首页 > 最新文献

Molecular Cancer Therapeutics最新文献

英文 中文
Co-blocking TIGIT and PVRIG using a novel bispecific antibody enhances anti-tumor immunity.
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-01-24 DOI: 10.1158/1535-7163.MCT-23-0614
Yuan Lin, Kan Lin, Qiang Fu, Xing Sun, Huan Wang, Lu Su, Yinghui Xu, Cheng Liao

TIGIT and PVRIG are immune checkpoints co-expressed on activated T and NK cells, contributing to tumor immune evasion. Simultaneous blockade of these pathways may enhance therapeutic efficacy, positioning them as promising dual targets for cancer immunotherapy. This study aimed to develop a bispecific antibody (BsAb) to co-target TIGIT and PVRIG. Expression of TIGIT and PVRIG was assessed on tumor-infiltrating lymphocytes (TILs) from patients with various cancers, including non-small cell lung cancer (n=63) and colorectal cancer (n=26). The BsAb was engineered by fusing anti-PVRIG nanobodies to the N terminus of anti-TIGIT antibodies. Functional characterization of the BsAb was performed in vitro and in vivo, including assessments of T and NK cell activation and cytotoxicity. Pharmacokinetics and safety profiles were evaluated in cynomolgus monkeys. Statistical analyses were conducted using the Student's t-test. The results showed that the BsAb effectively blocked TIGIT and PVRIG from binding their respective ligands, CD155 and CD112, leading to significant increases in T cell activation (2.8-fold, p<0.05) and NK cell cytotoxicity (1.8-fold, p<0.05). In vivo, the BsAb demonstrated potent anti-tumor activity, both as a monotherapy and in combination with anti-PD-1 or anti-PD-L1, in humanized PBMC and transgenic mouse models. Pharmacokinetic studies in cynomolgus monkeys revealed a favorable profile, with no dose-limiting toxicities observed after four repeated doses of 200 mg/kg. These findings provide compelling preclinical evidence for the therapeutic potential of targeting the TIGIT-PVRIG axis with a bispecific antibody. This approach shows promise for enhancing anti-tumor immunity and warrants further investigation in clinical trials.

{"title":"Co-blocking TIGIT and PVRIG using a novel bispecific antibody enhances anti-tumor immunity.","authors":"Yuan Lin, Kan Lin, Qiang Fu, Xing Sun, Huan Wang, Lu Su, Yinghui Xu, Cheng Liao","doi":"10.1158/1535-7163.MCT-23-0614","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-23-0614","url":null,"abstract":"<p><p>TIGIT and PVRIG are immune checkpoints co-expressed on activated T and NK cells, contributing to tumor immune evasion. Simultaneous blockade of these pathways may enhance therapeutic efficacy, positioning them as promising dual targets for cancer immunotherapy. This study aimed to develop a bispecific antibody (BsAb) to co-target TIGIT and PVRIG. Expression of TIGIT and PVRIG was assessed on tumor-infiltrating lymphocytes (TILs) from patients with various cancers, including non-small cell lung cancer (n=63) and colorectal cancer (n=26). The BsAb was engineered by fusing anti-PVRIG nanobodies to the N terminus of anti-TIGIT antibodies. Functional characterization of the BsAb was performed in vitro and in vivo, including assessments of T and NK cell activation and cytotoxicity. Pharmacokinetics and safety profiles were evaluated in cynomolgus monkeys. Statistical analyses were conducted using the Student's t-test. The results showed that the BsAb effectively blocked TIGIT and PVRIG from binding their respective ligands, CD155 and CD112, leading to significant increases in T cell activation (2.8-fold, p<0.05) and NK cell cytotoxicity (1.8-fold, p<0.05). In vivo, the BsAb demonstrated potent anti-tumor activity, both as a monotherapy and in combination with anti-PD-1 or anti-PD-L1, in humanized PBMC and transgenic mouse models. Pharmacokinetic studies in cynomolgus monkeys revealed a favorable profile, with no dose-limiting toxicities observed after four repeated doses of 200 mg/kg. These findings provide compelling preclinical evidence for the therapeutic potential of targeting the TIGIT-PVRIG axis with a bispecific antibody. This approach shows promise for enhancing anti-tumor immunity and warrants further investigation in clinical trials.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143029009","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
Unlocking the Potential of CAR-NK Cell Therapy: Overcoming Barriers and Challenges in the Treatment of Myeloid Malignancies. 释放CAR-NK细胞治疗的潜力:克服髓系恶性肿瘤治疗中的障碍和挑战。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-01-21 DOI: 10.1158/1535-7163.MCT-24-0721
Anqi Zhang, Xingcheng Yang, Yicheng Zhang, Xiaoxuan Yu, Wei Mu, Jia Wei

Myeloid malignancies include various types of cancers that arise from abnormal development or proliferation of myeloid cells within the bone marrow. Chimeric antigen receptor (CAR) T cell treatments, which show great potential for B cell and plasma cell cancers, face major challenges when used for myeloid malignancies. CAR natural killer (NK) cell-based immunotherapy encounters several challenges in treating myeloid cancers, including: (1) poor gene transfer efficiency and expansion platforms in vitro, (2) limited proliferation and persistence in vivo, (3) antigenic heterogeneity, and (4) an immunosuppressive tumor microenvironment. Despite these hurdles, "off-the-shelf" CAR-NK treatments showed encouraging results, marked by enhanced proliferation, prolonged persistence, enhanced tumor infiltration, and improved adaptability. This review offers a summary of the biological traits and cellular sources of NK cells along with a discussion of contemporary CAR designs. Furthermore, it addresses the challenges observed in preclinical research and clinical trials of CAR-NK cell therapy for myeloid cancers, suggesting enhancement strategies.

髓系恶性肿瘤包括骨髓内髓系细胞异常发育或增殖引起的各种类型的癌症。嵌合抗原受体(CAR) T细胞治疗在治疗B细胞癌和浆细胞癌方面显示出巨大的潜力,但在治疗髓系恶性肿瘤方面面临重大挑战。CAR -自然杀伤(NK)细胞免疫疗法在治疗骨髓癌方面面临着几个挑战,包括:(1)体外基因转移效率和扩增平台差,(2)体内增殖和持久性有限,(3)抗原异质性,(4)免疫抑制肿瘤微环境。尽管存在这些障碍,“现成的”CAR-NK治疗显示出令人鼓舞的结果,其特点是增强了增殖,延长了持久性,增强了肿瘤浸润,提高了适应性。本文综述了NK细胞的生物学特性和细胞来源,并讨论了当代CAR的设计。此外,它解决了CAR-NK细胞治疗髓系癌的临床前研究和临床试验中观察到的挑战,提出了增强策略。
{"title":"Unlocking the Potential of CAR-NK Cell Therapy: Overcoming Barriers and Challenges in the Treatment of Myeloid Malignancies.","authors":"Anqi Zhang, Xingcheng Yang, Yicheng Zhang, Xiaoxuan Yu, Wei Mu, Jia Wei","doi":"10.1158/1535-7163.MCT-24-0721","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0721","url":null,"abstract":"<p><p>Myeloid malignancies include various types of cancers that arise from abnormal development or proliferation of myeloid cells within the bone marrow. Chimeric antigen receptor (CAR) T cell treatments, which show great potential for B cell and plasma cell cancers, face major challenges when used for myeloid malignancies. CAR natural killer (NK) cell-based immunotherapy encounters several challenges in treating myeloid cancers, including: (1) poor gene transfer efficiency and expansion platforms in vitro, (2) limited proliferation and persistence in vivo, (3) antigenic heterogeneity, and (4) an immunosuppressive tumor microenvironment. Despite these hurdles, \"off-the-shelf\" CAR-NK treatments showed encouraging results, marked by enhanced proliferation, prolonged persistence, enhanced tumor infiltration, and improved adaptability. This review offers a summary of the biological traits and cellular sources of NK cells along with a discussion of contemporary CAR designs. Furthermore, it addresses the challenges observed in preclinical research and clinical trials of CAR-NK cell therapy for myeloid cancers, suggesting enhancement strategies.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143008573","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 Monounsaturated Fatty Acid Metabolism for Radiosensitization of KRAS Mutant 3D Lung Cancer Models. 靶向单不饱和脂肪酸代谢对KRAS突变体三维肺癌模型的放射增敏作用
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-01-21 DOI: 10.1158/1535-7163.MCT-24-0213
Shan Lu, Xiao Pan, Eva Volckova, Anjali Shinde, Schuyler R Fuller, Regina Egan, Jianli Ma, Jong Kung, Christopher J Ott, Aaron N Hata, Cyril H Benes, Jing X Kang, Henning Willers

Mutations in the KRAS oncogene can mediate resistance to radiation. KRAS mutation (mut) driven tumors have been reported to express cancer stem cell (CSC)-like features and may harbor metabolic liabilities through which CSC-associated radioresistance can be overcome. We established a radiation/drug screening approach that relies on the growth of 3D spheres under anchorage-independent and lipid-limiting culture conditions, which promote stemness and lipogenesis. In this format, we screened 32 KRASmut-enriched lung cancer models. As predicted from published data, CB-839, a glutaminase inhibitor, displayed the highest degree of radiosensitization in KRASmut models with LKB1 co-mutations. Radiosensitization by inhibition of stearoyl-CoA desaturase-1, SCD1, displayed a similar genotype preference though the data also implicated KEAP1 co-mutation and SCD1 expression as potential predictors of radiosensitization. In an isogenic model, KRASmut cells were characterized by increased SCD1 expression and a higher ratio of monounsaturated fatty acids (MUFA) to saturated fatty acids. Accordingly, pharmacological inhibition or depletion of SCD1 radiosensitized isogenic KRASmut but not wild-type cells. The radiosensitizing effect was notably small, especially compared to several DNA repair inhibitors. As an alternative strategy to targeting MUFA metabolism, adding polyunsaturated FAs (PUFA) phenocopied some aspects of SCD1 inhibition, suppressed tumor growth in vivo, and opposed the CSC-like phenotype of KRASmut cells. In conclusion, we report a 3D screening approach that recapitulates clinically relevant features of KRASmut tumors and can be leveraged for therapeutic targeting of metabolic vulnerabilities. Our data highlight pronounced inter-tumoral heterogeneity in radiation/drug responses and the complexity of underlying genomic dependencies.

KRAS癌基因的突变可以介导对辐射的抗性。据报道,KRAS突变(mut)驱动的肿瘤表达癌症干细胞(CSC)样特征,并可能具有代谢缺陷,通过代谢缺陷可以克服CSC相关的辐射抗性。我们建立了一种辐射/药物筛选方法,该方法依赖于3D球体在不依赖锚定和限脂培养条件下的生长,这促进了干细胞和脂肪的形成。在这种格式下,我们筛选了32个krasmut富集的肺癌模型。根据已发表的数据预测,谷氨酰胺酶抑制剂CB-839在LKB1共突变的KRASmut模型中显示出最高程度的放射致敏。抑制硬脂酰辅酶a去饱和酶-1 (SCD1)的放射致敏表现出类似的基因型偏好,尽管数据也表明KEAP1共突变和SCD1表达是放射致敏的潜在预测因子。在等基因模型中,KRASmut细胞的特征是SCD1表达增加,单不饱和脂肪酸(MUFA)与饱和脂肪酸的比例更高。因此,SCD1的药理学抑制或耗竭对KRASmut等基因放射致敏,但对野生型细胞没有作用。特别是与几种DNA修复抑制剂相比,放射增敏作用明显较小。作为靶向MUFA代谢的一种替代策略,添加多不饱和脂肪酸(PUFA)可以在一定程度上表型化SCD1抑制,在体内抑制肿瘤生长,并对抗KRASmut细胞的csc样表型。总之,我们报告了一种3D筛选方法,该方法概括了KRASmut肿瘤的临床相关特征,可以用于代谢脆弱性的治疗靶向。我们的数据突出了肿瘤间放射/药物反应的显著异质性以及潜在基因组依赖性的复杂性。
{"title":"Targeting Monounsaturated Fatty Acid Metabolism for Radiosensitization of KRAS Mutant 3D Lung Cancer Models.","authors":"Shan Lu, Xiao Pan, Eva Volckova, Anjali Shinde, Schuyler R Fuller, Regina Egan, Jianli Ma, Jong Kung, Christopher J Ott, Aaron N Hata, Cyril H Benes, Jing X Kang, Henning Willers","doi":"10.1158/1535-7163.MCT-24-0213","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0213","url":null,"abstract":"<p><p>Mutations in the KRAS oncogene can mediate resistance to radiation. KRAS mutation (mut) driven tumors have been reported to express cancer stem cell (CSC)-like features and may harbor metabolic liabilities through which CSC-associated radioresistance can be overcome. We established a radiation/drug screening approach that relies on the growth of 3D spheres under anchorage-independent and lipid-limiting culture conditions, which promote stemness and lipogenesis. In this format, we screened 32 KRASmut-enriched lung cancer models. As predicted from published data, CB-839, a glutaminase inhibitor, displayed the highest degree of radiosensitization in KRASmut models with LKB1 co-mutations. Radiosensitization by inhibition of stearoyl-CoA desaturase-1, SCD1, displayed a similar genotype preference though the data also implicated KEAP1 co-mutation and SCD1 expression as potential predictors of radiosensitization. In an isogenic model, KRASmut cells were characterized by increased SCD1 expression and a higher ratio of monounsaturated fatty acids (MUFA) to saturated fatty acids. Accordingly, pharmacological inhibition or depletion of SCD1 radiosensitized isogenic KRASmut but not wild-type cells. The radiosensitizing effect was notably small, especially compared to several DNA repair inhibitors. As an alternative strategy to targeting MUFA metabolism, adding polyunsaturated FAs (PUFA) phenocopied some aspects of SCD1 inhibition, suppressed tumor growth in vivo, and opposed the CSC-like phenotype of KRASmut cells. In conclusion, we report a 3D screening approach that recapitulates clinically relevant features of KRASmut tumors and can be leveraged for therapeutic targeting of metabolic vulnerabilities. Our data highlight pronounced inter-tumoral heterogeneity in radiation/drug responses and the complexity of underlying genomic dependencies.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143008570","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
PI3K/mTOR dual inhibitor GSK458 and arsenic trioxide exert synergistic anti-tumor effects against ovarian clear cell carcinoma. PI3K/mTOR双抑制剂GSK458与三氧化二砷协同抗卵巢透明细胞癌。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-01-21 DOI: 10.1158/1535-7163.MCT-24-0490
Yan Wang, Yasuto Kinose, Kanako Kasuya, Mai Koizumi, Aasa Shimizu, Koji Nakamura, Aska Toda, Mahiru Kawano, Michiko Kodama, Kae Hashimoto, Kenjiro Sawada, Tadashi Kimura

Ovarian clear cell carcinoma (OCCC), particularly advanced or recurrent settings, is generally resistant to platinum-based chemotherapy, warranting novel therapeutic strategies. Mutations in the phosphoinositide 3-kinase/protein kinase B/mechanistic target of rapamycin kinase (PI3K/AKT/mTOR) pathway are frequently reported in OCCC. Therefore, we hypothesized that the PI3K/mTOR dual inhibitor, GSK458, and arsenic trioxide may exert synergistic anti-tumor effects on OCCC. We investigated the effects of GSK458, arsenic trioxide, and the combination of GSK458 and arsenic trioxide on cell viability, colony formation, and apoptosis in seven OCCC cells. Mechanistically, transcriptomic differences were assessed among the groups. Additionally, their anti-tumor effects were evaluated on the three-dimensional cultures of OCCC patient-derived xenografts as well as in vivo. Low-dose combination of GSK458 and arsenic trioxide exerted synergistic anti-tumor effects in vitro. Viability of the three-dimensional OCCC patient-derived xenograft cultures treated with the combination of GSK458 and arsenic trioxide decreased to 23.8% of that of the control. RNA sequencing revealed that the mechanism was associated with cell cycle and DNA damage repair. The combination of GSK458 and arsenic trioxide synergistically inhibited the PI3K/AKT/mTOR pathway and angiogenesis and increased apoptosis. Compared to any monotherapy, the combination treatment significantly suppressed tumor growth in vivo, thereby enhancing survival. Overall, our findings highlight the potential of the novel combination of GSK458 and arsenic trioxide combination for OCCC treatment.

卵巢透明细胞癌(OCCC),特别是晚期或复发性卵巢透明细胞癌,通常对铂类化疗具有耐药性,因此需要新的治疗策略。磷酸肌苷3-激酶/蛋白激酶B/雷帕霉素激酶机制靶点(PI3K/AKT/mTOR)通路的突变在OCCC中经常被报道。因此,我们假设PI3K/mTOR双抑制剂GSK458和三氧化二砷可能对OCCC具有协同抗肿瘤作用。我们研究了GSK458、三氧化二砷以及GSK458和三氧化二砷联合使用对7个OCCC细胞的细胞活力、集落形成和凋亡的影响。机制上,评估各组之间的转录组差异。此外,在OCCC患者来源的异种移植物的三维培养和体内,评估了它们的抗肿瘤作用。GSK458与三氧化二砷低剂量联合体外抗肿瘤具有协同作用。GSK458和三氧化二砷联合处理的OCCC患者来源的三维异种移植物培养物的存活率下降到对照组的23.8%。RNA测序显示其机制与细胞周期和DNA损伤修复有关。GSK458与三氧化二砷联合使用可协同抑制PI3K/AKT/mTOR通路和血管生成,并增加细胞凋亡。与任何单一疗法相比,联合治疗在体内显著抑制肿瘤生长,从而提高生存期。总的来说,我们的研究结果突出了GSK458和三氧化二砷联合治疗OCCC的潜力。
{"title":"PI3K/mTOR dual inhibitor GSK458 and arsenic trioxide exert synergistic anti-tumor effects against ovarian clear cell carcinoma.","authors":"Yan Wang, Yasuto Kinose, Kanako Kasuya, Mai Koizumi, Aasa Shimizu, Koji Nakamura, Aska Toda, Mahiru Kawano, Michiko Kodama, Kae Hashimoto, Kenjiro Sawada, Tadashi Kimura","doi":"10.1158/1535-7163.MCT-24-0490","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0490","url":null,"abstract":"<p><p>Ovarian clear cell carcinoma (OCCC), particularly advanced or recurrent settings, is generally resistant to platinum-based chemotherapy, warranting novel therapeutic strategies. Mutations in the phosphoinositide 3-kinase/protein kinase B/mechanistic target of rapamycin kinase (PI3K/AKT/mTOR) pathway are frequently reported in OCCC. Therefore, we hypothesized that the PI3K/mTOR dual inhibitor, GSK458, and arsenic trioxide may exert synergistic anti-tumor effects on OCCC. We investigated the effects of GSK458, arsenic trioxide, and the combination of GSK458 and arsenic trioxide on cell viability, colony formation, and apoptosis in seven OCCC cells. Mechanistically, transcriptomic differences were assessed among the groups. Additionally, their anti-tumor effects were evaluated on the three-dimensional cultures of OCCC patient-derived xenografts as well as in vivo. Low-dose combination of GSK458 and arsenic trioxide exerted synergistic anti-tumor effects in vitro. Viability of the three-dimensional OCCC patient-derived xenograft cultures treated with the combination of GSK458 and arsenic trioxide decreased to 23.8% of that of the control. RNA sequencing revealed that the mechanism was associated with cell cycle and DNA damage repair. The combination of GSK458 and arsenic trioxide synergistically inhibited the PI3K/AKT/mTOR pathway and angiogenesis and increased apoptosis. Compared to any monotherapy, the combination treatment significantly suppressed tumor growth in vivo, thereby enhancing survival. Overall, our findings highlight the potential of the novel combination of GSK458 and arsenic trioxide combination for OCCC treatment.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143008567","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
Broad-spectrum efficacy of CEACAM6-targeted antibody-drug conjugate with BET protein degrader in colorectal, lung, and breast cancer mouse models. ceacam6靶向抗体-药物结合BET蛋白降解物在结直肠癌、肺癌和乳腺癌小鼠模型中的广谱疗效
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-01-15 DOI: 10.1158/1535-7163.MCT-24-0444
Hiroyuki Kogai, Shuntaro Tsukamoto, Minaho Koga, Masayuki Miyano, Tsuyoshi Akagi, Atsumi Yamaguchi, Kiyoshi Mori, Kunihito Gotoh, Youya Nakazawa

Despite remarkable advances in cancer treatment, most solid cancers remain difficult to cure. We recently developed an antibody-drug conjugate (ADC, 84-EBET) for pancreatic cancer by using the carcinoembryonic-antigen-related cell-adhesion molecule 6 (CEACAM6) antibody #84.7 and the bromodomain and extra-terminal (BET) protein degrader EBET. Here, we showed the overexpression of CEACAM6 in colorectal, lung, and breast cancers (CRC, LC, BC) and the broad-spectrum efficacy of 84-EBET in mouse models of these cancers. In vitro assays using cancer organoids and cell lines of CRC, LC, and BC revealed that 84-EBET was more potent than ADCs with known approved payloads-DXd, SN38, and monomethyl auristatin E (MMAE)-or standard chemotherapies. In mouse studies, a single injection of 84-EBET induced marked regression of CRC-, LC-, and BC-patient-derived xenograft tumors and cell-line-derived xenograft tumors. Moreover, in mouse syngeneic CRC, LC, and BC models resistant to PD-1 antibody, the combination of 84-EBET and PD-1 antibody induced complete regression of most tumors. Mechanistically, 84-EBET degraded BRD4 protein in both cancer and stromal cells via bystander efficacy. It decreased stromal inflammatory phenotypes and increased activated T-cell numbers in tumors. These results demonstrate that delivering BET protein degraders to tumors and their microenvironments via a CEACAM6-targeted ADC may be effective against a wide range of solid cancers.

尽管癌症治疗取得了显著进展,但大多数实体癌仍然难以治愈。我们最近利用癌胚-抗原相关细胞粘附分子6 (CEACAM6)抗体84.7和溴域和外端(BET)蛋白降解物EBET开发了一种用于胰腺癌的抗体-药物偶联物(ADC, 84-EBET)。在这里,我们展示了CEACAM6在结直肠癌、肺癌和乳腺癌(CRC、LC、BC)中的过表达,以及84-EBET在这些癌症小鼠模型中的广谱疗效。使用CRC、LC和BC的癌症类器官和细胞系进行的体外实验显示,84-EBET比已知批准有效载荷的adc (dxd、SN38和单甲基耳抑素E (MMAE))或标准化疗更有效。在小鼠研究中,单次注射84-EBET诱导CRC、LC和bc患者来源的异种移植肿瘤和细胞系来源的异种移植肿瘤显著消退。此外,在PD-1抗体耐药的小鼠同基因CRC、LC和BC模型中,84-EBET联合PD-1抗体可诱导大多数肿瘤完全消退。在机制上,84-EBET通过旁观者效应降解癌细胞和基质细胞中的BRD4蛋白。它能降低间质炎症表型,增加肿瘤中活化的t细胞数量。这些结果表明,通过ceacam6靶向ADC将BET蛋白降解物输送到肿瘤及其微环境可能对多种实体癌症有效。
{"title":"Broad-spectrum efficacy of CEACAM6-targeted antibody-drug conjugate with BET protein degrader in colorectal, lung, and breast cancer mouse models.","authors":"Hiroyuki Kogai, Shuntaro Tsukamoto, Minaho Koga, Masayuki Miyano, Tsuyoshi Akagi, Atsumi Yamaguchi, Kiyoshi Mori, Kunihito Gotoh, Youya Nakazawa","doi":"10.1158/1535-7163.MCT-24-0444","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0444","url":null,"abstract":"<p><p>Despite remarkable advances in cancer treatment, most solid cancers remain difficult to cure. We recently developed an antibody-drug conjugate (ADC, 84-EBET) for pancreatic cancer by using the carcinoembryonic-antigen-related cell-adhesion molecule 6 (CEACAM6) antibody #84.7 and the bromodomain and extra-terminal (BET) protein degrader EBET. Here, we showed the overexpression of CEACAM6 in colorectal, lung, and breast cancers (CRC, LC, BC) and the broad-spectrum efficacy of 84-EBET in mouse models of these cancers. In vitro assays using cancer organoids and cell lines of CRC, LC, and BC revealed that 84-EBET was more potent than ADCs with known approved payloads-DXd, SN38, and monomethyl auristatin E (MMAE)-or standard chemotherapies. In mouse studies, a single injection of 84-EBET induced marked regression of CRC-, LC-, and BC-patient-derived xenograft tumors and cell-line-derived xenograft tumors. Moreover, in mouse syngeneic CRC, LC, and BC models resistant to PD-1 antibody, the combination of 84-EBET and PD-1 antibody induced complete regression of most tumors. Mechanistically, 84-EBET degraded BRD4 protein in both cancer and stromal cells via bystander efficacy. It decreased stromal inflammatory phenotypes and increased activated T-cell numbers in tumors. These results demonstrate that delivering BET protein degraders to tumors and their microenvironments via a CEACAM6-targeted ADC may be effective against a wide range of solid cancers.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142984169","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
Novel combination therapy targeting oncogenic signaling kinase P21 activated Kinase-1 and chemotherapeutic drugs against triple negative breast cancer. 针对致癌信号激酶P21活化激酶-1和化疗药物联合治疗三阴性乳腺癌的新疗法。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-01-13 DOI: 10.1158/1535-7163.MCT-24-0404
Inemai Ezhil, Abirami Seetharaman, Rahul Kanumuri, Barathidasan R, Ranga Rao G, Ganesh Venkatraman, Suresh K Rayala

Most of the triple negative phenotype or basal-like molecular subtypes of breast cancers are associated with aggressive clinical behaviour and show poor disease prognosis. Current treatment options are constrained, emphasizing the need for novel combinatorial therapies for this particular tumor subtype. Our group has demonstrated that functionally active p21 activated kinase 1 (PAK1) exhibits significantly higher expression levels in clinical triple negative breast cancer (TNBC) samples compared to other subtypes, as well as adjacent normal tissues. Low PAK1 expression in TNBC was significantly linked to better prognosis, with improved overall survival (OS, p=0.00236) and relapse-free survival (RFS, p=0.0314), as shown by GOBO analysis. To confirm the role of PAK1 as a therapeutic target and to discover novel synergistic chemotherapy drug combinations, we conducted a drug combination screen using triple negative breast cancer cell lines and a mouse metastatic tumor cell line. We identified the combined inhibition of PAK1 inhibitor, NVS-PAK1 with doxorubicin/paclitaxel/methotrexate as a synergistic novel therapeutic approach for treating metastatic TNBC to improve overall survival. This study also indicated a reduction in the effective dosage of the chemotherapeutic drug when combined with NVS-PAK1. Our study demonstrates that combining NVS-PAK1 with each of the chemotherapeutic drugs' doxorubicin, paclitaxel, and methotrexate resulted in decreased colony formation, reduced wound healing capability, and diminished migratory and invasive potential in both TNBC cell lines and 4T1 in vitro. These findings were further validated in orthotopic mouse mammary tumors, confirming that simultaneous PAK1 inhibition alongside chemotherapy significantly enhanced anti-tumor efficacy and reduced metastasis.

大多数乳腺癌的三阴性表型或基底样分子亚型与侵袭性临床行为相关,疾病预后较差。目前的治疗方案是有限的,强调需要新的组合治疗这种特殊的肿瘤亚型。我们的研究小组已经证明,功能活跃的p21活化激酶1 (PAK1)在临床三阴性乳腺癌(TNBC)样本中的表达水平明显高于其他亚型以及邻近的正常组织。GOBO分析显示,TNBC中PAK1的低表达与更好的预后显著相关,总生存期(OS, p=0.00236)和无复发生存期(RFS, p=0.0314)均有改善。为了确认PAK1作为治疗靶点的作用,并发现新的协同化疗药物组合,我们使用三阴性乳腺癌细胞系和小鼠转移性肿瘤细胞系进行了药物联合筛选。我们发现联合抑制PAK1抑制剂NVS-PAK1与阿霉素/紫杉醇/甲氨蝶呤作为一种新的协同治疗方法来治疗转移性TNBC,以提高总生存率。该研究还表明,当与NVS-PAK1联合使用时,化疗药物的有效剂量降低。我们的研究表明,NVS-PAK1与每种化疗药物的阿霉素、紫杉醇和甲氨蝶呤联合使用,可减少TNBC细胞系和4T1的集落形成,降低伤口愈合能力,降低迁移和侵袭潜力。这些发现在原位小鼠乳腺肿瘤中得到进一步验证,证实在化疗的同时抑制PAK1可显著增强抗肿瘤疗效并减少转移。
{"title":"Novel combination therapy targeting oncogenic signaling kinase P21 activated Kinase-1 and chemotherapeutic drugs against triple negative breast cancer.","authors":"Inemai Ezhil, Abirami Seetharaman, Rahul Kanumuri, Barathidasan R, Ranga Rao G, Ganesh Venkatraman, Suresh K Rayala","doi":"10.1158/1535-7163.MCT-24-0404","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0404","url":null,"abstract":"<p><p>Most of the triple negative phenotype or basal-like molecular subtypes of breast cancers are associated with aggressive clinical behaviour and show poor disease prognosis. Current treatment options are constrained, emphasizing the need for novel combinatorial therapies for this particular tumor subtype. Our group has demonstrated that functionally active p21 activated kinase 1 (PAK1) exhibits significantly higher expression levels in clinical triple negative breast cancer (TNBC) samples compared to other subtypes, as well as adjacent normal tissues. Low PAK1 expression in TNBC was significantly linked to better prognosis, with improved overall survival (OS, p=0.00236) and relapse-free survival (RFS, p=0.0314), as shown by GOBO analysis. To confirm the role of PAK1 as a therapeutic target and to discover novel synergistic chemotherapy drug combinations, we conducted a drug combination screen using triple negative breast cancer cell lines and a mouse metastatic tumor cell line. We identified the combined inhibition of PAK1 inhibitor, NVS-PAK1 with doxorubicin/paclitaxel/methotrexate as a synergistic novel therapeutic approach for treating metastatic TNBC to improve overall survival. This study also indicated a reduction in the effective dosage of the chemotherapeutic drug when combined with NVS-PAK1. Our study demonstrates that combining NVS-PAK1 with each of the chemotherapeutic drugs' doxorubicin, paclitaxel, and methotrexate resulted in decreased colony formation, reduced wound healing capability, and diminished migratory and invasive potential in both TNBC cell lines and 4T1 in vitro. These findings were further validated in orthotopic mouse mammary tumors, confirming that simultaneous PAK1 inhibition alongside chemotherapy significantly enhanced anti-tumor efficacy and reduced metastasis.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142971555","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
Inhibition of DKK-1 by WAY262611 Inhibits Osteosarcoma Metastasis. WAY262611抑制DKK-1抑制骨肉瘤转移
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-01-09 DOI: 10.1158/1535-7163.MCT-24-0744
Adit Tal, Shimara Gunawardana-Zeigler, Da Peng, Yuqi Tan, Natalia Munoz Perez, Rachel Offenbacher, Laurel Kastner, Paul Ciero, Matthew E Randolph, Yun Gong, Hong-Wen Deng, Patrick Cahan, David M Loeb

Osteosarcoma (OS) is the most common primary malignant bone tumor in childhood. Patients who present with metastatic disease at diagnosis or relapse have a very poor prognosis, and this has not changed over the past four decades. The Wnt signaling pathway plays a role in regulating osteogenesis and is implicated in OS pathogenesis. DKK-1 inhibits the canonical Wnt signaling pathway, causing inhibition of osteoblast differentiation and disordered bone repair. Our lab previously demonstrated that a monoclonal antibody against DKK-1 prevented metastatic disease in a mouse model. This study expands upon those findings by demonstrating similar results with a small molecule inhibitor of DKK-1, WAY262611, both in vitro and in vivo. WAY262611 was evaluated in vitro on osteosarcoma cell lines, including proliferation, caspase activation, cell cycle analysis, and signaling pathway activation. We utilized our orthotopic implantation-amputation model of osteosarcoma metastasis in vivo to determine the impact of WAY262611 on primary tumor progression and metastatic outgrowth of disseminated tumor cells. Differentiation status was determined using single cell RNA sequencing. We show here that WAY262611 activates canonical Wnt signaling, enhances nuclear localization and transcriptional activity of beta-catenin, and slows proliferation of OS cell lines. We also show that WAY262611 induces osteoblastic differentiation of an OS patient-derived xenograft in vivo, as well as inhibiting metastasis. This work credentials DKK-1 as a therapeutic target in OS, allowing for manipulation of the Wnt signaling pathway and providing preclinical justification for the development of new biologics for prevention of osteosarcoma metastasis.

骨肉瘤(Osteosarcoma, OS)是儿童最常见的原发性骨恶性肿瘤。在诊断或复发时出现转移性疾病的患者预后非常差,这在过去四十年中没有改变。Wnt信号通路在骨形成调控中发挥作用,并参与骨肉瘤的发病机制。DKK-1抑制典型的Wnt信号通路,导致成骨细胞分化抑制和骨修复紊乱。我们的实验室先前在小鼠模型中证明了针对DKK-1的单克隆抗体可以预防转移性疾病。本研究通过在体外和体内证明DKK-1小分子抑制剂WAY262611的类似结果,扩展了这些发现。WAY262611在骨肉瘤细胞系上进行体外评估,包括增殖、caspase激活、细胞周期分析和信号通路激活。我们利用原位植入-截肢骨肉瘤体内转移模型来确定WAY262611对原发性肿瘤进展和播散性肿瘤细胞转移生长的影响。分化状态通过单细胞RNA测序测定。我们发现WAY262611激活典型Wnt信号,增强β -连环蛋白的核定位和转录活性,并减缓OS细胞系的增殖。我们还表明,WAY262611在体内诱导骨肉瘤患者来源的异种移植物的成骨细胞分化,并抑制转移。这项工作证明DKK-1是骨肉瘤的治疗靶点,允许操纵Wnt信号通路,并为开发预防骨肉瘤转移的新生物制剂提供临床前依据。
{"title":"Inhibition of DKK-1 by WAY262611 Inhibits Osteosarcoma Metastasis.","authors":"Adit Tal, Shimara Gunawardana-Zeigler, Da Peng, Yuqi Tan, Natalia Munoz Perez, Rachel Offenbacher, Laurel Kastner, Paul Ciero, Matthew E Randolph, Yun Gong, Hong-Wen Deng, Patrick Cahan, David M Loeb","doi":"10.1158/1535-7163.MCT-24-0744","DOIUrl":"10.1158/1535-7163.MCT-24-0744","url":null,"abstract":"<p><p>Osteosarcoma (OS) is the most common primary malignant bone tumor in childhood. Patients who present with metastatic disease at diagnosis or relapse have a very poor prognosis, and this has not changed over the past four decades. The Wnt signaling pathway plays a role in regulating osteogenesis and is implicated in OS pathogenesis. DKK-1 inhibits the canonical Wnt signaling pathway, causing inhibition of osteoblast differentiation and disordered bone repair. Our lab previously demonstrated that a monoclonal antibody against DKK-1 prevented metastatic disease in a mouse model. This study expands upon those findings by demonstrating similar results with a small molecule inhibitor of DKK-1, WAY262611, both in vitro and in vivo. WAY262611 was evaluated in vitro on osteosarcoma cell lines, including proliferation, caspase activation, cell cycle analysis, and signaling pathway activation. We utilized our orthotopic implantation-amputation model of osteosarcoma metastasis in vivo to determine the impact of WAY262611 on primary tumor progression and metastatic outgrowth of disseminated tumor cells. Differentiation status was determined using single cell RNA sequencing. We show here that WAY262611 activates canonical Wnt signaling, enhances nuclear localization and transcriptional activity of beta-catenin, and slows proliferation of OS cell lines. We also show that WAY262611 induces osteoblastic differentiation of an OS patient-derived xenograft in vivo, as well as inhibiting metastasis. This work credentials DKK-1 as a therapeutic target in OS, allowing for manipulation of the Wnt signaling pathway and providing preclinical justification for the development of new biologics for prevention of osteosarcoma metastasis.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142951624","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 : 2025-01-02 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 R Palmeri, Karl Dane Wittrup

Although 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 antitumor 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 and 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 prooxidant 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 R Palmeri, Karl Dane Wittrup","doi":"10.1158/1535-7163.MCT-24-0163","DOIUrl":"10.1158/1535-7163.MCT-24-0163","url":null,"abstract":"<p><p>Although 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 antitumor 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 and 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 prooxidant 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":" ","pages":"118-130"},"PeriodicalIF":5.3,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11695183/pdf/","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":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting Tumor Antigen 5T4 Using CAR T Cells for the Treatment of Acute Myeloid Leukemia. 利用 CAR-T 细胞靶向肿瘤抗原 5T4 治疗急性髓性白血病。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2025-01-02 DOI: 10.1158/1535-7163.MCT-24-0052
Richard Harrop, Daniel G Blount, Naeem Khan, Mayowa Soyombo, Laura Moyce, Mark T Drayson, Jenny Down, Michelle A Lawson, Deirdre O'Connor, Rachael Nimmo, Yatish Lad, Bernard Souberbielle, Kyriacos Mitrophanous, Anna Ettorre

Chimeric antigen receptor (CAR) T cells represent a novel targeted approach to overcome deficits in the ability of the host immune system to detect and subsequently eradicate tumors. The identification of antigens expressed specifically on the surface of tumor cells is a critical first step for a targeted therapy that selectively targets cancer cells without affecting normal tissues. 5T4 is a tumor-associated antigen expressed on the cell surface of most solid tumors. However, very little is known about its expression in hematologic malignancies. In this study, we assess the expression of 5T4 in different types of leukemias, specifically acute myeloid leukemia (AML), and normal hematopoietic stem cells (HSC). We also provide an in vitro assessment of safety and efficacy of 5T4-targeting CAR T cells against HSCs and AML tumor cell lines. 5T4 expression was seen in about 50% of AML cases; AML with mutated nucleophosmin 1, AML-myelodysplasia-related, and AML not otherwise specified showed the highest percentage of 5T4+ cases. 5T4 CAR T cells efficiently and specifically killed AML tumor cell lines, including leukemic stem cells. Coculture of 5T4 CAR T cells with HSCs from healthy donors showed no impact on subsequent colony formation, thus confirming the safety profile of 5T4. A proof-of-concept study using a murine model for AML demonstrated that CAR T cells recognize 5T4 expressed on cells and can kill tumor cells both in vitro and in vivo. These results highlight 5T4 as a promising target for immune intervention in AML and that CAR T cells can be considered a powerful personalized therapeutic approach to treat AML.

嵌合抗原受体(CAR)T细胞是克服宿主免疫系统检测和根除肿瘤能力缺陷的一种新型靶向方法。5T4 是一种表达在大多数实体瘤细胞表面的肿瘤相关抗原。然而,人们对它在血液恶性肿瘤中的表达却知之甚少。在此,我们评估了 5T4 在不同类型白血病(尤其是急性髓性白血病(AML))和正常造血干细胞(HSCs)中的表达。我们还对针对造血干细胞和急性髓性白血病肿瘤细胞系的 5T4 靶向 CAR-T 细胞的安全性和有效性进行了体外评估。约50%的急性髓细胞性白血病病例都有5T4表达,特别是NPM1、AML-MR和NOS突变的急性髓细胞性白血病。5T4 CAR-T 细胞能高效、特异地杀死急性髓细胞性白血病肿瘤细胞系,包括白血病干细胞。将 5T4 CAR-T 细胞与健康供体的造血干细胞共培养,结果显示对随后的集落形成没有影响,从而证实了 5T4 的安全性。一个急性髓细胞性白血病小鼠模型显示,CAR-T 细胞能识别并杀死体内表达 5T4 的肿瘤细胞。这些结果突出表明,5T4是治疗急性髓细胞性白血病的一个有希望的免疫干预靶点,CAR-T细胞可被视为治疗急性髓细胞性白血病的一种强有力的个性化治疗方法。
{"title":"Targeting Tumor Antigen 5T4 Using CAR T Cells for the Treatment of Acute Myeloid Leukemia.","authors":"Richard Harrop, Daniel G Blount, Naeem Khan, Mayowa Soyombo, Laura Moyce, Mark T Drayson, Jenny Down, Michelle A Lawson, Deirdre O'Connor, Rachael Nimmo, Yatish Lad, Bernard Souberbielle, Kyriacos Mitrophanous, Anna Ettorre","doi":"10.1158/1535-7163.MCT-24-0052","DOIUrl":"10.1158/1535-7163.MCT-24-0052","url":null,"abstract":"<p><p>Chimeric antigen receptor (CAR) T cells represent a novel targeted approach to overcome deficits in the ability of the host immune system to detect and subsequently eradicate tumors. The identification of antigens expressed specifically on the surface of tumor cells is a critical first step for a targeted therapy that selectively targets cancer cells without affecting normal tissues. 5T4 is a tumor-associated antigen expressed on the cell surface of most solid tumors. However, very little is known about its expression in hematologic malignancies. In this study, we assess the expression of 5T4 in different types of leukemias, specifically acute myeloid leukemia (AML), and normal hematopoietic stem cells (HSC). We also provide an in vitro assessment of safety and efficacy of 5T4-targeting CAR T cells against HSCs and AML tumor cell lines. 5T4 expression was seen in about 50% of AML cases; AML with mutated nucleophosmin 1, AML-myelodysplasia-related, and AML not otherwise specified showed the highest percentage of 5T4+ cases. 5T4 CAR T cells efficiently and specifically killed AML tumor cell lines, including leukemic stem cells. Coculture of 5T4 CAR T cells with HSCs from healthy donors showed no impact on subsequent colony formation, thus confirming the safety profile of 5T4. A proof-of-concept study using a murine model for AML demonstrated that CAR T cells recognize 5T4 expressed on cells and can kill tumor cells both in vitro and in vivo. These results highlight 5T4 as a promising target for immune intervention in AML and that CAR T cells can be considered a powerful personalized therapeutic approach to treat AML.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"93-104"},"PeriodicalIF":5.3,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142470226","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 Signaling 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 : 2025-01-02 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 C Y Wang, Luc G Berthiaume

Acute myeloid leukemia (AML) is a hematologic 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 (NMT), NMT1 and NMT2, with varying expressions in AML cell lines and patient samples. We identified NMT2 expression as a marker for survival of patients with AML, 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, induction of endoplasmic reticulum 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-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 leukemia stem cell 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 Signaling 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 C Y Wang, Luc G Berthiaume","doi":"10.1158/1535-7163.MCT-24-0307","DOIUrl":"10.1158/1535-7163.MCT-24-0307","url":null,"abstract":"<p><p>Acute myeloid leukemia (AML) is a hematologic 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 (NMT), NMT1 and NMT2, with varying expressions in AML cell lines and patient samples. We identified NMT2 expression as a marker for survival of patients with AML, 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, induction of endoplasmic reticulum 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-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 leukemia stem cell 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":" ","pages":"69-80"},"PeriodicalIF":5.3,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11694064/pdf/","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":"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