Immunogenic Cell Death

免疫原性细胞死亡
  • 文章类型: Journal Article
    在对抗癌症的斗争中,寻求靶向治疗至关重要。RAS/MAP激酶途径经常与肿瘤有关,ERK在RAS信号级联中作为最远端的激酶发挥关键作用。我们先前的研究表明,ERK和MYD88之间的相互作用,MYD88是先天免疫中的一种衔接蛋白。对于RAS依赖性转化和癌细胞存活至关重要。在这项研究中,我们检查了通过ERKD招募位点(DRS)破坏ERK-MYD88相互作用的生物学后果,同时保留ERK激酶活性。我们的结果表明,EI-52,一种靶向ERK-MYD88的小分子苯并咪唑相互作用诱导HRI介导的整合应激反应(ISR),导致特异性针对癌细胞的免疫原性凋亡。此外,EI-52在患者来源的肿瘤中表现出抗肿瘤功效,并在小鼠体内诱导抗肿瘤T细胞应答。这些发现表明,抑制ERK-MYD88相互作用可能是癌症治疗中一种有前途的治疗方法。
    The quest for targeted therapies is critical in the battle against cancer. The RAS/MAP kinase pathway is frequently implicated in neoplasia, with ERK playing a crucial role as the most distal kinase in the RAS signaling cascade. Our previous research demonstrated that the interaction between ERK and MYD88, an adaptor protein in innate immunity, is crucial for RAS-dependent transformation and cancer cell survival. In this study, we examine the biological consequences of disrupting the ERK-MYD88 interaction through the ERK D-recruitment site (DRS), while preserving ERK\'s kinase activity. Our results indicate that EI-52, a small-molecule benzimidazole targeting ERK-MYD88 interaction induces an HRI-mediated integrated stress response (ISR), resulting in immunogenic apoptosis specific to cancer cells. Additionally, EI-52 exhibits anti-tumor efficacy in patient-derived tumors and induces an anti-tumor T cell response in mice in vivo. These findings suggest that inhibiting the ERK-MYD88 interaction may be a promising therapeutic approach in cancer treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    在这里,我们报告了一种全新的生物活性聚合物,其特征是锍部分,具有诱导免疫原性细胞死亡(ICD)用于抗癌治疗的能力。优化的聚锍具有广泛的有效抗癌活性和显着的选择性。深入的机理研究表明,该聚合物通过膜破坏机制对癌细胞发挥其细胞毒性作用。这进一步引发了大量与损伤相关的分子模式的释放,有效触发ICD并导致全身抗癌免疫反应。值得注意的是,该化合物在B16-F10黑色素瘤肿瘤模型中表现出显著的抑制肿瘤生长的功效。此外,它表现出强大的免疫记忆效应,在再攻击模型和肺转移肿瘤模型中均能有效抑制肿瘤的复发和转移。据我们所知,这项研究代表了阳离子聚磺酸盐的开创性出口,不仅展示了它们对原发性肿瘤的显着安全性和有效性,还展示了它们激活长期免疫记忆的独特能力。
    Here we report a brand-new bioactive polymer featuring sulfonium moieties that exhibits the capability of inducing immunogenic cell death (ICD) for anticancer therapy. The optimized polysulfonium presents a wide spectrum of potent anticancer activity and remarkable selectivity. In-depth mechanistic studies reveal that the polymer exerts its cytotoxic effects on cancer cells through a membrane-disrupting mechanism. This further initiates the release of a plethora of damage-associated molecular patterns, effectively triggering ICD and resulting in systemic anticancer immune responses. Notably, the compound demonstrated significant efficacy in suppressing tumor growth in the B16-F10 melanoma tumor model. Furthermore, it exhibits robust immune memory effects, effectively suppressing tumor recurrence and metastasis in both the rechallenge model and the lung metastatic tumor model. To the best of our knowledge, the study represents the pioneering exportation of cationic polysulfoniums, showcasing not only their remarkable safety and efficacy against primary tumors but also their unique ability in activating long-term immune memory.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    由辐射诱导的免疫原性细胞死亡(ICD)驱动的放射免疫疗法正在成为解决仅适用于局部肿瘤治疗的常规放射疗法(RT)的潜在机会。然而,放疗期间ICD的有效激活受到辐射剂量的严重限制,弱肿瘤免疫原性,和肿瘤微环境(TME)引起的放射性抵抗。在这里,首先提出了一种新型的双金属混合纳米级配位纳米刺激器,由掺杂有铜离子(Cu2+)和铪离子(Hf4+)的磷酸盐骨架,然后用聚乙烯吡咯烷酮(PVP)改性。PVPylatedCu/Hf掺杂的磷酸盐纳米刺激器(表示为CHP)表现出有效的TME重编程,包括肿瘤内源性谷胱甘肽(GSH)的消耗,缓解肿瘤缺氧和M2表型巨噬细胞的复极化,从而在低X射线照射剂量下实现肿瘤放射增敏,肿瘤内源性活性氧(ROS)逐渐积累,并增加了细胞凋亡。此外,细胞凋亡可以通过ICD激活增强RT诱导的抗肿瘤免疫,最终导致强大的抗肿瘤免疫反应和长期免疫力,4T1荷瘤模型的远处肿瘤生长抑制证明。更有趣的是,发现在X射线照射过程中,CHP介导的角化可以增强。一起来看,这项工作提出了一种新的放射-投射-免疫疗法级联策略,为乳腺癌治疗领域的创新提供了新的视角。
    Radio-immunotherapy driven by radiation-induced immunogenic cell death (ICD) is emerging as a potential opportunity to address conventional radiotherapy (RT) that is only applicable to localized tumor treatment. However, the effective activation of ICD during RT is severely limited by radiation dose, weak tumor immunogenicity, and radio-resistance caused by tumor microenvironment (TME). Herein, a novel bimetallic hybrid nanoscale coordination nanostimulator is first proposed by phosphate backbone doped with copper ions (Cu2+) and hafnium ions (Hf4+), and then modified with polyvinylpyrrolidone (PVP). The PVPylated Cu/Hf-doped phosphate nanostimulator (denoted as CHP) exhibits effective reprogramming of TME, including depletion of tumor endogenous glutathione (GSH), relief of tumor hypoxia and repolarization of M2 phenotypic macrophages, thus achieving tumor radiosensitization at low X-ray irradiation dose, gradually accumulation of tumor endogenous reactive oxygen species (ROS) and augmenting cuproptosis. In addition, cuproptosis can amplify RT-induced anti-tumor immunity through ICD activation, ultimately resulting in a robust anti-tumor immune response and long-term immunity, evidenced by distant tumor growth inhibition of 4T1-tumor-bearing models. More interestingly, it is discovered that CHP-mediated cuproptosis can be intensifiable during X-ray irradiation. Taken together, this work presents a novel radio-cuproptosis-immunotherapy cascade strategy, offering a new perspective for innovation in the treatment field of breast cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    金属免疫疗法在治疗恶性肿瘤方面取得了巨大的临床前成功。尽管如此,肿瘤微环境(TME)内有限的免疫细胞浸润和受损的免疫原性显着阻碍了其向临床应用的转化。在这项研究中,开发了负载有氧化钙水合物(CaO2)纳米颗粒和STING激动剂diABZI-2(称为A-CaO2-Zn-LNP)的锌配位脂质纳米颗粒。从A-CaO2-Zn-LNP释放Zn2+和钙超载协同诱导免疫原性细胞死亡(ICD)。此外,在酸性条件下,CaO2纳米颗粒可以消耗H+并释放氧气(O2)。该处理增加了pH并减轻了TME的缺氧。随着diABZI-2激活cGAS-STING,A-CaO2-Zn-LNP最终通过减轻免疫抑制微环境导致增强的抗肿瘤全身免疫和长期免疫记忆。一起来看,A-CaO2-Zn-LNP提供了一种新的纳米平台,通过金属免疫疗法扩展了其在癌症治疗中的应用。
    Metalloimmunotherapy has achieved great preclinical success against malignant tumors. Nonetheless, the limited immune cell infiltration and impaired immunogenicity within the tumor microenvironment (TME) significantly hinder its translation to clinical applications. In this study, a zinc coordination lipid nanoparticle is developed loaded with calcium peroxide hydrate (CaO2) nanoparticles and the STING agonist diABZI-2, which is termed A-CaO2-Zn-LNP. The release of Zn2+ from the A-CaO2-Zn-LNP and the calcium overload synergistically induced immunogenic cell death (ICD). In addition, CaO2 nanoparticles can consume H+ and release oxygen (O2) under acidic conditions. This treatment increased the pH and alleviated the hypoxia of the TME. Along with cGAS-STING activation by diABZI-2, A-CaO2-Zn-LNP ultimately results in enhanced anti-tumor systemic immunity and long-term immune memory via alleviating the immunosuppressive microenvironment. Taken together, A-CaO2-Zn-LNP offers a new nanoplatform that expands its application for cancer treatment by metalloimmunotheray.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    程序性死亡(PD)1/PD配体1(PDL1)抑制剂是免疫检查点抑制剂(ICIs),可以促进HER2阳性乳腺癌的治疗;然而,他们的临床疗效仍然难以捉摸。氧增强光动力疗法(PDT)增加免疫原性细胞死亡(ICD),提供了一种有前途的策略,使肿瘤微环境对ICI更敏感。使用纳米气泡(NBs)水用于体内氧递送而获得的脂质包封的氧纳米气泡(Lipo-NBs-O2)可以促进增强的PDT。这里,双受体靶向的Lipo-NBs-O2(DRT@Lipo-NBs-O2)是通过用抗PDL1scFv和融合蛋白抗HER2scFv串联重复细胞色素c(抗HER2-nCytc)修饰Lipo-NBs-O2来制备的。酞菁铜是光敏剂(PS)。DRT@Lipo-PS-NBs-O2加上近红外照射导致强大的ICD诱导,增加DC活化和CD8+T细胞数量。用抗PDL1scFv修饰可改善DRT@Lipo-PS-NBs-O2的肿瘤分布并发挥ICI作用,激活CD8+T细胞并增强免疫治疗的效果。通过DRT@Lipo-PS-NBs-O2提供的氧减少P-糖蛋白表达。增强的PDT和Cytc可以导致肿瘤细胞死亡,从而减少免疫负担。在双受体靶向和激光局部照射下,肿瘤细胞受到PDT的联合作用,ICD,ICIs,和细胞凋亡;这有效地抑制了肿瘤的生长和转移。Lipo-NBs-O2提供了氧气输送和多种药物治疗的组合,以缓解HER2阳性乳腺癌。
    Programmed death (PD) 1/PD ligand 1 (PDL1) inhibitors are immune checkpoint inhibitors (ICIs) that may facilitate HER2-positive breast cancer treatment; however, their clinical efficacy remains elusive. Oxygen-enhanced photodynamic therapy (PDT) increases immunogenic cell death (ICD), providing a promising strategy to render the tumor microenvironment more sensitive to the ICIs. Lipid-encapsulated oxygen nanobubbles (Lipo-NBs-O2) obtained using nanobubbles (NBs) water for oxygen delivery in vivo can facilitate enhanced PDT. Here, dual-receptor targeted Lipo-NBs-O2 (DRT@Lipo-NBs-O2) is prepared by modifying Lipo-NBs-O2 with anti-PDL1 scFv and the fusion protein anti-HER2 scFv-tandem-repeat cytochrome c (anti-HER2-nCytc). Copper phthalocyanine is the photosensitizer (PS). DRT@Lipo-PS-NBs-O2 plus near-infrared irradiation leads to robust ICD induction, increasing DC activation and CD8+ T-cell numbers. Modification with anti-PDL1 scFv improves tumor distribution of DRT@Lipo-PS-NBs-O2 and plays the ICI role, invigorating CD8+ T cells and boosting the effects of immunotherapy. Oxygen supplied through DRT@Lipo-PS-NBs-O2 reduces P-glycoprotein expression. Enhanced PDT and Cytc can cause tumor cell death, thereby reducing the immune burden. Under dual receptor targeting and laser local irradiation, tumor cells become subject to the combination effects of PDT, ICD, ICIs, and apoptosis; this effectively suppresses tumor growth and metastasis. Lipo-NBs-O2 affords a combination of oxygen delivery and multidrug therapy to alleviate HER2-positive breast cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    肿瘤微环境(TME)中高水平的C(ROS)不仅会损害肿瘤细胞,还会降低免疫原性细胞死亡(ICD)的功效和肿瘤浸润性T淋巴细胞的活性。从而限制了免疫治疗的有效性。因此,ROS水平的精确调节对于有效消除肿瘤细胞和激活ICD诱导的免疫治疗至关重要。这里,一种智能蛋黄壳纳米平台(SPCCM),其特征是碳酸钙壳能够在酸性TME条件下分解,从而设计释放天然抗氧化剂原花青素(PAs)和光敏剂Ce6。PAs清除肿瘤内的ROS,延长T淋巴细胞的存活时间,而Ce6作为ICD诱导剂,在激光照射时产生高的ROS浓度,从而达到肿瘤细胞内的毒性阈值并诱导细胞凋亡。由此产生的凋亡细胞作为肿瘤相关抗原,促进树突状细胞(DC)成熟,并激活ICD。通过有效中和TME中的ROS,PA持续降低ROS水平,从而增强DC活化并恢复由ROS抑制的抗肿瘤免疫细胞活性(导致DC活化增加8倍)。这项研究通过精确调节ROS水平证明了光动力疗法和免疫疗法之间的有效协同作用。
    High level of C (ROS) within the tumor microenvironment (TME) not only damage tumor cells but also diminish the efficacy of immunogenic cell death (ICD) and the activity of tumor-infiltrating T lymphocytes, thereby limiting the effectiveness of immunotherapy. Therefore, precise modulation of ROS level is crucial to effectively eliminate tumor cells and activate ICD-induced immunotherapy. Here, an intelligent yolk shell nanoplatform (SPCCM) that features calcium carbonate shells capable of decomposing under acidic TME conditions, thereby releasing the natural antioxidant proanthocyanidins (PAs) and the photosensitizer Ce6 is designed. PAs scavenge ROS within tumors, extending the survival time of T lymphocytes, while Ce6, as an ICD inducer, generates high ROS concentrations upon laser irradiation, thus reaching the toxic threshold within tumor cells and inducing apoptosis. The resulting apoptotic cells serve as tumor-associated antigens, promoting dendritic cells (DCs) maturation, and activating ICD. By effectively neutralizing ROS in the TME, PAs sustainably reduce ROS level, thereby enhancing DCs activation and restoring antitumor immune cell activity suppressed by ROS (resulting in an eightfold increase in DCs activation). This study demonstrates effective synergistic effects between photodynamic therapy and immunotherapy by precisely modulating ROS level.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    骨肉瘤(OS),儿童和青少年骨最常见的原发性恶性肿瘤,免疫检查点抑制剂由于其抗肿瘤免疫应答不良而难以治疗。化疗和病毒疗法诱导免疫原性细胞死亡(ICD)和抗肿瘤免疫反应,在未经治疗的肿瘤中导致外镜效应。我们先前证明了端粒酶特异性复制型溶瘤腺病毒OBP-301和p53武装的OBP-702在人类OS细胞中的抗肿瘤活性。这里,我们展示了化疗药物(阿霉素,顺铂)和端粒酶特异性溶瘤腺病毒(OBP-301,p53武装的OBP-702)在人OS细胞中诱导ICD(U2OS,MNNG/HOS,SaOS-2)和鼠OS细胞(NHOS)。与化疗和OBP-301相比,OBP-702通过分泌三磷酸腺苷(ATP)和高迁移率族蛋白盒B1(HMGB1)在人OS细胞中诱导更深入的ICD。鼠NHOS细胞对OBP-702也比OBP-301更敏感。皮下NHOS肿瘤模型表明,与OBP-301相比,瘤内注射OBP-702显着增加了细胞毒性CD8T细胞的肿瘤浸润,并诱导了对未治疗肿瘤的外视作用。我们的结果表明,OBP-702是一种有前途的抗肿瘤试剂,可诱导ICD分泌ATP和HMGB1,并具有抗OS的作用。
    Osteosarcoma (OS), the most frequent primary malignant tumor of bone in children and adolescents, is refractory to immune checkpoint inhibitors due to its poor antitumor immune response. Chemotherapy and virotherapy induce immunogenic cell death (ICD) and antitumor immune responses, leading to the abscopal effect in untreated tumors. We previously demonstrated the antitumor activity of the telomerase-specific replication-competent oncolytic adenoviruses OBP-301 and p53-armed OBP-702 in human OS cells. Here, we show the therapeutic potential of chemotherapeutic drugs (doxorubicin, cisplatin) and telomerase-specific oncolytic adenoviruses (OBP-301, p53-armed OBP-702) to induce ICD in human OS cells (U2OS, MNNG/HOS, SaOS-2) and murine OS cells (NHOS). OBP-702 induced more profound ICD via the secretion of adenosine triphosphate (ATP) and high-mobility group box protein B1 (HMGB1) compared with chemotherapy and OBP-301 in human OS cells. Murine NHOS cells were also more sensitive to OBP-702 than OBP-301. Subcutaneous NHOS tumor models demonstrated that intratumoral injection of OBP-702 significantly increased the tumor infiltration of cytotoxic CD8+ T cells and induced the abscopal effect against non-treated tumors compared with OBP-301. Our results suggest that OBP-702 is a promising antitumor reagent to induce ICD with secretion of ATP and HMGB1 and the abscopal effect against OS.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    免疫原性细胞死亡(ICD)可以激活抗肿瘤免疫反应,这对于提高癌症治疗效果非常有吸引力。这里,这项工作报道了一种多功能砷(III)变构抑制剂Mech02,它通过敏化的生物催化反应诱导1O2的过度积累,导致细胞焦亡和放大的ICD效应。Mech02转化为Mech03后,对丙酮酸激酶M2的变构口袋产生更强的结合作用,进一步干扰肿瘤的无氧糖酵解途径。由Mech02引发的增强的DNA损伤和癌症干细胞的焦亡为完全清除肿瘤提供了保证。体内实验证明纳米粒Mech02-HANP能够激活免疫记忆效应并提高抗肿瘤免疫的持久性。总之,本研究首次将砷(Ⅲ)药效团作为增强ICD效应引发剂引入氮芥,为开发有效的多模式肿瘤治疗剂提供见解。
    Immunogenic cell death (ICD) could activate anti-tumor immune responses, which is highly attractive for improving cancer treatment effectiveness. Here, this work reports a multifunctional arsenic(III) allosteric inhibitor Mech02, which induces excessive accumulation of 1O2 through sensitized biocatalytic reactions, leading to cell pyroptosis and amplified ICD effect. After Mech02 is converted to Mech03, it could actualize stronger binding effects on the allosteric pocket of pyruvate kinase M2, further interfering with the anaerobic glycolysis pathway of tumors. The enhanced DNA damage triggered by Mech02 and the pyroptosis of cancer stem cells provide assurance for complete tumor clearance. In vivo experiments prove nanomicelle Mech02-HA NPs is able to activate immune memory effects and raise the persistence of anti-tumor immunity. In summary, this study for the first time to introduce the arsenic(III) pharmacophore as an enhanced ICD effect initiator into nitrogen mustard, providing insights for the development of efficient multimodal tumor therapy agents.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    胰腺导管腺癌(PDAC)的存活率为12%,并且针对PDAC的抗PD1疗法的多项临床试验均失败,表明需要新的治疗策略。在这项研究中,我们评估了米尔贝霉素肟(MBO)的潜力,一种抗寄生虫化合物,作为PDAC中的免疫调节剂。我们的结果表明,MBO通过诱导凋亡抑制多种PDAC细胞系的生长。体内研究表明,口服5mg/kgMBO可以抑制皮下和原位模型中PDAC肿瘤的生长49%和56%,分别。此外,与对照组相比,MBO治疗显著增加了荷瘤小鼠27天的存活率。有趣的是,MBO治疗小鼠的肿瘤CD8+T细胞浸润增加.值得注意的是,CD8+T细胞的消耗显著降低了MBO在小鼠中的抗肿瘤功效。Further,MBO显著增强抗PD1治疗的疗效,和联合治疗导致TME内更大比例的活性细胞毒性T细胞。在我们所有的临床前毒理学研究中,MBO是安全且耐受性良好的。总的来说,我们的研究为MBO对抗PDAC的应用提供了新的方向,并强调了MBO再利用以增强抗PD1免疫治疗的潜力.
    Pancreatic ductal adenocarcinoma (PDAC) has a survival rate of 12%, and multiple clinical trials testing anti-PD-1 therapies against PDAC have failed, suggesting a need for a novel therapeutic strategy. In this study, we evaluated the potential of milbemycin oxime (MBO), an antiparasitic compound, as an immunomodulatory agent in PDAC. Our results show that MBO inhibited the growth of multiple PDAC cell lines by inducing apoptosis. In vivo studies showed that the oral administration of 5 mg/kg MBO inhibited PDAC tumor growth in both subcutaneous and orthotopic models by 49% and 56%, respectively. Additionally, MBO treatment significantly increased the survival of tumor-bearing mice by 27 days as compared to the control group. Interestingly, tumors from MBO-treated mice had increased infiltration of CD8+ T cells. Notably, depletion of CD8+ T cells significantly reduced the anti-tumor efficacy of MBO in mice. Furthermore, MBO significantly augmented the efficacy of anti-PD-1 therapy, and the combination treatment resulted in a greater proportion of active cytotoxic T cells within the tumor microenvironment. MBO was safe and well tolerated in all our preclinical toxicological studies. Overall, our study provides a new direction for the use of MBO against PDAC and highlights the potential of repurposing MBO for enhancing anti-PD-1 immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    TartroonD(TRL)是由Teredinibacterturnerae产生的,一种共生的降解纤维素的细菌。免疫原性细胞死亡(ICD)诱导有助于更好和更持久的抗癌治疗反应。接受ICD的肿瘤细胞会触发免疫系统的激活,作为疫苗。
    目的:本研究旨在评估TRL诱导ICD。
    方法:通过SRB测定评价细胞活力。细胞应激,细胞死亡,通过流式细胞术和免疫印迹评估ICD特征和抗原呈递分子。
    结果:TRL对7种肿瘤细胞系(L929,HCT116,B16-F10,WM293A,SK-MEL-28,PC-3M,和MCF-7)和非肿瘤细胞(HEK293A),抑制浓度平均值(IC50)为0.03uM至13uM。转移性黑色素瘤,SK-MEL-28,B16-F10,和WM293A,是更敏感的细胞系,IC50范围为0.07至1.2uM。TRL诱导细胞凋亡以及自噬和内质网应激,并释放ICD的典型损伤相关分子模式(DAMPs),如钙网蛋白,ERp57和HSP70暴露,和HMGB1释放。此外,暴露于TRL的黑色素瘤B16-F10增加了抗原呈递分子MHCII和CD1d的表达,并诱导了C57BL/6小鼠脾细胞的激活。
    结论:尽管靶向治疗和免疫疗法取得了最新进展,超过一半的患者无法治愈晚期转移性黑色素瘤.ICD诱导剂对抗癌治疗产生更好和持久的反应。我们的发现揭示了一种在黑色素瘤中诱导ICD的海洋抗癌候选物。
    Tartrolon D (TRL) is produced by Teredinibacter turnerae, a symbiotic cellulose-degrading bacteria in shipworm gills. Immunogenic cell death (ICD) induction contributes to a better and longer-lasting response to anticancer treatment. Tumor cells undergoing ICD trigger activation of the immune system, as a vaccine.
    OBJECTIVE: This study aimed to evaluate ICD induction by TRL.
    METHODS: Cell viability was evaluated by SRB assay. Cell stress, cell death, ICD features and antigen-presenting molecules were evaluated by flow cytometry and immunoblot.
    RESULTS: TRL showed antiproliferative activity on 7 tumor cell lines (L929, HCT 116, B16-F10, WM293A, SK-MEL-28, PC-3M, and MCF-7) and a non-tumor cell (HEK293A), with an inhibition concentration mean (IC50) ranging from 0.03 μM to 13 μM. Metastatic melanomas, SK-MEL-28, B16-F10, and WM293A, were more sensitive cell lines, with IC50 ranging from 0.07 to 1.2 μM. TRL induced apoptosis along with autophagy and endoplasmic reticulum stress and release of typical damage-associated molecular patterns (DAMPs) of ICD such calreticulin, ERp57, and HSP70 exposure, and HMGB1 release. Additionally, melanoma B16-F10 exposed to TRL increased expression of antigen-presenting molecules MHC II and CD1d and induced activation of splenocytes of C57BL/6 mice.
    CONCLUSIONS: In spite of recent advances provided by target therapy and immunotherapy, advanced metastatic melanoma is incurable for more than half of patients. ICD inducers yield better and long-lasting responses to anticancer treatment. Our findings shed light on an anticancer candidate of marine origin that induces ICD in melanoma.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号