首页 > 最新文献

Molecular Cancer Therapeutics最新文献

英文 中文
Pretargeted Anti-CD20 Radioimmunotherapy with scFv Fusion Protein Safely Combines with BEAM and ASCT in Patients with High-risk B-cell Lymphomas. 预靶向抗cd20放射免疫治疗与scFv融合蛋白安全联合BEAM和ASCT治疗高危b细胞淋巴瘤
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-12-02 DOI: 10.1158/1535-7163.MCT-24-0550
Johnnie J Orozco, Manuela C Matesan, Sally J Lundberg, Robyn L Haaf, Robert S Miyaoka, Darrell R Fisher, Ted A Gooley, Damian J Green, Brenda M Sandmaier, Paul S Martin, Ajay K Gopal

Despite new therapies, many patients with non-Hodgkin lymphoma (NHL) relapse and need more effective salvage therapies. This study (NCT02483000) evaluated the safety of B9E9-FP, a tetrameric single-chain anti-CD20-streptavidin fusion protein used in pretargeted radioimmunotherapy, when combined with BEAM and autologous stem cell transplantation (ASCT) for patients with NHL. Patients with high-risk NHL received B9E9-FP on day -17, clearing agent on day -15, and DOTA-biotin (DOTA-Bt) equally divided and labeled with dose-escalated yttrium-90 (90Y) or with indium-111 (for imaging) on day -14. BEAM chemotherapy started at day -7 before stem cell infusion. Three patients with NHL (mantle cell lymphoma, transformed diffuse large B-cell lymphoma, and de novo diffuse large B-cell lymphoma), ages 52 to 62 years, were treated with 30, 50, or 70 mCi (1,110, 1,850, or 2,590 MBq) 90Y/m2 before ASCT without any dose-limiting toxicity. One case of diarrhea (grade 2) and one case of rash (grade 1) were possibly associated with B9E9-FP or DOTA-Bt, respectively. Pharmacokinetic studies showed peak blood biological percent injected dose per gram blood (% ID/g) of 90Y-DOTA-Bt at 15 minutes after infusion (14.8%-49.4% ID), with only 0.82% to 2.59% ID after 72 hours. Uptake was preferential at bone marrow (1.73-5.96 cGy/mCi injected) and spleen (2.4-4.17 cGy/mCi injected) compared with lungs (0.19-0.48 cGy/mCi). Unbound 90Y-DOTA-Bt was excreted renally without any renal dysfunction noted up to 2 years later. Two of the three enrolled patients are alive and in remission 3.5 to 4.9 years after transplant. Pharmacokinetic, dosimetry, and outcome data support that B9E9-FP pretargeted radioimmunotherapy combined with 90Y-augmented ASCT DOTA-Bt is feasible.

尽管有新的治疗方法,许多NHL患者复发,需要更有效的挽救治疗。这项研究(NCT02483000)评估了用于预靶向放射免疫治疗(PRIT)的四聚体单链抗cd20链亲和素融合蛋白B9E9-FP与BEAM和自体干细胞移植(ASCT)联合治疗NHL患者的安全性。高危NHL患者在第17天接受B9E9-FP治疗,第15天接受清除剂治疗,第14天接受dota -生物素(DOTA-Bt)等量分配并标记为剂量递增的钇-90 (90Y)或铟-111 (111In用于成像)。BEAM化疗开始于干细胞输注前第7天。3例NHL患者(MCL、转化型DLBCL和新生DLBCL),年龄52-62岁,在ASCT前接受30、50或70 mCi(1110、1850或2590 MBq) 90Y/m2治疗,无任何剂量限制性毒性。1例腹泻(2级)和1例皮疹(1级)可能分别与B9E9-FP或DOTA-Bt有关。药代动力学(PK)研究显示,90Y-DOTA-Bt在输注后15 min血中生物注射剂量百分比(% ID/g)达到峰值(14.8 ~ 49.4% ID), 72 h后仅为0.82 ~ 2.59% ID。与肺(0.19 - 0.48 cGy/mCi)相比,骨髓(注射1.73 - 5.96 cGy/mCi)和脾脏(注射2.4 - 4.17 cGy/mCi)的摄取优先。未结合的90Y-DOTA-Bt在2年后无任何肾功能障碍。3例入组患者中有2例存活,移植后3.5 - 4.9年处于缓解期。PK、剂量学和结果数据支持B9E9-FP PRIT和90y增强ASCT DOTA-Bt是可行的。
{"title":"Pretargeted Anti-CD20 Radioimmunotherapy with scFv Fusion Protein Safely Combines with BEAM and ASCT in Patients with High-risk B-cell Lymphomas.","authors":"Johnnie J Orozco, Manuela C Matesan, Sally J Lundberg, Robyn L Haaf, Robert S Miyaoka, Darrell R Fisher, Ted A Gooley, Damian J Green, Brenda M Sandmaier, Paul S Martin, Ajay K Gopal","doi":"10.1158/1535-7163.MCT-24-0550","DOIUrl":"10.1158/1535-7163.MCT-24-0550","url":null,"abstract":"<p><p>Despite new therapies, many patients with non-Hodgkin lymphoma (NHL) relapse and need more effective salvage therapies. This study (NCT02483000) evaluated the safety of B9E9-FP, a tetrameric single-chain anti-CD20-streptavidin fusion protein used in pretargeted radioimmunotherapy, when combined with BEAM and autologous stem cell transplantation (ASCT) for patients with NHL. Patients with high-risk NHL received B9E9-FP on day -17, clearing agent on day -15, and DOTA-biotin (DOTA-Bt) equally divided and labeled with dose-escalated yttrium-90 (90Y) or with indium-111 (for imaging) on day -14. BEAM chemotherapy started at day -7 before stem cell infusion. Three patients with NHL (mantle cell lymphoma, transformed diffuse large B-cell lymphoma, and de novo diffuse large B-cell lymphoma), ages 52 to 62 years, were treated with 30, 50, or 70 mCi (1,110, 1,850, or 2,590 MBq) 90Y/m2 before ASCT without any dose-limiting toxicity. One case of diarrhea (grade 2) and one case of rash (grade 1) were possibly associated with B9E9-FP or DOTA-Bt, respectively. Pharmacokinetic studies showed peak blood biological percent injected dose per gram blood (% ID/g) of 90Y-DOTA-Bt at 15 minutes after infusion (14.8%-49.4% ID), with only 0.82% to 2.59% ID after 72 hours. Uptake was preferential at bone marrow (1.73-5.96 cGy/mCi injected) and spleen (2.4-4.17 cGy/mCi injected) compared with lungs (0.19-0.48 cGy/mCi). Unbound 90Y-DOTA-Bt was excreted renally without any renal dysfunction noted up to 2 years later. Two of the three enrolled patients are alive and in remission 3.5 to 4.9 years after transplant. Pharmacokinetic, dosimetry, and outcome data support that B9E9-FP pretargeted radioimmunotherapy combined with 90Y-augmented ASCT DOTA-Bt is feasible.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"1928-1937"},"PeriodicalIF":5.5,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12396841/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144553996","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The histone modifier KANSL2 is an actionable biomarker in multiple myeloma. 组蛋白修饰因子KANSL2在多发性骨髓瘤中是一种可行的生物标志物。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-26 DOI: 10.1158/1535-7163.MCT-25-0379
Kaiting Jiang, Marieluise Kirchner, Frederik Herzberg, Yan Zhao, Amelie Gasper, Francis Baumgartner, Paul Jung, Jan Braune, Veronika Schulze, Konstandina Isaakidis, Philipp Mertins, Jan Krönke, Matthias Wirth, Ulrich Keller, Stefan Habringer

Epigenetic aberrations are key drivers of multiple myeloma (MM), yet targeted therapies exploiting epigenetic alterations have not been established. By integrating clinical and molecular MM patient data sets with an unbiased genetic in vivo screen, we identified KAT8 regulatory NSL complex subunit 2 (KANSL2) as a histone posttranslational modification (PTM)-associated candidate oncogene. High expression of KANSL2 was associated with adverse prognosis in MM patients. Genetic gain and loss of function models identified a protective role of KANSL2 towards genotoxic stress. By transcriptomics, proteomics and quantitative acetylome profiling, we identified a KANSL2-dependent specific molecular program targetable by acetylation-related modifiers. High KANSL2 levels increased sensitivity to the histone deacetylase (HDAC) inhibitor panobinostat and bromodomain and extra-terminal motif (BET) inhibitor OTX-015 and their combination. Ex vivo drug response profiling in relapsed/refractory MM patient samples confirmed that high KANSL2 expression is associated with selective MM cell killing by HDAC and BET inhibitors. Collectively, these findings position KANSL2 as a mediator of chemotherapy resistance and actionable biomarker for response to drugs targeting its epigenetic program.

表观遗传畸变是多发性骨髓瘤(MM)的关键驱动因素,但利用表观遗传改变的靶向治疗尚未建立。通过整合临床和分子MM患者数据集以及无偏倚的体内遗传筛选,我们确定KAT8调节NSL复合物亚基2 (KANSL2)是组蛋白翻译后修饰(PTM)相关的候选癌基因。高表达的KANSL2与MM患者的不良预后相关。遗传增益和功能丧失模型确定了KANSL2对遗传毒性胁迫的保护作用。通过转录组学、蛋白质组学和定量乙酰化分析,我们确定了一个依赖于kansl2的特定分子程序,可被乙酰化相关修饰子靶向。高KANSL2水平增加了对组蛋白去乙酰化酶(HDAC)抑制剂panobinostat和bromodomain和extra-terminal motif (BET)抑制剂OTX-015及其组合的敏感性。复发/难治性MM患者样本的体外药物反应分析证实,高KANSL2表达与HDAC和BET抑制剂选择性杀死MM细胞有关。总的来说,这些发现表明KANSL2是化疗耐药的中介,也是针对其表观遗传程序的药物反应的可操作的生物标志物。
{"title":"The histone modifier KANSL2 is an actionable biomarker in multiple myeloma.","authors":"Kaiting Jiang, Marieluise Kirchner, Frederik Herzberg, Yan Zhao, Amelie Gasper, Francis Baumgartner, Paul Jung, Jan Braune, Veronika Schulze, Konstandina Isaakidis, Philipp Mertins, Jan Krönke, Matthias Wirth, Ulrich Keller, Stefan Habringer","doi":"10.1158/1535-7163.MCT-25-0379","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0379","url":null,"abstract":"<p><p>Epigenetic aberrations are key drivers of multiple myeloma (MM), yet targeted therapies exploiting epigenetic alterations have not been established. By integrating clinical and molecular MM patient data sets with an unbiased genetic in vivo screen, we identified KAT8 regulatory NSL complex subunit 2 (KANSL2) as a histone posttranslational modification (PTM)-associated candidate oncogene. High expression of KANSL2 was associated with adverse prognosis in MM patients. Genetic gain and loss of function models identified a protective role of KANSL2 towards genotoxic stress. By transcriptomics, proteomics and quantitative acetylome profiling, we identified a KANSL2-dependent specific molecular program targetable by acetylation-related modifiers. High KANSL2 levels increased sensitivity to the histone deacetylase (HDAC) inhibitor panobinostat and bromodomain and extra-terminal motif (BET) inhibitor OTX-015 and their combination. Ex vivo drug response profiling in relapsed/refractory MM patient samples confirmed that high KANSL2 expression is associated with selective MM cell killing by HDAC and BET inhibitors. Collectively, these findings position KANSL2 as a mediator of chemotherapy resistance and actionable biomarker for response to drugs targeting its epigenetic program.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145605053","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
Discovery of potent and brain-penetrable tubulin inhibitors that effectively suppress breast cancer brain metastasis. 发现有效抑制乳腺癌脑转移的强效和可穿透脑的微管蛋白抑制剂。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-24 DOI: 10.1158/1535-7163.MCT-25-0538
Raisa I Krutilina, Kelli L Adeleye, Hilaire C Playa, Satyanarayana Pochampally, Souvik Banjeree, Damilola Oluwalana, Mir Shahriar Kamal, Di Tian, Duane D Miller, Wei Li, Tiffany N Seagroves

Breast cancer brain metastasis (BCBM) remains a clinical challenge marked by limited therapeutic options and poor survival rates. Approximately 30% of all metastatic breast cancer patients develop BCBM, with the highest incidence in patients with aggressive molecular subtypes, including triple-negative breast cancer (TNBC). TNBC patients with brain metastasis experience rapid disease progression and significantly reduced survival times due to a lack of targeted treatments that can penetrate the blood-brain barrier (BBB) and effectively control metastatic expansion. Current treatment options, such as whole-brain radiotherapy and chemotherapy, offer limited efficacy and are associated with significant toxicities, underscoring the urgent need for novel therapeutics that can target BCBM directly. We developed innovative colchicine binding site inhibitors (CBSIs) targeting tubulin, SB-216 and SP-1-39, that show potent preclinical efficacy against brain and extracranial metastases in BCBM models. Both CBSIs cross the BBB, inhibit cell growth and migration, and induce apoptosis with low nM potencies, similar to Azixa (MPC-6827), another CBSI previously evaluated in clinical trials. SB-216 reduced brain and concurrent extracranial metastases in a preventive dosing paradigm, extending overall survival. SB-216 also suppressed the expansion of pre-established brain lesions. In a taxane-refractory TNBC PDX model, SP-1-39 therapy markedly reduced brain and extracranial tumor burden. Together, these results highlight the promising therapeutic potential of SB-216 and SP-1-39 in treating taxane-sensitive or -resistant BCBM, filling a critical gap in TNBC management by offering targeted treatments that can cross the BBB and combat chemorefractory disease.

乳腺癌脑转移(BCBM)仍然是一个临床挑战,其特点是治疗选择有限,生存率低。大约30%的转移性乳腺癌患者发生BCBM,在侵袭性分子亚型患者中发病率最高,包括三阴性乳腺癌(TNBC)。由于缺乏能够穿透血脑屏障(BBB)并有效控制转移扩张的靶向治疗,伴有脑转移的TNBC患者疾病进展迅速,生存时间显着缩短。目前的治疗方案,如全脑放疗和化疗,提供有限的疗效,并与显著的毒性相关,强调迫切需要新的治疗方法,可以直接针对BCBM。我们开发了针对微管蛋白、SB-216和SP-1-39的创新秋水仙碱结合位点抑制剂(CBSIs),在BCBM模型中对脑和颅外转移显示出强有力的临床前疗效。这两种CBSI都能穿过血脑屏障,抑制细胞生长和迁移,并以低nM效价诱导细胞凋亡,类似于Azixa (MPC-6827),这是另一种CBSI,此前在临床试验中进行了评估。SB-216在预防性给药模式下减少脑和并发颅外转移,延长总生存期。SB-216还抑制了预先建立的脑病变的扩张。在紫杉烷难治性TNBC PDX模型中,SP-1-39治疗可显著减轻脑和颅外肿瘤负担。总之,这些结果突出了SB-216和SP-1-39在治疗紫杉烷敏感或耐药的BCBM方面的治疗潜力,通过提供可以跨越血脑屏障和对抗化疗难治疾病的靶向治疗,填补了TNBC管理的关键空白。
{"title":"Discovery of potent and brain-penetrable tubulin inhibitors that effectively suppress breast cancer brain metastasis.","authors":"Raisa I Krutilina, Kelli L Adeleye, Hilaire C Playa, Satyanarayana Pochampally, Souvik Banjeree, Damilola Oluwalana, Mir Shahriar Kamal, Di Tian, Duane D Miller, Wei Li, Tiffany N Seagroves","doi":"10.1158/1535-7163.MCT-25-0538","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0538","url":null,"abstract":"<p><p>Breast cancer brain metastasis (BCBM) remains a clinical challenge marked by limited therapeutic options and poor survival rates. Approximately 30% of all metastatic breast cancer patients develop BCBM, with the highest incidence in patients with aggressive molecular subtypes, including triple-negative breast cancer (TNBC). TNBC patients with brain metastasis experience rapid disease progression and significantly reduced survival times due to a lack of targeted treatments that can penetrate the blood-brain barrier (BBB) and effectively control metastatic expansion. Current treatment options, such as whole-brain radiotherapy and chemotherapy, offer limited efficacy and are associated with significant toxicities, underscoring the urgent need for novel therapeutics that can target BCBM directly. We developed innovative colchicine binding site inhibitors (CBSIs) targeting tubulin, SB-216 and SP-1-39, that show potent preclinical efficacy against brain and extracranial metastases in BCBM models. Both CBSIs cross the BBB, inhibit cell growth and migration, and induce apoptosis with low nM potencies, similar to Azixa (MPC-6827), another CBSI previously evaluated in clinical trials. SB-216 reduced brain and concurrent extracranial metastases in a preventive dosing paradigm, extending overall survival. SB-216 also suppressed the expansion of pre-established brain lesions. In a taxane-refractory TNBC PDX model, SP-1-39 therapy markedly reduced brain and extracranial tumor burden. Together, these results highlight the promising therapeutic potential of SB-216 and SP-1-39 in treating taxane-sensitive or -resistant BCBM, filling a critical gap in TNBC management by offering targeted treatments that can cross the BBB and combat chemorefractory disease.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145588450","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
AXL kinase inhibition promotes cytosolic DNA sensor cGAS activity and sensitizes poorly immunogenic tumors to chemo-immunotherapy. AXL激酶抑制促进细胞质DNA传感器cGAS活性,使免疫原性差的肿瘤对化学免疫治疗敏感。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-20 DOI: 10.1158/1535-7163.MCT-24-0440
Sushil Dhakal, Muntequa I Siraji, Sturla M Grøndal, Gard N Skarsten, Emmanuel E Moutoussamy, Gro Gausdal, James B Lorens, Sébastien Bougnaud

AXL is an important negative regulator of type I interferon (IFN) responses during viral infections. In the context of tumors, AXL is associated with driving tumor progression, spread, immune evasion, and therapy resistance. AXL regulation of tumor cell intrinsic IFN responses remains unexplored. We show that AXL suppresses tumor intrinsic IFN responses by inhibiting the cytosolic DNA sensor cGAS via an AKT-dependent pathway. AXL inhibition in combination with chemo-immunotherapy demonstrated potent anti-tumor effects in poorly immunogenic tumors that are refractory to immunotherapy. The inhibition of AXL correlated with increased cGAMP levels, activation of IFN, and enhanced infiltration of T cells and NK cells into the tumor microenvironment. These findings reveal a novel role for AXL in suppressing IFN within tumors and support AXL targeting as a promising strategy in conjunction with chemo-immunotherapy for treating therapy-resistant tumors.

AXL是病毒感染期间I型干扰素(IFN)应答的重要负调节因子。在肿瘤的背景下,AXL与驱动肿瘤进展、扩散、免疫逃避和治疗抵抗有关。AXL对肿瘤细胞内生性IFN反应的调控尚不清楚。我们发现AXL通过akt依赖性途径抑制细胞质DNA传感器cGAS,从而抑制肿瘤内生性IFN反应。AXL抑制联合化学免疫治疗在免疫治疗难治性低免疫原性肿瘤中显示出有效的抗肿瘤作用。AXL的抑制与cGAMP水平升高、IFN激活、T细胞和NK细胞向肿瘤微环境的浸润增强相关。这些发现揭示了AXL在肿瘤中抑制IFN的新作用,并支持AXL靶向作为一种有希望的策略,与化学免疫疗法一起治疗治疗耐药肿瘤。
{"title":"AXL kinase inhibition promotes cytosolic DNA sensor cGAS activity and sensitizes poorly immunogenic tumors to chemo-immunotherapy.","authors":"Sushil Dhakal, Muntequa I Siraji, Sturla M Grøndal, Gard N Skarsten, Emmanuel E Moutoussamy, Gro Gausdal, James B Lorens, Sébastien Bougnaud","doi":"10.1158/1535-7163.MCT-24-0440","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0440","url":null,"abstract":"<p><p>AXL is an important negative regulator of type I interferon (IFN) responses during viral infections. In the context of tumors, AXL is associated with driving tumor progression, spread, immune evasion, and therapy resistance. AXL regulation of tumor cell intrinsic IFN responses remains unexplored. We show that AXL suppresses tumor intrinsic IFN responses by inhibiting the cytosolic DNA sensor cGAS via an AKT-dependent pathway. AXL inhibition in combination with chemo-immunotherapy demonstrated potent anti-tumor effects in poorly immunogenic tumors that are refractory to immunotherapy. The inhibition of AXL correlated with increased cGAMP levels, activation of IFN, and enhanced infiltration of T cells and NK cells into the tumor microenvironment. These findings reveal a novel role for AXL in suppressing IFN within tumors and support AXL targeting as a promising strategy in conjunction with chemo-immunotherapy for treating therapy-resistant tumors.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145557542","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
MCT8 modulates metabolite uptake and T cell exhaustion to promote immunosuppression and tumor progression in hepatocellular carcinoma. MCT8调节代谢物摄取和T细胞耗竭,促进肝细胞癌的免疫抑制和肿瘤进展。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-20 DOI: 10.1158/1535-7163.MCT-25-0564
Bin Bi, Liqin Tang, Ranxi Liang, Haijing Wu, Qilan Huang, Li Pan

Solute carrier family 16 member 2 (SLC16A2), also known as monocarboxylate transporter 8 (MCT8), is a member of the SLC16 family that exerts essential functions in the transport of elemental cell nutrients. This study explores the function of SLC16A2 in hepatocellular carcinoma (HCC) progression, particularly its impact on T cell function. We established Slc16a2 gene knockout (Slc16a2ko) C57BL/6 mice and injected H22 cells subcutaneously to analyze tumor growth and T cell activity in vivo. Additionally, Slc16a2fl/fl mice were crossed with Cd8aCre mice to obtain Slc16a2-Cd8a-ko mice, where Slc16a2 was specifically knocked out in CD8+T cells. In addition to subcutaneous models, luciferase-labeled H22 cells were injected into the liver lobe of mice for orthotopic models. SLC16A2 alteration did not affect proliferation or migration of mouse Hepa1-6 and H22 cells in vitro, though, tumorigenic activity of H22 cells was substantially reduced in Slc16a2ko and Slc16a2-Cd8a-ko C57BL/6 mice. Slc6a2 was highly expressed in Tex cells, and its expression in Tex, as well as the population of Tex cells in tumors, was increased by lactate or other chronic stimuli. An MCT8 monoclonal antibody (mAb) reduced lactate uptake by Tex cells, thus enhancing CD8+ T cell activity and reducing tumor growth in mice. The MCT8 mAb treatment also enhanced the efficacy of anti-PD-L1 in mice bearing tumors. This study supports that SLC16A2 contributes to Tex accumulation in association with increased lactate uptake and hampers immune activity in HCC, supporting SLC16A2 as a promising target to enhance immune activity in HCC management.

溶质载体家族16成员2 (SLC16A2),也被称为单羧酸转运体8 (MCT8),是SLC16家族的一员,在细胞元素营养物质的运输中发挥重要作用。本研究探讨了SLC16A2在肝细胞癌(HCC)进展中的功能,特别是其对T细胞功能的影响。我们建立Slc16a2基因敲除(Slc16a2ko) C57BL/6小鼠,皮下注射H22细胞,在体内分析肿瘤生长和T细胞活性。此外,将Slc16a2fl/fl小鼠与Cd8aCre小鼠杂交获得Slc16a2- cd8a -ko小鼠,其中Slc16a2在CD8+T细胞中被特异性敲除。除皮下模型外,还将荧光素酶标记的H22细胞注射到小鼠肝叶中建立原位模型。SLC16A2的改变不影响小鼠Hepa1-6和H22细胞的体外增殖和迁移,但在Slc16a2ko和SLC16A2 - cd8a -ko C57BL/6小鼠中,H22细胞的致瘤活性明显降低。Slc6a2在Tex细胞中高表达,其在Tex中的表达,以及肿瘤中Tex细胞的数量,在乳酸或其他慢性刺激下增加。MCT8单克隆抗体(mAb)减少了Tex细胞对乳酸的摄取,从而增强了CD8+ T细胞的活性,降低了小鼠肿瘤的生长。MCT8单抗治疗也增强了荷瘤小鼠抗pd - l1的疗效。本研究支持SLC16A2促进Tex积累,增加乳酸摄取并阻碍HCC的免疫活性,支持SLC16A2作为HCC管理中增强免疫活性的有希望的靶点。
{"title":"MCT8 modulates metabolite uptake and T cell exhaustion to promote immunosuppression and tumor progression in hepatocellular carcinoma.","authors":"Bin Bi, Liqin Tang, Ranxi Liang, Haijing Wu, Qilan Huang, Li Pan","doi":"10.1158/1535-7163.MCT-25-0564","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0564","url":null,"abstract":"<p><p>Solute carrier family 16 member 2 (SLC16A2), also known as monocarboxylate transporter 8 (MCT8), is a member of the SLC16 family that exerts essential functions in the transport of elemental cell nutrients. This study explores the function of SLC16A2 in hepatocellular carcinoma (HCC) progression, particularly its impact on T cell function. We established Slc16a2 gene knockout (Slc16a2ko) C57BL/6 mice and injected H22 cells subcutaneously to analyze tumor growth and T cell activity in vivo. Additionally, Slc16a2fl/fl mice were crossed with Cd8aCre mice to obtain Slc16a2-Cd8a-ko mice, where Slc16a2 was specifically knocked out in CD8+T cells. In addition to subcutaneous models, luciferase-labeled H22 cells were injected into the liver lobe of mice for orthotopic models. SLC16A2 alteration did not affect proliferation or migration of mouse Hepa1-6 and H22 cells in vitro, though, tumorigenic activity of H22 cells was substantially reduced in Slc16a2ko and Slc16a2-Cd8a-ko C57BL/6 mice. Slc6a2 was highly expressed in Tex cells, and its expression in Tex, as well as the population of Tex cells in tumors, was increased by lactate or other chronic stimuli. An MCT8 monoclonal antibody (mAb) reduced lactate uptake by Tex cells, thus enhancing CD8+ T cell activity and reducing tumor growth in mice. The MCT8 mAb treatment also enhanced the efficacy of anti-PD-L1 in mice bearing tumors. This study supports that SLC16A2 contributes to Tex accumulation in association with increased lactate uptake and hampers immune activity in HCC, supporting SLC16A2 as a promising target to enhance immune activity in HCC management.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145557645","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
Involvement of the FGF8/FGF receptor signaling pathway in the maintenance and progression of fusion-positive rhabdomyosarcoma. FGF8/FGF受体信号通路参与融合阳性横纹肌肉瘤的维持和发展
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-20 DOI: 10.1158/1535-7163.MCT-24-0328
Salah Boudjadi, Hana Kim, Bishwanath Chatterjee, Pawan Kumar Raut, Thanh Hung Nguyen, Puspa Raj Pandey, Reyaz Ur Rasool, Wenyue Sun, Frederic G Barr

We previously used a myoblast model of fusion-positive rhabdomyosarcoma (FP-RMS) to show that FGF8, a PAX3-FOXO1 (P3F) transcriptional target, is required for P3F-driven tumorigenicity and, when aberrantly expressed, can maintain tumorigenicity in P3F-independent recurrent tumors. We report in this study that FGF8, FGFR1 and FGFR4 are often highly expressed in FP-RMS tumors. High FGF8 expression in FP-RMS cells is associated with high sensitivity to an FGFR4 inhibitor and a pan-FGFR inhibitor. While downregulating FGF8 resulted in loss of sensitivity to these inhibitors, FGF8 upregulation in myoblasts decreased FGFR4 expression and sensitized the cells to an FGFR1 inhibitor and a pan-FGFR inhibitor. FGF8 downregulation of FGFR4 expression was reverted by inhibitors of FGFR1, MEK or ERK, thus defining a signaling pathway by which FGF8 mediates this regulatory effect. Finally, high FGF8 expression in P3F-independent recurrent tumors was attributable to a rearrangement of viral LTR sequences into the FGF8 3' UTR region, resulting in increased FGF8 mRNA stability. These findings indicate that FGF8 exerts oncogenic effects in FP-RMS via FGFR4 and may exert oncogenic effects in P3F-independent relapses via FGFR1. Our study reveals the functional significance of FGF8 in FP-RMS and provides a rationale for preclinical studies of FGFR inhibitors in FP-RMS.

我们之前使用融合阳性横纹肌肉瘤(FP-RMS)的成肌细胞模型表明,FGF8, pax3 - fox01 (P3F)转录靶点,是P3F驱动的致瘤性所必需的,当异常表达时,可以在不依赖P3F的复发肿瘤中维持致瘤性。我们在本研究中报道,FGF8、FGFR1和FGFR4通常在FP-RMS肿瘤中高表达。FGF8在FP-RMS细胞中的高表达与FGFR4抑制剂和泛fgfr抑制剂的高敏感性相关。虽然下调FGF8导致对这些抑制剂的敏感性丧失,但在成肌细胞中上调FGF8会降低FGFR4的表达,并使细胞对FGFR1抑制剂和泛fgfr抑制剂敏感。FGFR1、MEK或ERK抑制剂可以逆转FGF8对FGFR4表达的下调,从而确定了FGF8介导这种调节作用的信号通路。最后,在不依赖p3f的复发肿瘤中,FGF8的高表达可归因于病毒LTR序列重排到FGF8 3' UTR区域,从而增加了FGF8 mRNA的稳定性。这些发现表明,FGF8通过FGFR4在FP-RMS中发挥致癌作用,并可能通过FGFR1在不依赖p3f的复发中发挥致癌作用。我们的研究揭示了FGF8在FP-RMS中的功能意义,并为FGFR抑制剂在FP-RMS中的临床前研究提供了依据。
{"title":"Involvement of the FGF8/FGF receptor signaling pathway in the maintenance and progression of fusion-positive rhabdomyosarcoma.","authors":"Salah Boudjadi, Hana Kim, Bishwanath Chatterjee, Pawan Kumar Raut, Thanh Hung Nguyen, Puspa Raj Pandey, Reyaz Ur Rasool, Wenyue Sun, Frederic G Barr","doi":"10.1158/1535-7163.MCT-24-0328","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0328","url":null,"abstract":"<p><p>We previously used a myoblast model of fusion-positive rhabdomyosarcoma (FP-RMS) to show that FGF8, a PAX3-FOXO1 (P3F) transcriptional target, is required for P3F-driven tumorigenicity and, when aberrantly expressed, can maintain tumorigenicity in P3F-independent recurrent tumors. We report in this study that FGF8, FGFR1 and FGFR4 are often highly expressed in FP-RMS tumors. High FGF8 expression in FP-RMS cells is associated with high sensitivity to an FGFR4 inhibitor and a pan-FGFR inhibitor. While downregulating FGF8 resulted in loss of sensitivity to these inhibitors, FGF8 upregulation in myoblasts decreased FGFR4 expression and sensitized the cells to an FGFR1 inhibitor and a pan-FGFR inhibitor. FGF8 downregulation of FGFR4 expression was reverted by inhibitors of FGFR1, MEK or ERK, thus defining a signaling pathway by which FGF8 mediates this regulatory effect. Finally, high FGF8 expression in P3F-independent recurrent tumors was attributable to a rearrangement of viral LTR sequences into the FGF8 3' UTR region, resulting in increased FGF8 mRNA stability. These findings indicate that FGF8 exerts oncogenic effects in FP-RMS via FGFR4 and may exert oncogenic effects in P3F-independent relapses via FGFR1. Our study reveals the functional significance of FGF8 in FP-RMS and provides a rationale for preclinical studies of FGFR inhibitors in FP-RMS.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145557531","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
Pertuzumab enhances the antitumor activity of T-DXd in HER2-positive gastric cancer cells. 帕妥珠单抗增强T-DXd在her2阳性胃癌细胞中的抗肿瘤活性。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-13 DOI: 10.1158/1535-7163.MCT-24-0453
Minsu Kang, Kui-Jin Kim, Hyeon Jeong Oh, Ji Hea Sung, Milang Nam, Bo-Ram Park, Sung-Hyun Hwang, Eun Hee Jung, Koung Jin Suh, Ji-Won Kim, Se Hyun Kim, Jin Won Kim, Yu Jung Kim, Jee Hyun Kim, Keun-Wook Lee

For HER2-positive advanced gastric cancer (GC), a recent major therapeutic advancement is the development of trastuzumab deruxtecan (T-DXd), a HER2-directed antibody-drug conjugate. In this disease, simultaneously targeting HER2 and HER3 pathways has the potential to be a promising therapeutic strategy. However, the therapeutic approach of combining T-DXd with pertuzumab, which disrupts HER2-HER3 heterodimerization, has not yet been explored in GC, making this study a pioneering effort. In vitro, T-DXd efficacy correlated with high levels of membrane HER2 expression. Among the 12 cell lines tested, two cell lines (NCI-N87 and OE19) confirmed as HER2 3+ by immunohistochemistry showed the most effective proliferation inhibition by T-DXd. When comparing NCI-N87 and OE19, HER2-HER3 dimerization was found to be more abundant in NCI-N87, and combination treatment with pertuzumab and T-DXd showed synergy in cell growth inhibition in NCI-N87, but not in OE19. NRG1 stimulation attenuated the antiproliferative effect of T-DXd. This attenuation of T-DXd activity by NRG1 was partially reversed by the addition of pertuzumab in NCI-N87, but not in OE19. Notably, the combination of T-DXd and pertuzumab enhanced membrane HER2 internalization more effectively in NCI-N87 than in OE19. In vivo mouse experiments using NCI-N87 cells showed the combination treatment significantly suppressed tumor growth compared to either monotherapy. Taken together, our findings suggest that dual targeting of HER2 and HER3 with T-DXd and pertuzumab may improve therapeutic outcomes in HER2-positive GC, particularly in tumors enriched with HER2-HER3 heterodimers. These preclinical data provide strong rationale for clinical trials evaluating this combination strategy in HER2-positive GC.

对于her2阳性晚期胃癌(GC),最近的主要治疗进展是曲妥珠单抗德鲁西替康(T-DXd)的开发,这是一种her2导向的抗体-药物偶联物。在这种疾病中,同时靶向HER2和HER3通路有可能成为一种有前途的治疗策略。然而,T-DXd联合pertuzumab的治疗方法,破坏HER2-HER3异源二聚化,尚未在GC中探索,使本研究成为开创性的努力。在体外,T-DXd的疗效与高水平的膜HER2表达相关。在12个细胞系中,免疫组化证实为HER2 3+的2个细胞系NCI-N87和OE19对T-DXd的增殖抑制效果最好。对比NCI-N87和OE19,发现HER2-HER3二聚体在NCI-N87中更丰富,并且帕妥珠单抗和T-DXd联合治疗在NCI-N87中显示出协同抑制细胞生长的作用,而在OE19中没有。NRG1刺激可减弱T-DXd的抗增殖作用。NRG1对T-DXd活性的减弱在NCI-N87中被添加pertuzumab部分逆转,但在OE19中没有。值得注意的是,在NCI-N87中,T-DXd和pertuzumab联合使用比在OE19中更有效地增强了膜HER2的内化。使用NCI-N87细胞的小鼠体内实验显示,与单一治疗相比,联合治疗显著抑制肿瘤生长。综上所述,我们的研究结果表明,T-DXd和帕妥珠单抗联合靶向HER2和HER3可能改善HER2阳性胃癌的治疗效果,特别是在富含HER2-HER3异源二聚体的肿瘤中。这些临床前数据为在her2阳性GC中评估这种联合策略的临床试验提供了强有力的依据。
{"title":"Pertuzumab enhances the antitumor activity of T-DXd in HER2-positive gastric cancer cells.","authors":"Minsu Kang, Kui-Jin Kim, Hyeon Jeong Oh, Ji Hea Sung, Milang Nam, Bo-Ram Park, Sung-Hyun Hwang, Eun Hee Jung, Koung Jin Suh, Ji-Won Kim, Se Hyun Kim, Jin Won Kim, Yu Jung Kim, Jee Hyun Kim, Keun-Wook Lee","doi":"10.1158/1535-7163.MCT-24-0453","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0453","url":null,"abstract":"<p><p>For HER2-positive advanced gastric cancer (GC), a recent major therapeutic advancement is the development of trastuzumab deruxtecan (T-DXd), a HER2-directed antibody-drug conjugate. In this disease, simultaneously targeting HER2 and HER3 pathways has the potential to be a promising therapeutic strategy. However, the therapeutic approach of combining T-DXd with pertuzumab, which disrupts HER2-HER3 heterodimerization, has not yet been explored in GC, making this study a pioneering effort. In vitro, T-DXd efficacy correlated with high levels of membrane HER2 expression. Among the 12 cell lines tested, two cell lines (NCI-N87 and OE19) confirmed as HER2 3+ by immunohistochemistry showed the most effective proliferation inhibition by T-DXd. When comparing NCI-N87 and OE19, HER2-HER3 dimerization was found to be more abundant in NCI-N87, and combination treatment with pertuzumab and T-DXd showed synergy in cell growth inhibition in NCI-N87, but not in OE19. NRG1 stimulation attenuated the antiproliferative effect of T-DXd. This attenuation of T-DXd activity by NRG1 was partially reversed by the addition of pertuzumab in NCI-N87, but not in OE19. Notably, the combination of T-DXd and pertuzumab enhanced membrane HER2 internalization more effectively in NCI-N87 than in OE19. In vivo mouse experiments using NCI-N87 cells showed the combination treatment significantly suppressed tumor growth compared to either monotherapy. Taken together, our findings suggest that dual targeting of HER2 and HER3 with T-DXd and pertuzumab may improve therapeutic outcomes in HER2-positive GC, particularly in tumors enriched with HER2-HER3 heterodimers. These preclinical data provide strong rationale for clinical trials evaluating this combination strategy in HER2-positive GC.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145505919","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
NOUS-209 off-the-shelf immunotherapy has the potential to hit primary and metachronous colorectal and urothelial cancer in Lynch syndrome. NOUS-209现成的免疫疗法有可能治疗Lynch综合征的原发性和异时性结直肠癌和尿路上皮癌。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-13 DOI: 10.1158/1535-7163.MCT-25-0864
Lorenzo De Marco, Elisa Micarelli, Joni Panula, Jussi Nikkola, Lauri Moilanen, Matti Annala, Jouni Härkönen, Kalle E Hokkanen, Anna Morena D'Alise, Kirsi Pylvänäinen, Päivi T Peltomäki, Maarit Ahtiainen, Jan Böhm, Jukka-Pekka Mecklin, Elisa Scarselli, Toni T Seppälä

Lynch syndrome (LS) is characterized by the development of microsatellite instable (MSI) cancers that share neoantigens, offering an opportunity for targeted immunotherapy. NOUS-209 is a heterologous prime-boost cancer vaccine in clinical development, employing viral vectors encoding 209 shared neoantigen peptides derived from frameshift mutations (FSMs) commonly found in MSI tumors. In this study, we investigated the presence and dynamics of NOUS-209 targeted FSMs in both primary and metachronous LS-associated cancers. Whole-exome sequencing was performed for 73 tumors, including 58 colorectal cancers (CRC) and 15 urothelial cancers (UC), from 58 individuals with confirmed LS. A median of 57 FSMs per CRC and 24 FSMs per UC was observed, with similar FSM burdens in both primary and metachronous tumors. Analysis of nine matched primary-metachronous tumor pairs revealed evidence of immune editing: FSMs predicted to encode highly immunogenic neoepitopes were selectively lost in metachronous tumors, while those with lower predicted immunogenicity were retained. Importantly, all subsequent primary cancers acquired novel FSMs encoding neoantigens with strong predicted HLA-binding affinity, supporting the rationale for NOUS-209-mediated immune interception. These findings demonstrated that NOUS-209 FSMs are present in both CRCs and UCs in LS, expanding the therapeutic potential of NOUS-209 beyond CRC. Moreover, the emergence of novel targetable FSMs in metachronous tumors suggests that NOUS-209 immunotherapy may be effective in the prevention of both primary and metachronous LS-associated cancers.

Lynch综合征(LS)的特点是发展为共享新抗原的微卫星不稳定(MSI)癌症,为靶向免疫治疗提供了机会。NOUS-209是一种处于临床开发阶段的异源癌疫苗,利用病毒载体编码209个共享的新抗原肽,这些新抗原肽来源于MSI肿瘤中常见的移码突变(FSMs)。在这项研究中,我们研究了NOUS-209靶向FSMs在原发性和异时性ls相关癌症中的存在和动态。对来自58例确诊LS患者的73个肿瘤进行了全外显子组测序,包括58例结直肠癌(CRC)和15例尿路上皮癌(UC)。观察到每个结直肠癌中有57个FSM,每个UC中有24个FSM,原发和异时性肿瘤中有相似的FSM负担。对9对匹配的原发异时性肿瘤的分析揭示了免疫编辑的证据:预测编码高免疫原性新表位的fsm在异时性肿瘤中选择性丢失,而预测免疫原性较低的fsm保留下来。重要的是,所有随后的原发性癌症都获得了编码新抗原的新型FSMs,具有很强的预测hla结合亲和力,支持nous -209介导的免疫拦截的基本原理。这些发现表明,NOUS-209 FSMs存在于LS的CRC和UCs中,扩大了NOUS-209在CRC以外的治疗潜力。此外,异时性肿瘤中新型靶向fsm的出现表明,NOUS-209免疫疗法可能有效预防原发性和异时性ls相关癌症。
{"title":"NOUS-209 off-the-shelf immunotherapy has the potential to hit primary and metachronous colorectal and urothelial cancer in Lynch syndrome.","authors":"Lorenzo De Marco, Elisa Micarelli, Joni Panula, Jussi Nikkola, Lauri Moilanen, Matti Annala, Jouni Härkönen, Kalle E Hokkanen, Anna Morena D'Alise, Kirsi Pylvänäinen, Päivi T Peltomäki, Maarit Ahtiainen, Jan Böhm, Jukka-Pekka Mecklin, Elisa Scarselli, Toni T Seppälä","doi":"10.1158/1535-7163.MCT-25-0864","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0864","url":null,"abstract":"<p><p>Lynch syndrome (LS) is characterized by the development of microsatellite instable (MSI) cancers that share neoantigens, offering an opportunity for targeted immunotherapy. NOUS-209 is a heterologous prime-boost cancer vaccine in clinical development, employing viral vectors encoding 209 shared neoantigen peptides derived from frameshift mutations (FSMs) commonly found in MSI tumors. In this study, we investigated the presence and dynamics of NOUS-209 targeted FSMs in both primary and metachronous LS-associated cancers. Whole-exome sequencing was performed for 73 tumors, including 58 colorectal cancers (CRC) and 15 urothelial cancers (UC), from 58 individuals with confirmed LS. A median of 57 FSMs per CRC and 24 FSMs per UC was observed, with similar FSM burdens in both primary and metachronous tumors. Analysis of nine matched primary-metachronous tumor pairs revealed evidence of immune editing: FSMs predicted to encode highly immunogenic neoepitopes were selectively lost in metachronous tumors, while those with lower predicted immunogenicity were retained. Importantly, all subsequent primary cancers acquired novel FSMs encoding neoantigens with strong predicted HLA-binding affinity, supporting the rationale for NOUS-209-mediated immune interception. These findings demonstrated that NOUS-209 FSMs are present in both CRCs and UCs in LS, expanding the therapeutic potential of NOUS-209 beyond CRC. Moreover, the emergence of novel targetable FSMs in metachronous tumors suggests that NOUS-209 immunotherapy may be effective in the prevention of both primary and metachronous LS-associated cancers.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145513283","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
Preclinical development and a case report of a nanobody-based CLDN18.2 CAR-T IMC002 with reduced on-target off-tumor toxicity. 基于纳米体的CLDN18.2 CAR-T IMC002的临床前开发和病例报告,降低了靶外肿瘤毒性。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-10 DOI: 10.1158/1535-7163.MCT-25-0171
Sicheng Du, Ying Zhang, Ruidong Hao, Lupeng Qiu, Chao Li, Yuting Li, Rong-Rui Liu, Chuan-Hua Zhao, Juan Li, Sisi Ye, Jun Zhou, Yantao Li, Qiaoyong Yi, Shuangshuang Zhang, Minmin Sun, Tianhang Luo, Jianming Xu

Claudin18.2 (CLDN18.2)-targeted chimeric antigen receptor T (CAR-T) cell therapy has shown promising antitumor activity in gastrointestinal cancers. However, limited persistence in solid tumors and on-target off-tumor (OTOT) toxicity remain significant challenges. Here, we report on the preclinical development of a nanobody-based CLDN18.2-targeted CAR-T (IMC002) with effectiveness and safety in CLDN18.2-positive gastric and pancreatic cancer and present an efficacious clinical case. IMC002 exhibited robust antitumor activity and tolerability in multiple CLDN18.2-positive cell-derived xenograft and patient-derived xenograft models of gastric and pancreatic cancer with reduced OTOT toxicity. In vivo pharmacological studies revealed that peak concentrations of CAR gene DNA copies in total DNA in the tumor and lung tissues occurred on seven days after administration, while the peak in stomach tissues was observed an additional seven days later. Toxicity studies showed no obvious body weight loss induced by IMC002. The highest non-severely toxic dose was 5×108 CAR-T cells/kg. In the clinical case report, we present a case with unresectable advanced gastric cancer achieved pathological complete response 10 months after IMC002 infusion and no signs of recurrence were indicated in subsequent clinical and radiological follow-ups. IMC002 shows effectiveness and safety in CLDN18.2-positive gastric and pancreatic cancer and its favorable profiles support further clinical development.

CLDN18.2 (CLDN18.2)靶向嵌合抗原受体T (CAR-T)细胞治疗在胃肠道肿瘤中显示出良好的抗肿瘤活性。然而,有限的持久性实体瘤和靶外肿瘤(OTOT)毒性仍然是一个重大挑战。在这里,我们报告了一种基于纳米体的cldn18.2靶向CAR-T (IMC002)的临床前开发,该药物对cldn18.2阳性的胃癌和胰腺癌具有有效性和安全性,并提出了一个有效的临床病例。IMC002在多种cldn18.2阳性细胞源异种移植瘤和患者源异种移植瘤模型中表现出强大的抗肿瘤活性和耐受性,并降低了OTOT毒性。体内药理学研究显示,在给药后7天,肿瘤和肺组织中总DNA中CAR基因DNA拷贝的浓度达到峰值,而胃组织中CAR基因DNA拷贝的浓度又在给药后7天达到峰值。毒性研究表明,IMC002没有引起明显的体重减轻。最高非严重毒性剂量为5×108 CAR-T细胞/kg。在临床病例报告中,我们报告了一例不可切除的晚期胃癌患者,在IMC002输注10个月后达到病理完全缓解,在随后的临床和放射学随访中未发现复发迹象。IMC002在cldn18.2阳性胃癌和胰腺癌中显示出有效性和安全性,其良好的特性支持进一步的临床开发。
{"title":"Preclinical development and a case report of a nanobody-based CLDN18.2 CAR-T IMC002 with reduced on-target off-tumor toxicity.","authors":"Sicheng Du, Ying Zhang, Ruidong Hao, Lupeng Qiu, Chao Li, Yuting Li, Rong-Rui Liu, Chuan-Hua Zhao, Juan Li, Sisi Ye, Jun Zhou, Yantao Li, Qiaoyong Yi, Shuangshuang Zhang, Minmin Sun, Tianhang Luo, Jianming Xu","doi":"10.1158/1535-7163.MCT-25-0171","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0171","url":null,"abstract":"<p><p>Claudin18.2 (CLDN18.2)-targeted chimeric antigen receptor T (CAR-T) cell therapy has shown promising antitumor activity in gastrointestinal cancers. However, limited persistence in solid tumors and on-target off-tumor (OTOT) toxicity remain significant challenges. Here, we report on the preclinical development of a nanobody-based CLDN18.2-targeted CAR-T (IMC002) with effectiveness and safety in CLDN18.2-positive gastric and pancreatic cancer and present an efficacious clinical case. IMC002 exhibited robust antitumor activity and tolerability in multiple CLDN18.2-positive cell-derived xenograft and patient-derived xenograft models of gastric and pancreatic cancer with reduced OTOT toxicity. In vivo pharmacological studies revealed that peak concentrations of CAR gene DNA copies in total DNA in the tumor and lung tissues occurred on seven days after administration, while the peak in stomach tissues was observed an additional seven days later. Toxicity studies showed no obvious body weight loss induced by IMC002. The highest non-severely toxic dose was 5×108 CAR-T cells/kg. In the clinical case report, we present a case with unresectable advanced gastric cancer achieved pathological complete response 10 months after IMC002 infusion and no signs of recurrence were indicated in subsequent clinical and radiological follow-ups. IMC002 shows effectiveness and safety in CLDN18.2-positive gastric and pancreatic cancer and its favorable profiles support further clinical development.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145482553","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
T2R5 agonist phendione decreases cell viability and induces apoptosis in head and neck squamous cell carcinoma. T2R5激动剂苯地酮降低头颈部鳞状细胞癌细胞活力,诱导细胞凋亡。
IF 5.5 2区 医学 Q1 ONCOLOGY Pub Date : 2025-11-07 DOI: 10.1158/1535-7163.MCT-25-0358
Sarah Sywanycz, Brianna L Hill, Zoey A Miller, Gavin Turner, Lily Huang, Kyle Polen, Robert J Lee, Ryan M Carey

Bitter taste receptors (T2Rs), a family of G-protein coupled receptors, are emerging as potential therapeutic targets in head and neck squamous cell carcinoma (HNSCC). Phendione, a known T2R5 agonist, has not been previously investigated in HNSCC. Here, we show that phendione activates endogenously expressed T2R5 in HNSCC cells and ex vivo tumor samples, inducing sustained calcium responses, reducing cell viability, and promoting apoptosis through a T2R5-dependent mechanism. Analysis of The Cancer Genome Atlas data revealed that high T2R5 expression in HNSCC tumors correlates with improved long-term disease-specific survival, suggesting a potential tumor-suppressive role for T2R5. These findings highlight T2R5 as a promising therapeutic target in HNSCC and support further investigation of phendione or other T2R5 agonists as potential anti-cancer agents.

苦味受体(T2Rs)是一个g蛋白偶联受体家族,正在成为头颈部鳞状细胞癌(HNSCC)的潜在治疗靶点。Phendione是一种已知的T2R5激动剂,以前没有在HNSCC中研究过。本研究表明,苯二酮激活HNSCC细胞和离体肿瘤样本中内源性表达的T2R5,诱导持续的钙反应,降低细胞活力,并通过T2R5依赖机制促进细胞凋亡。对癌症基因组图谱数据的分析显示,高T2R5在HNSCC肿瘤中的表达与提高长期疾病特异性生存率相关,提示T2R5具有潜在的肿瘤抑制作用。这些发现强调了T2R5作为HNSCC的一个有希望的治疗靶点,并支持进一步研究phendione或其他T2R5激动剂作为潜在的抗癌药物。
{"title":"T2R5 agonist phendione decreases cell viability and induces apoptosis in head and neck squamous cell carcinoma.","authors":"Sarah Sywanycz, Brianna L Hill, Zoey A Miller, Gavin Turner, Lily Huang, Kyle Polen, Robert J Lee, Ryan M Carey","doi":"10.1158/1535-7163.MCT-25-0358","DOIUrl":"10.1158/1535-7163.MCT-25-0358","url":null,"abstract":"<p><p>Bitter taste receptors (T2Rs), a family of G-protein coupled receptors, are emerging as potential therapeutic targets in head and neck squamous cell carcinoma (HNSCC). Phendione, a known T2R5 agonist, has not been previously investigated in HNSCC. Here, we show that phendione activates endogenously expressed T2R5 in HNSCC cells and ex vivo tumor samples, inducing sustained calcium responses, reducing cell viability, and promoting apoptosis through a T2R5-dependent mechanism. Analysis of The Cancer Genome Atlas data revealed that high T2R5 expression in HNSCC tumors correlates with improved long-term disease-specific survival, suggesting a potential tumor-suppressive role for T2R5. These findings highlight T2R5 as a promising therapeutic target in HNSCC and support further investigation of phendione or other T2R5 agonists as potential anti-cancer agents.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.5,"publicationDate":"2025-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145471548","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Molecular Cancer Therapeutics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1