首页 > 最新文献

Experimental Hematology & Oncology最新文献

英文 中文
Novel CAR T-cell therapies for relapsed/refractory B-cell malignancies: latest updates from 2023 ASH annual meeting 治疗复发/难治性B细胞恶性肿瘤的新型CAR T细胞疗法:2023年ASH年会的最新进展
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-04-18 DOI: 10.1186/s40164-024-00508-4
Wenjie Zhang, Sumei Li, Jinlan Long, Shufeng Xie, Minghui Wang, Han Liu, Zhenshu Xu
Chimeric antigen receptors (CAR) are engineered fusion proteins that target T-cells to specific surface antigens of tumor cells to generate effective anti-tumor responses. CAR T-cell therapy is playing an increasingly important role in the treatment of relapsed/refractory B-cell malignancies (R/R BCM). Attempting to make CAR T-cells safer and more effective in treating R/R BCM, various novel engineered CAR T-cell agents are currently in the research and development or clinical trial stages. We have summarized here the latest reports on the novel CAR T-cell therapies for R/R BCM presented at the 2023 ASH Annual Meeting as well as the latest updates in related clinical trials.
嵌合抗原受体(CAR)是一种经过设计的融合蛋白,能将 T 细胞靶向肿瘤细胞的特异性表面抗原,从而产生有效的抗肿瘤反应。CAR T细胞疗法在治疗复发/难治性B细胞恶性肿瘤(R/R BCM)中发挥着越来越重要的作用。为了使 CAR T 细胞在治疗复发性/难治性 B 细胞恶性肿瘤(R/R BCM)时更安全、更有效,各种新型工程 CAR T 细胞制剂目前正处于研发或临床试验阶段。我们在此总结了2023年ASH年会上关于治疗R/R BCM的新型CAR T细胞疗法的最新报告以及相关临床试验的最新进展。
{"title":"Novel CAR T-cell therapies for relapsed/refractory B-cell malignancies: latest updates from 2023 ASH annual meeting","authors":"Wenjie Zhang, Sumei Li, Jinlan Long, Shufeng Xie, Minghui Wang, Han Liu, Zhenshu Xu","doi":"10.1186/s40164-024-00508-4","DOIUrl":"https://doi.org/10.1186/s40164-024-00508-4","url":null,"abstract":"Chimeric antigen receptors (CAR) are engineered fusion proteins that target T-cells to specific surface antigens of tumor cells to generate effective anti-tumor responses. CAR T-cell therapy is playing an increasingly important role in the treatment of relapsed/refractory B-cell malignancies (R/R BCM). Attempting to make CAR T-cells safer and more effective in treating R/R BCM, various novel engineered CAR T-cell agents are currently in the research and development or clinical trial stages. We have summarized here the latest reports on the novel CAR T-cell therapies for R/R BCM presented at the 2023 ASH Annual Meeting as well as the latest updates in related clinical trials.","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-04-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140630342","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pan-cancer transcriptional atlas of minimal residual disease links DUSP1 to chemotherapy persistence 最小残留病的泛癌症转录图谱将 DUSP1 与化疗持久性联系起来
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-04-16 DOI: 10.1186/s40164-024-00509-3
Yuanhui Liu, Bi Peng, Ziqi Chen, Yimin Shen, Jingmin Zhang, Xianglin Yuan
Chemotherapy is a commonly effective treatment for most types of cancer. However, many patients experience a relapse due to minimal residual disease (MRD) after chemotherapy. Previous studies have analyzed the changes induced by chemotherapy for specific types of cancer, but our study is the first to comprehensively analyze MRD across various types of cancer. We included both bulk and single-cell RNA sequencing datasets. We compared the expression of the entire genome and calculated scores for canonical pathway signatures and immune infiltrates before and after chemotherapy across different types of cancer. Our findings revealed that DUSP1 was the most significantly and widely enriched gene in pan-cancer MRD. DUSP1 was found to be essential for MRD formation and played a role in T cell-fibroblast communications and the cytotoxic function of CD4 + T cells. Overall, our analysis provides a comprehensive understanding of the changes caused by chemotherapy and identifies potential targets for preventing and eliminating MRD, which could lead to long-term survival benefits for patients.
化疗是治疗大多数类型癌症的常用有效方法。然而,许多患者在化疗后会因极小残留病(MRD)而复发。以前的研究分析了特定类型癌症化疗引起的变化,但我们的研究是首次全面分析各种类型癌症的MRD。我们纳入了大量和单细胞 RNA 测序数据集。我们比较了全基因组的表达,并计算了不同类型癌症化疗前后卡农通路特征和免疫浸润的得分。我们的研究结果表明,DUSP1是泛癌症MRD中最显著、最广泛富集的基因。研究发现,DUSP1对MRD的形成至关重要,并在T细胞与成纤维细胞的沟通以及CD4 + T细胞的细胞毒性功能中发挥作用。总之,我们的分析提供了对化疗引起的变化的全面了解,并确定了预防和消除MRD的潜在靶点,这将为患者带来长期生存益处。
{"title":"Pan-cancer transcriptional atlas of minimal residual disease links DUSP1 to chemotherapy persistence","authors":"Yuanhui Liu, Bi Peng, Ziqi Chen, Yimin Shen, Jingmin Zhang, Xianglin Yuan","doi":"10.1186/s40164-024-00509-3","DOIUrl":"https://doi.org/10.1186/s40164-024-00509-3","url":null,"abstract":"Chemotherapy is a commonly effective treatment for most types of cancer. However, many patients experience a relapse due to minimal residual disease (MRD) after chemotherapy. Previous studies have analyzed the changes induced by chemotherapy for specific types of cancer, but our study is the first to comprehensively analyze MRD across various types of cancer. We included both bulk and single-cell RNA sequencing datasets. We compared the expression of the entire genome and calculated scores for canonical pathway signatures and immune infiltrates before and after chemotherapy across different types of cancer. Our findings revealed that DUSP1 was the most significantly and widely enriched gene in pan-cancer MRD. DUSP1 was found to be essential for MRD formation and played a role in T cell-fibroblast communications and the cytotoxic function of CD4 + T cells. Overall, our analysis provides a comprehensive understanding of the changes caused by chemotherapy and identifies potential targets for preventing and eliminating MRD, which could lead to long-term survival benefits for patients.","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-04-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140561488","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A Fab of trastuzumab to treat HER2 overexpressing breast cancer brain metastases 用曲妥珠单抗治疗 HER2 过度表达乳腺癌脑转移瘤的 Fab疗法
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-04-15 DOI: 10.1186/s40164-024-00513-7
Eurydice Angeli, Justine Paris, Olivier Le Tilly, Céline Desvignes, Guillaume Gapihan, Didier Boquet, Frédéric Pamoukdjian, Diaddin Hamdan, Marthe Rigal, Florence Poirier, Didier Lutomski, Feriel Azibani, Alexandre Mebazaa, Amaury Herbet, Aloïse Mabondzo, Géraldine Falgarone, Anne Janin, Gilles Paintaud, Guilhem Bousquet
Despite major therapeutic advances for two decades, including the most recently approved anti-HER2 drugs, brain metastatic localizations remain the major cause of death for women with metastatic HER2 breast cancer. The main reason is the limited drug passage of the blood-brain barrier after intravenous injection and the significant efflux of drugs, including monoclocal antibodies, after administration into the cerebrospinal fluid. We hypothesized that this efflux was linked to the presence of a FcRn receptor in the blood-brain barrier. To overcome this efflux, we engineered two Fab fragments of trastuzumab, an anti-HER2 monoclonal antibody, and did a thorough preclinical development for therapeutic translational purpose. We demonstrated the safety and equal efficacy of the Fabs with trastuzumab in vitro, and in vivo using a patient-derived xenograft model of HER2 overexpressing breast cancer. For the pharmacokinetic studies of intra-cerebrospinal fluid administration, we implemented original rat models with catheter implanted into the cisterna magna. After intraventricular administration in rats, we demonstrated that the brain-to-blood efflux of Fab was up to 10 times lower than for trastuzumab, associated with a two-fold higher brain penetration compared to trastuzumab. This Fab, capable of significantly reducing brain-to-blood efflux and enhancing brain penetration after intra-cerebrospinal fluid injection, could thus be a new and original effective drug in the treatment of HER2 breast cancer brain metastases, which will be demonstrated by a phase I clinical trial dedicated to women in resort situations.
尽管二十年来在治疗方面取得了重大进展,包括最近批准的抗 HER2 药物,但脑转移灶仍是转移性 HER2 乳腺癌女性患者的主要死因。主要原因是静脉注射后药物通过血脑屏障的能力有限,而且药物(包括单克隆抗体)在进入脑脊液后会大量外流。我们假设这种外流与血脑屏障中存在 FcRn 受体有关。为了克服这种外流现象,我们设计了两种抗 HER2 单克隆抗体曲妥珠单抗的 Fab 片段,并为治疗转化目的进行了全面的临床前开发。我们在体外和体内使用源自患者的 HER2 过度表达乳腺癌异种移植模型证明了 Fabs 与曲妥珠单抗的安全性和同等疗效。在脑脊液内给药的药代动力学研究中,我们采用了将导管植入大鼠脑室的原始大鼠模型。在大鼠脑室内给药后,我们证实法布从脑到血液的外流比曲妥珠单抗低 10 倍,脑穿透力比曲妥珠单抗高 2 倍。因此,这种能够显著降低脑脊液内注射后的脑-血外流并增强脑穿透力的 Fab 可以成为治疗 HER2 乳腺癌脑转移的一种新的原创有效药物。
{"title":"A Fab of trastuzumab to treat HER2 overexpressing breast cancer brain metastases","authors":"Eurydice Angeli, Justine Paris, Olivier Le Tilly, Céline Desvignes, Guillaume Gapihan, Didier Boquet, Frédéric Pamoukdjian, Diaddin Hamdan, Marthe Rigal, Florence Poirier, Didier Lutomski, Feriel Azibani, Alexandre Mebazaa, Amaury Herbet, Aloïse Mabondzo, Géraldine Falgarone, Anne Janin, Gilles Paintaud, Guilhem Bousquet","doi":"10.1186/s40164-024-00513-7","DOIUrl":"https://doi.org/10.1186/s40164-024-00513-7","url":null,"abstract":"Despite major therapeutic advances for two decades, including the most recently approved anti-HER2 drugs, brain metastatic localizations remain the major cause of death for women with metastatic HER2 breast cancer. The main reason is the limited drug passage of the blood-brain barrier after intravenous injection and the significant efflux of drugs, including monoclocal antibodies, after administration into the cerebrospinal fluid. We hypothesized that this efflux was linked to the presence of a FcRn receptor in the blood-brain barrier. To overcome this efflux, we engineered two Fab fragments of trastuzumab, an anti-HER2 monoclonal antibody, and did a thorough preclinical development for therapeutic translational purpose. We demonstrated the safety and equal efficacy of the Fabs with trastuzumab in vitro, and in vivo using a patient-derived xenograft model of HER2 overexpressing breast cancer. For the pharmacokinetic studies of intra-cerebrospinal fluid administration, we implemented original rat models with catheter implanted into the cisterna magna. After intraventricular administration in rats, we demonstrated that the brain-to-blood efflux of Fab was up to 10 times lower than for trastuzumab, associated with a two-fold higher brain penetration compared to trastuzumab. This Fab, capable of significantly reducing brain-to-blood efflux and enhancing brain penetration after intra-cerebrospinal fluid injection, could thus be a new and original effective drug in the treatment of HER2 breast cancer brain metastases, which will be demonstrated by a phase I clinical trial dedicated to women in resort situations. ","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-04-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140561477","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mechanistic insights and the clinical prospects of targeted therapies for glioblastoma: a comprehensive review 胶质母细胞瘤靶向疗法的机制认识和临床前景:综述
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-04-13 DOI: 10.1186/s40164-024-00512-8
Yating Shen, Dexter Kai Hao Thng, Andrea Li Ann Wong, Tan Boon Toh
Glioblastoma (GBM) is a fatal brain tumour that is traditionally diagnosed based on histological features. Recent molecular profiling studies have reshaped the World Health Organization approach in the classification of central nervous system tumours to include more pathogenetic hallmarks. These studies have revealed that multiple oncogenic pathways are dysregulated, which contributes to the aggressiveness and resistance of GBM. Such findings have shed light on the molecular vulnerability of GBM and have shifted the disease management paradigm from chemotherapy to targeted therapies. Targeted drugs have been developed to inhibit oncogenic targets in GBM, including receptors involved in the angiogenic axis, the signal transducer and activator of transcription 3 (STAT3), the PI3K/AKT/mTOR signalling pathway, the ubiquitination-proteasome pathway, as well as IDH1/2 pathway. While certain targeted drugs showed promising results in vivo, the translatability of such preclinical achievements in GBM remains a barrier. We also discuss the recent developments and clinical assessments of targeted drugs, as well as the prospects of cell-based therapies and combinatorial therapy as novel ways to target GBM. Targeted treatments have demonstrated preclinical efficacy over chemotherapy as an alternative or adjuvant to the current standard of care for GBM, but their clinical efficacy remains hindered by challenges such as blood-brain barrier penetrance of the drugs. The development of combinatorial targeted therapies is expected to improve therapeutic efficacy and overcome drug resistance.
胶质母细胞瘤(GBM)是一种致命的脑肿瘤,传统上根据组织学特征进行诊断。最近的分子图谱研究重塑了世界卫生组织对中枢神经系统肿瘤的分类方法,纳入了更多的致病特征。这些研究揭示了多种致癌通路失调,从而导致了 GBM 的侵袭性和耐药性。这些发现揭示了 GBM 的分子脆弱性,并使疾病治疗模式从化疗转向靶向治疗。目前已开发出抑制 GBM 致癌靶点的靶向药物,包括参与血管生成轴的受体、转录信号转导和激活因子 3 (STAT3)、PI3K/AKT/mTOR 信号通路、泛素化-蛋白酶体通路以及 IDH1/2 通路。虽然某些靶向药物在体内显示出良好的效果,但这些临床前研究成果在 GBM 中的转化仍然存在障碍。我们还讨论了靶向药物的最新进展和临床评估,以及细胞疗法和组合疗法作为靶向 GBM 的新方法的前景。靶向治疗在临床前已显示出优于化疗的疗效,可作为 GBM 现行标准疗法的替代或辅助疗法,但其临床疗效仍受到药物血脑屏障穿透性等挑战的阻碍。组合靶向疗法的开发有望提高疗效并克服耐药性。
{"title":"Mechanistic insights and the clinical prospects of targeted therapies for glioblastoma: a comprehensive review","authors":"Yating Shen, Dexter Kai Hao Thng, Andrea Li Ann Wong, Tan Boon Toh","doi":"10.1186/s40164-024-00512-8","DOIUrl":"https://doi.org/10.1186/s40164-024-00512-8","url":null,"abstract":"Glioblastoma (GBM) is a fatal brain tumour that is traditionally diagnosed based on histological features. Recent molecular profiling studies have reshaped the World Health Organization approach in the classification of central nervous system tumours to include more pathogenetic hallmarks. These studies have revealed that multiple oncogenic pathways are dysregulated, which contributes to the aggressiveness and resistance of GBM. Such findings have shed light on the molecular vulnerability of GBM and have shifted the disease management paradigm from chemotherapy to targeted therapies. Targeted drugs have been developed to inhibit oncogenic targets in GBM, including receptors involved in the angiogenic axis, the signal transducer and activator of transcription 3 (STAT3), the PI3K/AKT/mTOR signalling pathway, the ubiquitination-proteasome pathway, as well as IDH1/2 pathway. While certain targeted drugs showed promising results in vivo, the translatability of such preclinical achievements in GBM remains a barrier. We also discuss the recent developments and clinical assessments of targeted drugs, as well as the prospects of cell-based therapies and combinatorial therapy as novel ways to target GBM. Targeted treatments have demonstrated preclinical efficacy over chemotherapy as an alternative or adjuvant to the current standard of care for GBM, but their clinical efficacy remains hindered by challenges such as blood-brain barrier penetrance of the drugs. The development of combinatorial targeted therapies is expected to improve therapeutic efficacy and overcome drug resistance.","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-04-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140561652","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Myeloid-derived suppressor cells in cancer: therapeutic targets to overcome tumor immune evasion 癌症中的髓源性抑制细胞:克服肿瘤免疫逃避的治疗目标
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-04-12 DOI: 10.1186/s40164-024-00505-7
Junli Lu, Yiming Luo, Dean Rao, Tiantian Wang, Zhen Lei, Xiaoping Chen, Bixiang Zhang, Yiwei Li, Bifeng Liu, Limin Xia, Wenjie Huang
Paradoxically, tumor development and progression can be inhibited and promoted by the immune system. After three stages of immune editing, namely, elimination, homeostasis and escape, tumor cells are no longer restricted by immune surveillance and thus develop into clinical tumors. The mechanisms of immune escape include abnormalities in antitumor-associated immune cells, selection for immune resistance to tumor cells, impaired transport of T cells, and the formation of an immunosuppressive tumor microenvironment. A population of distinct immature myeloid cells, myeloid-derived suppressor cells (MDSCs), mediate immune escape primarily by exerting immunosuppressive effects and participating in the constitution of an immunosuppressive microtumor environment. Clinical trials have found that the levels of MDSCs in the peripheral blood of cancer patients are strongly correlated with tumor stage, metastasis and prognosis. Moreover, animal experiments have confirmed that elimination of MDSCs inhibits tumor growth and metastasis to some extent. Therefore, MDSCs may become the target of immunotherapy for many cancers, and eliminating MDSCs can help improve the response rate to cancer treatment and patient survival. However, a clear definition of MDSCs and the specific mechanism involved in immune escape are lacking. In this paper, we review the role of the MDSCs population in tumor development and the mechanisms involved in immune escape in different tumor contexts. In addition, we discuss the use of these cells as targets for tumor immunotherapy. This review not only contributes to a systematic and comprehensive understanding of the essential role of MDSCs in immune system reactions against tumors but also provides information to guide the development of cancer therapies targeting MDSCs.
矛盾的是,免疫系统既可以抑制肿瘤的发展,也可以促进肿瘤的发展。经过消除、平衡和逃逸三个阶段的免疫编辑后,肿瘤细胞不再受免疫监视的限制,从而发展成为临床肿瘤。免疫逃逸的机制包括抗肿瘤相关免疫细胞的异常、对肿瘤细胞免疫抵抗的选择、T 细胞转运受损以及免疫抑制性肿瘤微环境的形成。一种独特的未成熟髓系细胞--髓源性抑制细胞(MDSCs)主要通过发挥免疫抑制作用和参与形成免疫抑制性微肿瘤环境来介导免疫逃逸。临床试验发现,癌症患者外周血中的 MDSCs 水平与肿瘤分期、转移和预后密切相关。此外,动物实验证实,消除 MDSCs 可在一定程度上抑制肿瘤的生长和转移。因此,MDSCs 有可能成为多种癌症的免疫治疗靶点,消除 MDSCs 有助于提高癌症治疗反应率和患者生存率。然而,目前尚缺乏对MDSCs的明确定义以及参与免疫逃逸的具体机制。在本文中,我们回顾了 MDSCs 群体在肿瘤发生发展中的作用以及在不同肿瘤环境中参与免疫逃逸的机制。此外,我们还讨论了如何利用这些细胞作为肿瘤免疫疗法的靶点。这篇综述不仅有助于系统全面地了解 MDSCs 在免疫系统对抗肿瘤反应中的重要作用,还为开发针对 MDSCs 的癌症疗法提供了指导信息。
{"title":"Myeloid-derived suppressor cells in cancer: therapeutic targets to overcome tumor immune evasion","authors":"Junli Lu, Yiming Luo, Dean Rao, Tiantian Wang, Zhen Lei, Xiaoping Chen, Bixiang Zhang, Yiwei Li, Bifeng Liu, Limin Xia, Wenjie Huang","doi":"10.1186/s40164-024-00505-7","DOIUrl":"https://doi.org/10.1186/s40164-024-00505-7","url":null,"abstract":"Paradoxically, tumor development and progression can be inhibited and promoted by the immune system. After three stages of immune editing, namely, elimination, homeostasis and escape, tumor cells are no longer restricted by immune surveillance and thus develop into clinical tumors. The mechanisms of immune escape include abnormalities in antitumor-associated immune cells, selection for immune resistance to tumor cells, impaired transport of T cells, and the formation of an immunosuppressive tumor microenvironment. A population of distinct immature myeloid cells, myeloid-derived suppressor cells (MDSCs), mediate immune escape primarily by exerting immunosuppressive effects and participating in the constitution of an immunosuppressive microtumor environment. Clinical trials have found that the levels of MDSCs in the peripheral blood of cancer patients are strongly correlated with tumor stage, metastasis and prognosis. Moreover, animal experiments have confirmed that elimination of MDSCs inhibits tumor growth and metastasis to some extent. Therefore, MDSCs may become the target of immunotherapy for many cancers, and eliminating MDSCs can help improve the response rate to cancer treatment and patient survival. However, a clear definition of MDSCs and the specific mechanism involved in immune escape are lacking. In this paper, we review the role of the MDSCs population in tumor development and the mechanisms involved in immune escape in different tumor contexts. In addition, we discuss the use of these cells as targets for tumor immunotherapy. This review not only contributes to a systematic and comprehensive understanding of the essential role of MDSCs in immune system reactions against tumors but also provides information to guide the development of cancer therapies targeting MDSCs.","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-04-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140561493","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
TRIB3 silencing promotes the downregulation of Akt pathway and PAX3-FOXO1 in high-risk rhabdomyosarcoma TRIB3沉默可促进高危横纹肌肉瘤中Akt通路和PAX3-FOXO1的下调
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-04-05 DOI: 10.1186/s40164-024-00503-9
Gabriel Gallo-Oller, Guillem Pons, Júlia Sansa-Girona, Natalia Navarro, Patricia Zarzosa, Lia García-Gilabert, Paula Cabré-Fernandez, Gabriela Guillén Burrieza, Lorena Valero-Arrese, Miguel F. Segura, José M. Lizcano, José Sánchez de Toledo, Lucas Moreno, Soledad Gallego, Josep Roma
Rhabdomyosarcoma (RMS), such as other childhood tumors, has witnessed treatment advancements in recent years. However, high-risk patients continue to face poor survival rates, often attributed to the presence of the PAX3/7-FOXO1 fusion proteins, which has been associated with metastasis and treatment resistance. Despite efforts to directly target these chimeric proteins, clinical success remains elusive. In this study, the main aim was to address this challenge by investigating regulators of FOXO1. Specifically, we focused on TRIB3, a potential regulator of the fusion protein in RMS. Our findings revealed a prominent TRIB3 expression in RMS tumors, highlighting its correlation with the presence of fusion protein. By conducting TRIB3 genetic inhibition experiments, we observed an impairment on cell proliferation. Notably, the knockdown of TRIB3 led to a decrease in PAX3-FOXO1 and its target genes at protein level, accompanied by a reduction in the activity of the Akt signaling pathway. Additionally, inducible silencing of TRIB3 significantly delayed tumor growth and improved overall survival in vivo. Based on our analysis, we propose that TRIB3 holds therapeutic potential for treating the most aggressive subtype of RMS. The findings herein reported contribute to our understanding of the underlying molecular mechanisms driving RMS progression and provide novel insights into the potential use of TRIB3 as a therapeutic intervention for high-risk RMS patients.
与其他儿童肿瘤一样,横纹肌肉瘤(RMS)的治疗近年来也取得了进展。然而,高危患者的生存率仍然很低,这通常归因于PAX3/7-FOXO1融合蛋白的存在,它与转移和耐药性有关。尽管人们一直在努力直接靶向这些嵌合蛋白,但临床成功仍然遥遥无期。本研究的主要目的是通过研究 FOXO1 的调控因子来应对这一挑战。具体来说,我们重点研究了 TRIB3,它是 RMS 中融合蛋白的潜在调控因子。我们的研究结果表明,TRIB3在RMS肿瘤中的表达非常突出,这凸显了它与融合蛋白存在的相关性。通过TRIB3基因抑制实验,我们观察到细胞增殖受到了影响。值得注意的是,TRIB3的基因敲除会导致PAX3-FOXO1及其靶基因蛋白水平的下降,并伴随着Akt信号通路活性的降低。此外,诱导性沉默TRIB3可明显延缓肿瘤生长并提高体内总生存率。根据我们的分析,我们认为 TRIB3 具有治疗最具侵袭性亚型 RMS 的潜力。本文报告的研究结果有助于我们了解驱动 RMS 进展的潜在分子机制,并为 TRIB3 作为高风险 RMS 患者治疗干预措施的潜在用途提供了新的见解。
{"title":"TRIB3 silencing promotes the downregulation of Akt pathway and PAX3-FOXO1 in high-risk rhabdomyosarcoma","authors":"Gabriel Gallo-Oller, Guillem Pons, Júlia Sansa-Girona, Natalia Navarro, Patricia Zarzosa, Lia García-Gilabert, Paula Cabré-Fernandez, Gabriela Guillén Burrieza, Lorena Valero-Arrese, Miguel F. Segura, José M. Lizcano, José Sánchez de Toledo, Lucas Moreno, Soledad Gallego, Josep Roma","doi":"10.1186/s40164-024-00503-9","DOIUrl":"https://doi.org/10.1186/s40164-024-00503-9","url":null,"abstract":"Rhabdomyosarcoma (RMS), such as other childhood tumors, has witnessed treatment advancements in recent years. However, high-risk patients continue to face poor survival rates, often attributed to the presence of the PAX3/7-FOXO1 fusion proteins, which has been associated with metastasis and treatment resistance. Despite efforts to directly target these chimeric proteins, clinical success remains elusive. In this study, the main aim was to address this challenge by investigating regulators of FOXO1. Specifically, we focused on TRIB3, a potential regulator of the fusion protein in RMS. Our findings revealed a prominent TRIB3 expression in RMS tumors, highlighting its correlation with the presence of fusion protein. By conducting TRIB3 genetic inhibition experiments, we observed an impairment on cell proliferation. Notably, the knockdown of TRIB3 led to a decrease in PAX3-FOXO1 and its target genes at protein level, accompanied by a reduction in the activity of the Akt signaling pathway. Additionally, inducible silencing of TRIB3 significantly delayed tumor growth and improved overall survival in vivo. Based on our analysis, we propose that TRIB3 holds therapeutic potential for treating the most aggressive subtype of RMS. The findings herein reported contribute to our understanding of the underlying molecular mechanisms driving RMS progression and provide novel insights into the potential use of TRIB3 as a therapeutic intervention for high-risk RMS patients.","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-04-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140561369","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Novel insights into TCR-T cell therapy in solid neoplasms: optimizing adoptive immunotherapy TCR-T细胞疗法治疗实体瘤的新见解:优化采用性免疫疗法
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-04-03 DOI: 10.1186/s40164-024-00504-8
Weihuan Shao, Yiran Yao, Ludi Yang, Xiaoran Li, Tongxin Ge, Yue Zheng, Qiuyi Zhu, Shengfang Ge, Xiang Gu, Renbing Jia, Xin Song, Ai Zhuang
Adoptive immunotherapy in the T cell landscape exhibits efficacy in cancer treatment. Over the past few decades, genetically modified T cells, particularly chimeric antigen receptor T cells, have enabled remarkable strides in the treatment of hematological malignancies. Besides, extensive exploration of multiple antigens for the treatment of solid tumors has led to clinical interest in the potential of T cells expressing the engineered T cell receptor (TCR). TCR-T cells possess the capacity to recognize intracellular antigen families and maintain the intrinsic properties of TCRs in terms of affinity to target epitopes and signal transduction. Recent research has provided critical insight into their capability and therapeutic targets for multiple refractory solid tumors, but also exposes some challenges for durable efficacy. In this review, we describe the screening and identification of available tumor antigens, and the acquisition and optimization of TCRs for TCR-T cell therapy. Furthermore, we summarize the complete flow from laboratory to clinical applications of TCR-T cells. Last, we emerge future prospects for improving therapeutic efficacy in cancer world with combination therapies or TCR-T derived products. In conclusion, this review depicts our current understanding of TCR-T cell therapy in solid neoplasms, and provides new perspectives for expanding its clinical applications and improving therapeutic efficacy.
T 细胞的适应性免疫疗法在癌症治疗中显示出疗效。过去几十年来,转基因 T 细胞,特别是嵌合抗原受体 T 细胞在治疗血液恶性肿瘤方面取得了显著进展。此外,对治疗实体瘤的多种抗原的广泛探索,也使临床对表达工程化 T 细胞受体(TCR)的 T 细胞的潜力产生了兴趣。TCR-T 细胞具有识别细胞内抗原家族的能力,并在与目标表位的亲和力和信号转导方面保持 TCR 的固有特性。最近的研究对TCR-T细胞的能力和多种难治性实体瘤的治疗靶点提出了重要的见解,但也暴露出了持久疗效方面的一些挑战。在这篇综述中,我们介绍了现有肿瘤抗原的筛选和鉴定,以及用于 TCR-T 细胞疗法的 TCR 的获取和优化。此外,我们还总结了 TCR-T 细胞从实验室到临床应用的完整流程。最后,我们展望了利用联合疗法或 TCR-T 衍生产品提高癌症疗效的未来前景。总之,这篇综述描述了我们目前对实体瘤 TCR-T 细胞疗法的理解,并为扩大其临床应用和提高疗效提供了新的视角。
{"title":"Novel insights into TCR-T cell therapy in solid neoplasms: optimizing adoptive immunotherapy","authors":"Weihuan Shao, Yiran Yao, Ludi Yang, Xiaoran Li, Tongxin Ge, Yue Zheng, Qiuyi Zhu, Shengfang Ge, Xiang Gu, Renbing Jia, Xin Song, Ai Zhuang","doi":"10.1186/s40164-024-00504-8","DOIUrl":"https://doi.org/10.1186/s40164-024-00504-8","url":null,"abstract":"Adoptive immunotherapy in the T cell landscape exhibits efficacy in cancer treatment. Over the past few decades, genetically modified T cells, particularly chimeric antigen receptor T cells, have enabled remarkable strides in the treatment of hematological malignancies. Besides, extensive exploration of multiple antigens for the treatment of solid tumors has led to clinical interest in the potential of T cells expressing the engineered T cell receptor (TCR). TCR-T cells possess the capacity to recognize intracellular antigen families and maintain the intrinsic properties of TCRs in terms of affinity to target epitopes and signal transduction. Recent research has provided critical insight into their capability and therapeutic targets for multiple refractory solid tumors, but also exposes some challenges for durable efficacy. In this review, we describe the screening and identification of available tumor antigens, and the acquisition and optimization of TCRs for TCR-T cell therapy. Furthermore, we summarize the complete flow from laboratory to clinical applications of TCR-T cells. Last, we emerge future prospects for improving therapeutic efficacy in cancer world with combination therapies or TCR-T derived products. In conclusion, this review depicts our current understanding of TCR-T cell therapy in solid neoplasms, and provides new perspectives for expanding its clinical applications and improving therapeutic efficacy.","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140561474","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
VISTA drives macrophages towards a pro-tumoral phenotype that promotes cancer cell phagocytosis yet down-regulates T cell responses VISTA 促使巨噬细胞向有利于肿瘤的表型发展,从而促进癌细胞的吞噬作用,同时下调 T 细胞反应
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-03-29 DOI: 10.1186/s40164-024-00501-x
Yusheng Lin, Ghizlane Choukrani, Lena Dubbel, Lena Rockstein, Jimena Alvarez Freile, Yuzhu Qi, Valerie Wiersma, Hao Zhang, Karl-Wilhelm Koch, Emanuele Ammatuna, Jan Jacob Schuringa, Tom van Meerten, Gerwin Huls, Edwin Bremer
VISTA is a well-known immune checkpoint in T cell biology, but its role in innate immunity is less established. Here, we investigated the role of VISTA on anticancer macrophage immunity, with a focus on phagocytosis, macrophage polarization and concomitant T cell activation. Macrophages, differentiated from VISTA overexpressed THP-1 cells and cord blood CD34+ cell-derived monocytes, were used in phagocytosis assay using B lymphoma target cells opsonized with Rituximab. PBMC-derived macrophages were used to assess the correlation between phagocytosis and VISTA expression. qRT-PCR, flow cytometry, and enzyme-linked immunosorbent assay were performed to analyze the impact of VISTA on other checkpoints and M1/M2-like macrophage biology. Additionally, flow cytometry was used to assess the frequency of CD14+ monocytes expressing VISTA in PBMCs from 65 lymphoma patients and 37 healthy donors. Ectopic expression of VISTA in the monocytic model cell line THP-1 or in primary monocytes triggered differentiation towards the macrophage lineage, with a marked increase in M2-like macrophage-related gene expression and decrease in M1-like macrophage-related gene expression. VISTA expression in THP-1 and monocyte-derived macrophages strongly downregulated expression of SIRPα, a prominent ‘don’t eat me’ signal, and augmented phagocytic activity of macrophages against cancer cells. Intriguingly, expression of VISTA’s extracellular domain alone sufficed to trigger phagocytosis in ∼ 50% of cell lines, with those cell lines also directly binding to recombinant human VISTA, indicating ligand-dependent and -independent mechanisms. Endogenous VISTA expression was predominantly higher in M2-like macrophages compared to M0- or M1-like macrophages, with a positive correlation observed between VISTA expression in M2c macrophages and their phagocytic activity. VISTA-expressing macrophages demonstrated a unique cytokine profile, characterized by reduced IL-1β and elevated IL-10 secretion. Furthermore, VISTA interacted with MHC-I and downregulated its surface expression, leading to diminished T cell activation. Notably, VISTA surface expression was identified in monocytes from all lymphoma patients but was less prevalent in healthy donors. Collectively, VISTA expression associates with and drives M2-like activation of macrophages with a high phagocytic capacity yet a decrease in antigen presentation capability to T cells. Therefore, VISTA is a negative immune checkpoint regulator in macrophage-mediated immune suppression.
VISTA 是 T 细胞生物学中一个众所周知的免疫检查点,但它在先天免疫中的作用却鲜为人知。在这里,我们研究了 VISTA 对抗癌巨噬细胞免疫的作用,重点是吞噬、巨噬细胞极化和伴随的 T 细胞活化。由 VISTA 过表达的 THP-1 细胞和脐带血 CD34+ 细胞衍生的单核细胞分化出的巨噬细胞被用于利妥昔单抗鸦片化的 B 淋巴瘤靶细胞的吞噬试验。采用 qRT-PCR、流式细胞术和酶联免疫吸附试验分析 VISTA 对其他检查点和 M1/M2 样巨噬细胞生物学的影响。此外,还使用流式细胞术评估了 65 名淋巴瘤患者和 37 名健康供体的 PBMC 中表达 VISTA 的 CD14+ 单核细胞的频率。在单核细胞模型细胞系 THP-1 或原代单核细胞中异位表达 VISTA 会引发向巨噬细胞系的分化,M2 样巨噬细胞相关基因表达明显增加,M1 样巨噬细胞相关基因表达减少。VISTA 在 THP-1 和单核细胞衍生巨噬细胞中的表达强烈下调了 SIRPα 的表达,这是一个显著的 "别吃我 "信号,并增强了巨噬细胞对癌细胞的吞噬活性。耐人寻味的是,仅表达VISTA的胞外结构域就足以触发50%细胞系的吞噬作用,这些细胞系还能直接与重组人VISTA结合,这表明了配体依赖性和非依赖性机制。与 M0 或 M1 样巨噬细胞相比,M2 样巨噬细胞中内源性 VISTA 的表达主要较高,在 M2c 巨噬细胞中观察到 VISTA 的表达与其吞噬活性呈正相关。表达 VISTA 的巨噬细胞表现出独特的细胞因子谱,其特点是 IL-1β 分泌减少,IL-10 分泌增加。此外,VISTA 与 MHC-I 相互作用并下调其表面表达,从而导致 T 细胞活化减弱。值得注意的是,在所有淋巴瘤患者的单核细胞中都发现了 VISTA 的表面表达,但在健康供体中却不常见。总之,VISTA 的表达与巨噬细胞的 M2 类活化有关,并驱动巨噬细胞的 M2 类活化,巨噬细胞的吞噬能力很强,但向 T 细胞呈递抗原的能力却下降了。因此,在巨噬细胞介导的免疫抑制中,VISTA 是一种负性免疫检查点调节因子。
{"title":"VISTA drives macrophages towards a pro-tumoral phenotype that promotes cancer cell phagocytosis yet down-regulates T cell responses","authors":"Yusheng Lin, Ghizlane Choukrani, Lena Dubbel, Lena Rockstein, Jimena Alvarez Freile, Yuzhu Qi, Valerie Wiersma, Hao Zhang, Karl-Wilhelm Koch, Emanuele Ammatuna, Jan Jacob Schuringa, Tom van Meerten, Gerwin Huls, Edwin Bremer","doi":"10.1186/s40164-024-00501-x","DOIUrl":"https://doi.org/10.1186/s40164-024-00501-x","url":null,"abstract":"VISTA is a well-known immune checkpoint in T cell biology, but its role in innate immunity is less established. Here, we investigated the role of VISTA on anticancer macrophage immunity, with a focus on phagocytosis, macrophage polarization and concomitant T cell activation. Macrophages, differentiated from VISTA overexpressed THP-1 cells and cord blood CD34+ cell-derived monocytes, were used in phagocytosis assay using B lymphoma target cells opsonized with Rituximab. PBMC-derived macrophages were used to assess the correlation between phagocytosis and VISTA expression. qRT-PCR, flow cytometry, and enzyme-linked immunosorbent assay were performed to analyze the impact of VISTA on other checkpoints and M1/M2-like macrophage biology. Additionally, flow cytometry was used to assess the frequency of CD14+ monocytes expressing VISTA in PBMCs from 65 lymphoma patients and 37 healthy donors. Ectopic expression of VISTA in the monocytic model cell line THP-1 or in primary monocytes triggered differentiation towards the macrophage lineage, with a marked increase in M2-like macrophage-related gene expression and decrease in M1-like macrophage-related gene expression. VISTA expression in THP-1 and monocyte-derived macrophages strongly downregulated expression of SIRPα, a prominent ‘don’t eat me’ signal, and augmented phagocytic activity of macrophages against cancer cells. Intriguingly, expression of VISTA’s extracellular domain alone sufficed to trigger phagocytosis in ∼ 50% of cell lines, with those cell lines also directly binding to recombinant human VISTA, indicating ligand-dependent and -independent mechanisms. Endogenous VISTA expression was predominantly higher in M2-like macrophages compared to M0- or M1-like macrophages, with a positive correlation observed between VISTA expression in M2c macrophages and their phagocytic activity. VISTA-expressing macrophages demonstrated a unique cytokine profile, characterized by reduced IL-1β and elevated IL-10 secretion. Furthermore, VISTA interacted with MHC-I and downregulated its surface expression, leading to diminished T cell activation. Notably, VISTA surface expression was identified in monocytes from all lymphoma patients but was less prevalent in healthy donors. Collectively, VISTA expression associates with and drives M2-like activation of macrophages with a high phagocytic capacity yet a decrease in antigen presentation capability to T cells. Therefore, VISTA is a negative immune checkpoint regulator in macrophage-mediated immune suppression.","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-03-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140323889","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Vaccination with a combination of STING agonist-loaded lipid nanoparticles and CpG-ODNs protects against lung metastasis via the induction of CD11bhighCD27low memory-like NK cells 通过诱导 CD11bhighCD27low 记忆样 NK 细胞,STING 激动剂负载脂质纳米颗粒和 CpG-ODNs 组合疫苗可防止肺转移
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-03-29 DOI: 10.1186/s40164-024-00502-w
Alaa M. Khalifa, Takashi Nakamura, Yusuke Sato, Hideyoshi Harashima
Natural killer (NK) cells are effective in attacking tumor cells that escape T cell attack. Memory NK cells are believed to function as potent effector cells in cancer immunotherapy. However, knowledge of their induction, identification, and potential in vivo is limited. Herein, we report on the induction and identification of memory-like NK cells via the action of a combination of a stimulator of interferon genes (STING) agonist loaded into lipid nanoparticles (STING-LNPs) and cytosine-phosphorothioate-guanine oligodeoxynucleotides (CpG-ODNs), and the potential of the inducted memory-like NK cells to prevent melanoma lung metastasis. The antitumor effects of either the STING-LNPs, CpG-ODNs, or the combination therapy were evaluated using a B16-F10 lung metastasis model. The effect of the combined treatment was evaluated by measuring cytokine production. The induction of memory-like NK cells was demonstrated via flow cytometry and confirmed through their preventative effect. The combination of STING-LNPs and CpG-ODNs tended to enhance the production of interleukin 12 (IL-12) and IL-18, and exerted a therapeutic effect against B16-F10 lung metastasis. The combination therapy increased the population of CD11bhighCD27low NK cells. Although monotherapies failed to show preventative effects, the combination therapy induced a surprisingly strong preventative effect, which indicates that CD11bhighCD27low cells could be a phenotype of memory-like NK cells. As far as could be ascertained, this is the first report of the in vivo induction, identification, and confirmation of a phenotype of the memory-like NK cells through a prophylactic effect via the use of an immunotherapeutic drug. Our findings provide novel insights into the in vivo induction of CD11bhighCD27low memory-like NK cells thus paving the way for the development of efficient immunotherapies.
自然杀伤(NK)细胞能有效攻击躲过 T 细胞攻击的肿瘤细胞。记忆 NK 细胞被认为是癌症免疫疗法中有效的效应细胞。然而,人们对记忆 NK 细胞的诱导、鉴定和体内潜能了解有限。在此,我们报告了通过将干扰素基因刺激剂(STING)激动剂装入脂质纳米颗粒(STING-LNPs)和胞嘧啶-硫代磷酸鸟嘌呤寡脱氧核苷酸(CpG-ODNs)的组合作用诱导和识别记忆样NK细胞的情况,以及诱导的记忆样NK细胞预防黑色素瘤肺转移的潜力。利用 B16-F10 肺转移模型评估了 STING-LNPs、CpG-ODNs 或联合疗法的抗肿瘤效果。联合疗法的效果通过测量细胞因子的产生来评估。通过流式细胞术证明了记忆样 NK 细胞的诱导作用,并通过其预防效果予以证实。STING-LNPs 和 CpG-ODNs 的联合治疗往往能提高白细胞介素 12(IL-12)和 IL-18 的产生,并对 B16-F10 肺转移产生治疗效果。联合疗法增加了 CD11bhighCD27low NK 细胞的数量。虽然单一疗法未能显示出预防效果,但联合疗法却产生了令人惊讶的强大预防效果,这表明 CD11bhighCD27low 细胞可能是记忆类 NK 细胞的一种表型。据目前所知,这是首次报道通过使用免疫治疗药物的预防作用,在体内诱导、鉴定和确认记忆样 NK 细胞的表型。我们的发现为体内诱导 CD11bhighCD27low 记忆样 NK 细胞提供了新的见解,从而为开发高效的免疫疗法铺平了道路。
{"title":"Vaccination with a combination of STING agonist-loaded lipid nanoparticles and CpG-ODNs protects against lung metastasis via the induction of CD11bhighCD27low memory-like NK cells","authors":"Alaa M. Khalifa, Takashi Nakamura, Yusuke Sato, Hideyoshi Harashima","doi":"10.1186/s40164-024-00502-w","DOIUrl":"https://doi.org/10.1186/s40164-024-00502-w","url":null,"abstract":"Natural killer (NK) cells are effective in attacking tumor cells that escape T cell attack. Memory NK cells are believed to function as potent effector cells in cancer immunotherapy. However, knowledge of their induction, identification, and potential in vivo is limited. Herein, we report on the induction and identification of memory-like NK cells via the action of a combination of a stimulator of interferon genes (STING) agonist loaded into lipid nanoparticles (STING-LNPs) and cytosine-phosphorothioate-guanine oligodeoxynucleotides (CpG-ODNs), and the potential of the inducted memory-like NK cells to prevent melanoma lung metastasis. The antitumor effects of either the STING-LNPs, CpG-ODNs, or the combination therapy were evaluated using a B16-F10 lung metastasis model. The effect of the combined treatment was evaluated by measuring cytokine production. The induction of memory-like NK cells was demonstrated via flow cytometry and confirmed through their preventative effect. The combination of STING-LNPs and CpG-ODNs tended to enhance the production of interleukin 12 (IL-12) and IL-18, and exerted a therapeutic effect against B16-F10 lung metastasis. The combination therapy increased the population of CD11bhighCD27low NK cells. Although monotherapies failed to show preventative effects, the combination therapy induced a surprisingly strong preventative effect, which indicates that CD11bhighCD27low cells could be a phenotype of memory-like NK cells. As far as could be ascertained, this is the first report of the in vivo induction, identification, and confirmation of a phenotype of the memory-like NK cells through a prophylactic effect via the use of an immunotherapeutic drug. Our findings provide novel insights into the in vivo induction of CD11bhighCD27low memory-like NK cells thus paving the way for the development of efficient immunotherapies. ","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-03-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140323815","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cyclin dependent kinase 4/6 inhibitor palbociclib synergizes with BCL2 inhibitor venetoclax in experimental models of mantle cell lymphoma without RB1 deletion. 在无RB1缺失的套细胞淋巴瘤实验模型中,细胞周期蛋白依赖性激酶4/6抑制剂palbociclib与BCL2抑制剂venetoclax具有协同作用。
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-03-25 DOI: 10.1186/s40164-024-00499-2
Diana Malarikova, Radek Jorda, Kristyna Kupcova, Jana Senavova, Alexandra Dolnikova, Eva Pokorna, Dmitry Kazantsev, Kristina Nozickova, Dana Sovilj, Celine Bellanger, David Chiron, Ladislav Andera, Vladimir Krystof, Miroslav Strnad, Karel Helman, Magdalena Klanova, Marek Trneny, Ondrej Havranek, Pavel Klener

Background: Mantle cell lymphoma (MCL) is a chronically relapsing malignancy with deregulated cell cycle progression. We analyzed efficacy, mode of action, and predictive markers of susceptibility to palbociclib, an approved CDK 4/6 inhibitor, and its combination with venetoclax, a BCL2 inhibitor.

Methods: A panel of nine MCL cell lines were used for in vitro experiments. Four patient derived xenografts (PDX) obtained from patients with chemotherapy and ibrutinib-refractory MCL were used for in vivo proof-of-concept studies. Changes of the mitochondrial membrane potential, energy-metabolic pathways, AKT activity, and pro-apoptotic priming of MCL cells were evaluated by JC-1 staining, Seahorse XF analyser, genetically encoded fluorescent AKT reporter, and BH3 profiling, respectively. MCL clones with gene knockout or transgenic (over)expression of CDKN2A, MYC, CDK4, and RB1 were used to estimate impact of these aberrations on sensitivity to palbociclib, and venetoclax.

Results: Co-targeting MCL cells with palbociclib and venetoclax induced cytotoxic synergy in vitro and in vivo. Molecular mechanisms responsible for the observed synthetic lethality comprised palbociclib-mediated downregulation of anti-apoptotic MCL1, increased levels of proapoptotic BIM bound on both BCL2, and BCL-XL and increased pro-apoptotic priming of MCL cells mediated by BCL2-independent mechanisms, predominantly palbociclib-triggered metabolic and mitochondrial stress. Loss of RB1 resulted in palbociclib resistance, while deletion of CDKN2A or overexpression of CDK4, and MYC genes did not change sensitivity to palbociclib.

Conclusions: Our data strongly support investigation of the chemotherapy-free palbociclib and venetoclax combination as an innovative treatment strategy for post-ibrutinib MCL patients without RB1 deletion.

背景:套细胞淋巴瘤(MCL套细胞淋巴瘤(MCL)是一种细胞周期进展失调的慢性复发性恶性肿瘤。我们分析了已获批准的CDK 4/6抑制剂palbociclib及其与BCL2抑制剂venetoclax联用的疗效、作用方式和易感性预测指标:方法:体外实验使用了九种 MCL 细胞系。从化疗和伊布替尼难治性MCL患者身上获得的四个患者衍生异种移植(PDX)被用于体内概念验证研究。通过JC-1染色、Seahorse XF分析仪、基因编码荧光AKT报告物和BH3分析,分别评估了MCL细胞线粒体膜电位、能量代谢途径、AKT活性和促凋亡引物的变化。使用基因敲除或转基因(过度)表达CDKN2A、MYC、CDK4和RB1的MCL克隆来评估这些畸变对palbociclib和venetoclax敏感性的影响:结果:用palbociclib和venetoclax共同靶向MCL细胞可在体外和体内产生细胞毒性协同作用。导致所观察到的合成致死的分子机制包括:palbociclib介导的抗凋亡MCL1下调、结合在BCL2和BCL-XL上的促凋亡BIM水平升高,以及由BCL2依赖机制介导的MCL细胞促凋亡引物增加,主要是palbociclib触发的代谢和线粒体应激。RB1缺失会导致palbociclib耐药,而CDKN2A基因缺失或CDK4和MYC基因过表达不会改变对palbociclib的敏感性:我们的数据有力地支持了将无化疗的palbociclib和venetoclax联合疗法作为一种创新的治疗策略,用于治疗无RB1缺失的伊布替尼后MCL患者。
{"title":"Cyclin dependent kinase 4/6 inhibitor palbociclib synergizes with BCL2 inhibitor venetoclax in experimental models of mantle cell lymphoma without RB1 deletion.","authors":"Diana Malarikova, Radek Jorda, Kristyna Kupcova, Jana Senavova, Alexandra Dolnikova, Eva Pokorna, Dmitry Kazantsev, Kristina Nozickova, Dana Sovilj, Celine Bellanger, David Chiron, Ladislav Andera, Vladimir Krystof, Miroslav Strnad, Karel Helman, Magdalena Klanova, Marek Trneny, Ondrej Havranek, Pavel Klener","doi":"10.1186/s40164-024-00499-2","DOIUrl":"10.1186/s40164-024-00499-2","url":null,"abstract":"<p><strong>Background: </strong>Mantle cell lymphoma (MCL) is a chronically relapsing malignancy with deregulated cell cycle progression. We analyzed efficacy, mode of action, and predictive markers of susceptibility to palbociclib, an approved CDK 4/6 inhibitor, and its combination with venetoclax, a BCL2 inhibitor.</p><p><strong>Methods: </strong>A panel of nine MCL cell lines were used for in vitro experiments. Four patient derived xenografts (PDX) obtained from patients with chemotherapy and ibrutinib-refractory MCL were used for in vivo proof-of-concept studies. Changes of the mitochondrial membrane potential, energy-metabolic pathways, AKT activity, and pro-apoptotic priming of MCL cells were evaluated by JC-1 staining, Seahorse XF analyser, genetically encoded fluorescent AKT reporter, and BH3 profiling, respectively. MCL clones with gene knockout or transgenic (over)expression of CDKN2A, MYC, CDK4, and RB1 were used to estimate impact of these aberrations on sensitivity to palbociclib, and venetoclax.</p><p><strong>Results: </strong>Co-targeting MCL cells with palbociclib and venetoclax induced cytotoxic synergy in vitro and in vivo. Molecular mechanisms responsible for the observed synthetic lethality comprised palbociclib-mediated downregulation of anti-apoptotic MCL1, increased levels of proapoptotic BIM bound on both BCL2, and BCL-XL and increased pro-apoptotic priming of MCL cells mediated by BCL2-independent mechanisms, predominantly palbociclib-triggered metabolic and mitochondrial stress. Loss of RB1 resulted in palbociclib resistance, while deletion of CDKN2A or overexpression of CDK4, and MYC genes did not change sensitivity to palbociclib.</p><p><strong>Conclusions: </strong>Our data strongly support investigation of the chemotherapy-free palbociclib and venetoclax combination as an innovative treatment strategy for post-ibrutinib MCL patients without RB1 deletion.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-03-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10962182/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140287261","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Experimental Hematology & Oncology
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