6 was determined in complex with a MUC16 SEA (Sea urchin sperm, Enterokinase, Agrin) domain. Binding of huAR9.6 to recombinant, shed, and cell-surface MUC16 was characterized, and anti-PDAC activity was evaluated in vitro and in vivo. HuAR9.6 bound a discontinuous, SEA domain epitope with an overall affinity of 88 nmol/L. Binding affinity depended on the specific SEA domain(s) present, and glycosylation modestly enhanced affinity driven by favorable entropy and enthalpy and via distinct transition state thermodynamic pathways. Treatment with huAR9.6 reduced the in vitro growth, migration, invasion, and clonogenicity of MUC16-positive PDAC cells and patient-derived organoids (PDO). HuAR9.6 blocked MUC16-mediated ErbB and AKT activation in PDAC cells, PDOs, and patient-derived xenografts and induced antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. More importantly, huAR9.6 treatment caused substantial PDAC regression in subcutaneous and orthotopic tumor models. The mechanism of action of huAR9.6 may depend on dense avid binding to homologous SEA domains on MUC16. The results of this study validate the translational therapeutic potential of huAR9.6 against MUC16-positive PDACs.

粘蛋白-16(MUC16)是胰腺导管腺癌(PDAC)和其他恶性肿瘤中抗体介导免疫疗法的靶点。MUC16特异性单克隆抗体AR9.6已在PDAC免疫治疗和成像方面显示出前景。在此,我们报告了人源化 AR9.6 抗体(huAR9.6)的结构和生物学特性。我们确定了 huAR9.6 与 MUC16 SEA(海胆精子、肠激酶、Agrin)结构域的复合物结构。对 huAR9.6 与重组、脱落和细胞表面 MUC16 的结合进行了表征,并在体外和体内评估了其抗 PDAC 活性。结合亲和力取决于存在的特定 SEA 结构域,糖基化可通过有利的熵和焓以及不同的过渡态热力学途径将亲和力提高 3-7 倍。用huAR9.6处理可减少MUC16阳性PDAC细胞和患者衍生的器官组织(PDOs)的体外生长、迁移、侵袭和克隆性。HuAR9.6 阻止了 MUC16 介导的 ErbB 和 AKT 在 PDAC 细胞、PDOs 和患者衍生异种移植物中的激活,并诱导了抗体依赖性细胞毒性和补体依赖性细胞毒性。更重要的是,huAR9.6 治疗可使皮下和正位肿瘤模型中的 PDAC 大量消退。huAR9.6的作用机制可能取决于与MUC16上同源SEA结构域的高密度亲和性结合。本研究的结果验证了 huAR9.6 对 MUC16 阳性 PDAC 的转化治疗潜力。
{"title":"Structural Basis for Multivalent MUC16 Recognition and Robust Anti-Pancreatic Cancer Activity of Humanized Antibody AR9.6.","authors":"Eric N Aguilar, Satish Sagar, Brandy R Murray, Christabelle Rajesh, Eric K Lei, Sarah A Michaud, David R Goodlett, Thomas C Caffrey, Paul M Grandgenett, Benjamin Swanson, Teresa M Brooks, Adrian R Black, Henk van Faassen, Greg Hussack, Kevin A Henry, Michael A Hollingsworth, Cory L Brooks, Prakash Radhakrishnan","doi":"10.1158/1535-7163.MCT-23-0868","DOIUrl":"10.1158/1535-7163.MCT-23-0868","url":null,"abstract":"<p><p>Mucin-16 (MUC16) is a target for antibody-mediated immunotherapy in pancreatic ductal adenocarcinoma (PDAC) among other malignancies. The MUC16-specific monoclonal antibody AR9.6 has shown promise for PDAC immunotherapy and imaging. Here, we report the structural and biological characterization of the humanized AR9.6 antibody (huAR9.6). The structure of huAR9.6 was determined in complex with a MUC16 SEA (Sea urchin sperm, Enterokinase, Agrin) domain. Binding of huAR9.6 to recombinant, shed, and cell-surface MUC16 was characterized, and anti-PDAC activity was evaluated in vitro and in vivo. HuAR9.6 bound a discontinuous, SEA domain epitope with an overall affinity of 88 nmol/L. Binding affinity depended on the specific SEA domain(s) present, and glycosylation modestly enhanced affinity driven by favorable entropy and enthalpy and via distinct transition state thermodynamic pathways. Treatment with huAR9.6 reduced the in vitro growth, migration, invasion, and clonogenicity of MUC16-positive PDAC cells and patient-derived organoids (PDO). HuAR9.6 blocked MUC16-mediated ErbB and AKT activation in PDAC cells, PDOs, and patient-derived xenografts and induced antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. More importantly, huAR9.6 treatment caused substantial PDAC regression in subcutaneous and orthotopic tumor models. The mechanism of action of huAR9.6 may depend on dense avid binding to homologous SEA domains on MUC16. The results of this study validate the translational therapeutic potential of huAR9.6 against MUC16-positive PDACs.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"836-853"},"PeriodicalIF":5.3,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139940295","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting Chemoresistance in Advanced Bladder Cancers with a Novel Adjuvant Strategy. 用新型辅助策略瞄准晚期膀胱癌的耐药性
IF 5.7 2区 医学 Q1 ONCOLOGY Pub Date : 2024-05-30 DOI: 10.1158/1535-7163.MCT-23-0806
Juliette R Seremak, Kunj Bihari Gupta, Sunilkanth Bonigala, Elise Liu, Brendan Marshall, Wenbo Zhi, Riham M Bokhtia, Siva S Panda, Vinata B Lokeshwar, Bal L Lokeshwar

Advanced urinary bladder cancer (BC) is characterized by rapid progression and development of therapy resistance. About 30% of the patients are diagnosed with high-grade tumors (Grade >T2a). A typical non-surgical treatment is systemic chemotherapy using Cisplatin (C) and Gemcitabine (G). However, treatment failure and subsequent disease progression are common in treated patients, and adjuvant therapies are not significantly effective. The therapeutic potential of a molecular hybrid of Ursolic Acid (UA), a pentacyclic-triterpene conjugated to N-methyl piperazine (UA4), was tested on both naïve (WT) and Gemcitabine-resistant (GemR) variants of two human invasive BC cell lines, 5637 and T24. UA4 killed 5637 (4µM), T24 (4µM) WT, and GemR cells invitro at equal potency. Pretreatment with UA4 followed by G synergistically killed WT and GemR cells by >50% compared to G followed by UA4. Oral gavage of UA4 (100 mg/kg) inhibited WT and GemR tumor growth in athymic mice. UA4 + G was more effective against GemR tumors than either drug alone. Studies revealed cytotoxic autophagy as a mechanism of UA4 cytotoxicity. UA4 induced moderate apoptosis in T24 but not in 5637 cells. Mitochondrial integrity and function were most affected by UA4 due to high levels of reactive oxygen species (ROS), disruption of mitochondrial membrane, and cell cycle arrest. These effects were enhanced in the UA4+G combination. UA4 was well-tolerated in mice, and oral gavage led to a serum level >1µM with no systemic toxicity. These results show the potential of UA4 as a non-toxic alternative treatment for high-grade BC.

晚期泌尿系统膀胱癌(BC)的特点是病情进展迅速和出现耐药性。约 30% 的患者被确诊为高级别肿瘤(级别 >T2a)。典型的非手术治疗方法是使用顺铂(C)和吉西他滨(G)进行全身化疗。然而,治疗失败和随后的疾病进展在接受治疗的患者中很常见,辅助疗法的效果也不明显。我们对熊果酸(UA)分子杂交体(UA4)的治疗潜力进行了测试,该杂交体是一种与 N-甲基哌嗪共轭的五环三萜类化合物,适用于 5637 和 T24 这两种人类侵袭性 BC 细胞系的天真型(WT)和吉西他滨耐药型(GemR)变体。UA4 对 5637(4µM)、T24(4µM)WT 和 GemR 细胞的杀伤力相同。与先用 G 再用 UA4 相比,先用 UA4 再用 G 的协同作用可杀死 WT 和 GemR 细胞 50%以上。口服 UA4(100 毫克/千克)可抑制 WT 和 GemR 肿瘤在无胸腺小鼠体内的生长。与单独使用两种药物相比,UA4 + G 对 GemR 肿瘤更有效。研究显示细胞毒性自噬是 UA4 细胞毒性的一种机制。UA4 可诱导 T24 细胞中度凋亡,但不能诱导 5637 细胞中度凋亡。线粒体的完整性和功能受 UA4 的影响最大,这是由于高水平的活性氧(ROS)、线粒体膜破坏和细胞周期停滞造成的。这些影响在 UA4+G 组合中得到了加强。小鼠对 UA4 的耐受性良好,口服后血清中 UA4 的浓度大于 1µM,且无全身毒性。这些结果表明,UA4 有可能成为治疗高级别 BC 的一种无毒替代疗法。
{"title":"Targeting Chemoresistance in Advanced Bladder Cancers with a Novel Adjuvant Strategy.","authors":"Juliette R Seremak, Kunj Bihari Gupta, Sunilkanth Bonigala, Elise Liu, Brendan Marshall, Wenbo Zhi, Riham M Bokhtia, Siva S Panda, Vinata B Lokeshwar, Bal L Lokeshwar","doi":"10.1158/1535-7163.MCT-23-0806","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-23-0806","url":null,"abstract":"<p><p>Advanced urinary bladder cancer (BC) is characterized by rapid progression and development of therapy resistance. About 30% of the patients are diagnosed with high-grade tumors (Grade >T2a). A typical non-surgical treatment is systemic chemotherapy using Cisplatin (C) and Gemcitabine (G). However, treatment failure and subsequent disease progression are common in treated patients, and adjuvant therapies are not significantly effective. The therapeutic potential of a molecular hybrid of Ursolic Acid (UA), a pentacyclic-triterpene conjugated to N-methyl piperazine (UA4), was tested on both naïve (WT) and Gemcitabine-resistant (GemR) variants of two human invasive BC cell lines, 5637 and T24. UA4 killed 5637 (4µM), T24 (4µM) WT, and GemR cells invitro at equal potency. Pretreatment with UA4 followed by G synergistically killed WT and GemR cells by >50% compared to G followed by UA4. Oral gavage of UA4 (100 mg/kg) inhibited WT and GemR tumor growth in athymic mice. UA4 + G was more effective against GemR tumors than either drug alone. Studies revealed cytotoxic autophagy as a mechanism of UA4 cytotoxicity. UA4 induced moderate apoptosis in T24 but not in 5637 cells. Mitochondrial integrity and function were most affected by UA4 due to high levels of reactive oxygen species (ROS), disruption of mitochondrial membrane, and cell cycle arrest. These effects were enhanced in the UA4+G combination. UA4 was well-tolerated in mice, and oral gavage led to a serum level >1µM with no systemic toxicity. These results show the potential of UA4 as a non-toxic alternative treatment for high-grade BC.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.7,"publicationDate":"2024-05-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141176096","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Molecular Cancer Therapeutics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号: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