intratumoral injection

肿瘤内注射
  • 文章类型: Journal Article
    对于肺癌患者,静脉内(i.v.)化疗增加静脉内(i.v.)免疫疗法可改善总生存率,但受到协同副作用和未知的限制。肿瘤内高度可变的最终细胞毒性剂量。假设静脉内化疗和免疫疗法之间的协同作用是由于化疗引起的细胞损伤而发生的。一种被证明在肿瘤微环境中驱动抗原呈递的机制。因此可以优化化疗的肿瘤内递送以使肿瘤细胞损伤最大化。为了评估肿瘤细胞损伤与死亡之间的平衡,我们建立了直接注射后三种不同化疗药物在肺癌肿瘤内的瘤内动力学作为注射部位和数量的函数的计算模型.我们基于从胸部CT扫描获得的肺肿瘤的形态模型。我们发现中央注射与外周注射药物在肿瘤细胞损伤程度上没有有意义的差异,但单次注射与在多个注射部位之间分配总剂量相比存在显著差异.重要的是,我们还发现,用于静脉内给药的标准化疗浓度可有效引起细胞死亡,但浓度过高,不会产生明显的细胞损伤.这表明诱导最大的肿瘤细胞损伤,最佳浓度应该比通常用于静脉治疗的浓度低几个数量级。
    The addition of intravenous (i.v.) chemotherapy to i.v. immunotherapy for patients with lung cancer results in improved overall survival but is limited by synergistic side effects and an unknown, highly variable final cytotoxic dose within the tumor. The synergy between i.v. chemo- and immunotherapies is hypothesized to occur as a result of cell injury caused by chemotherapy, a mechanism demonstrated to drive antigen presentation within the tumor microenvironment. Intratumoral delivery of chemotherapy may thus be optimized to maximize tumor cell injury. To assess the balance between the damage versus the death of tumor cells, we developed a computational model of intratumoral dynamics within a lung cancer tumor for three different chemotherapy agents following direct injection as a function of location and number of injection sites. We based the model on the morphology of a lung tumor obtained from a thoracic CT scan. We found no meaningful difference in the extent of tumor cell damage between a centrally injected versus peripherally injected agent, but there were significant differences between a single injection versus when the total dose was apportioned between multiple injection sites. Importantly, we also found that the standard chemotherapeutic concentrations used for intravenous administration were effective at causing cell death but were too high to generate significant cell injury. This suggests that to induce maximal tumor cell injury, the optimal concentration should be several orders of magnitude lower than those typically used for intravenous therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    肉瘤来源于间充质肿瘤,有许多亚型,占所有成人恶性肿瘤的1%和儿童恶性肿瘤的15%。转移性或复发性肉瘤的预后仍然较差。目前的研究提出了两例肉瘤,他们参加了实体肿瘤的I期剂量递增试验,以前所有标准疗法都失败了。这些患者接受了VG161治疗,VG161是一种免疫刺激的单纯疱疹病毒1型溶瘤病毒,具有IL-12,IL-15和IL-15受体α单位的有效载荷,和程序性细胞死亡1(PD-1)/PD-1配体1阻断肽。两种情况都证明疾病稳定是最好的反应,伴随着无进展生存期的显著延长(软骨肉瘤11.8个月,软组织肉瘤11.9个月,分别),剂量为2.5×108PFU/周期。此外,这种治疗导致了抗癌免疫力的激活,从细胞因子中可以明显看出,外周血和/或肿瘤活检样本的淋巴细胞亚群和相关通路分析。这些有希望的结果表明,VG161单一疗法有望作为肉瘤的有效治疗方法,并需要通过临床试验进行进一步研究。报告的两名患者是进行的I期临床试验的一部分,并在澳大利亚的澳大利亚新西兰临床试验注册中心注册(注册编号:ACTRN12620000244909;注册日期,2月26日,2020)。
    Sarcoma is derived from mesenchymal neoplasms and has numerous subtypes, accounting for 1% of all adult malignancies and 15% of childhood malignancies. The prognosis of metastatic or recurrent sarcoma remains poor. The current study presents two cases of sarcoma enrolled in a phase I dose escalation trial for solid tumor, who had previously failed all standard therapies. These patients were treated with VG161, an immune-stimulating herpes simplex virus type 1 oncolytic virus with payloads of IL-12, IL-15 and IL-15 receptor α unit, and a programmed cell death 1 (PD-1)/PD-1 ligand 1 blocking peptide. Both cases demonstrated stable disease as the best response, accompanied by a noteworthy prolongation of progression-free survival (11.8 months for chondrosarcoma and 11.9 months for soft tissue sarcoma, respectively) at a dose of 2.5×108 PFU/cycle. In addition, the treatment led to the activation of anti-cancer immunity, as evident from cytokine, lymphocyte subset and related pathway analyses of peripheral blood and/or tumor biopsy samples. These promising results suggest that VG161 monotherapy holds promise as an effective treatment for sarcoma and warrants further investigation through clinical trials. The two reported patients were part of a phase I clinical trial conducted and registered on the Australian New Zealand Clinical Trials Registry in Australia (registration no. ACTRN12620000244909; registration date, 26 February, 2020).
    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
    背景:免疫治疗为癌症治疗开辟了一系列新的策略。作为一种新兴的方法,化学免疫疗法创新性地扩展了用于癌症管理的常规化学治疗剂的成就。
    目的:开发了一种有效的化学免疫疗法,利用极简静电复合纳米颗粒(NP)整合的肿瘤免疫原性细胞死亡(ICD)和免疫激动剂,作为一种水密的“原位”疫苗,用于癌症治疗,通过方便的肿瘤内给药和最小化的全身毒性。
    方法:将化学修饰的pH敏感的顺式-乌亚甲基-多柔比星(CAD)和免疫佐剂未甲基化的胞嘧啶-磷酸-鸟嘌呤(CpG)与聚阳离子聚乙烯亚胺(PEI)共包装通过静电相互作用构建PEI/CpG/CADNP。通过肿瘤内注射,这种带正电荷的NP可以毫不费力地滞留在肿瘤部位并被肿瘤细胞内吞.然后,多柔比星通过内体酸度诱导的顺式-乌头酰裂解释放,并进一步触发肿瘤ICD,垂死的肿瘤细胞可以释放损伤相关分子模式(DAMPs)以募集树突状细胞(DCs).同时,整个肿瘤碎片衍生为多种抗原,并与免疫刺激CpG合作,激发DC成熟并激活全面的抗肿瘤免疫。
    结果:在侵袭性小鼠黑色素瘤肿瘤模型中实现了显著的肿瘤抑制,验证了这种极简CAD/CpG联合交付NP的可行性和有效性。
    结论:这项研究为有效的癌症化学免疫疗法提供了方便和有希望的范例。
    BACKGROUND: Immunotherapy has unprecedentedly opened up a series of neoteric tactics for cancer treatment. As a burgeoning approach, chemo-immunotherapy has innovatively expanded the accomplishments of conventional chemotherapeutic agents for cancer governing.
    OBJECTIVE: An efficacious chemo-immunotherapy leveraging minimalist electrostatic complex nanoparticle (NP) integrated tumor immunogenic cell death (ICD) and immunoagonist was developed as a watertight \"in situ\" vaccine for cancer therapy through convenient intratumoral administration with minimized systemic toxicity.
    METHODS: Chemical-modified pH-sensitive cis-aconityl-doxorubicin (CAD) and immunoadjuvant unmethylated cytosine-phosphate-guanine (CpG) were co-packaged by polycationic polyethylenimine (PEI) though electrostatic-interaction to construct PEI/CpG/CAD NP. By intratumoral injection, this positively charged NP could be detained at tumor site and endocytosed by tumor cells effortlessly. Then, doxorubicin was released through cis-aconityl cleavage induced by endosomal-acidity and further triggered tumor ICD, the moribund tumor cells could release damage-associated molecular patterns (DAMPs) to recruit dendritic cells (DCs). Meanwhile, the entire tumor debris derived into diversified antigens and cooperated with immunostimulatory CpG to excite DC maturation and activated comprehensive antitumor immunity.
    RESULTS: Prominent tumor suppression was achieved in aggressive mouse melanoma tumor model, which verified the feasibility and effectiveness of this minimalist CAD/CpG-codelivered NP.
    CONCLUSIONS: This study has provided a convenient and promising paradigm for potent cancer chemo-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
    背景:作者的临床前研究证实,注射到局部病变中的RO佐剂(由TLR7激动剂[咪喹莫特/R837]和OX40激动剂组成)可诱导原发肿瘤和远处转移的消退。作者建议通过“R-ISV-RO”原位策略加抗PD-1单克隆抗体在晚期肿瘤中实现局部控制并发挥远视作用。方法:本研究为单中心,探索性,评估R-ISV-RO联合抗PD-1单克隆抗体在晚期肿瘤中的疗效和安全性的II期试验。将招募30名具有一个或多个可进行放射或注射的可测量的脑外病变的患者。主要终点是目标病变的客观反应率。讨论/结论:新策略的有效性和安全性将通过本临床试验得到进一步验证。临床试验注册:ChiCTR2100053870(www。chictr.org.cn/)。
    Background: The authors\' preclinical study has confirmed that RO adjuvant (composed of TLR 7 agonists [imiquimod/R837] and OX40 agonists) injected into local lesions induces the regression of both primary tumor and distant metastasis. The authors propose to realize local control and exert abscopal effect through an \'R-ISV-RO\' in situ strategy plus anti-PD-1 monoclonal antibody in advanced tumors. Methods: This study is a single-center, exploratory, phase II trial to evaluate the efficacy and safety of R-ISV-RO plus anti-PD-1 monoclonal antibody in advanced tumors. 30 patients with one or more measurable extracerebral lesions that are accessible for radiation or injection will be enrolled. The primary endpoint is the objective response rate of target lesions. Discussion/Conclusion: The efficacy and safety of the novel strategy will be further validated through this clinical trial.Clinical trial registration: ChiCTR2100053870 (www.chictr.org.cn/).
    [Box: see text].
    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
    这篇综述总结了大表面积微粒紫杉醇(LSAM-PTX)和多西他赛(LSAM-DTX)用于局部治疗原发性癌的发展,重点是免疫调节。LSAM-PTX和LSAM-DTX的肿瘤内(IT)递送提供持续、几个星期的治疗药物水平。临床前研究和临床试验报道了原发性肿瘤和外周血中肿瘤体积(TV)和免疫调节的减少,先天和适应性免疫细胞增加,抑制细胞减少。在高风险非肌肉浸润性膀胱癌(LSAM-DTX)和不可切除的局部胰腺癌(LSAM-PTX)的临床试验中,免疫细胞的检查点表达水平升高。在ITLSAM-DTX和ITLSAM-PTX以及抗mCTLA-4和抗mPD-1之后,免疫效应细胞中的TV减少和增加分别发生。在4T1-Luc乳腺癌模型中组合疗法的协同益处包括用ITLSAM-DTX+抗mCTLA-4减少转移。它LSAM-PTX和LSAM-DTX是杀肿瘤的,免疫增强,并且可以改善实体瘤对免疫检查点抑制剂的反应,而没有额外的全身毒性。
    This review summarizes development of large surface area microparticle paclitaxel (LSAM-PTX) and docetaxel (LSAM-DTX) for local treatment of primary carcinomas with emphasis on immunomodulation. Intratumoral (IT) delivery of LSAM-PTX and LSAM-DTX provides continuous, therapeutic drug levels for several weeks. Preclinical studies and clinical trials reported a reduction in tumor volume (TV) and immunomodulation in primary tumor and peripheral blood with increases in innate and adaptive immune cells and decreases in suppressor cells. Increased levels of checkpoint expression of immune cells occurred in clinical trials of high-risk non-muscle-invasive bladder cancer (LSAM-DTX) and unresectable localized pancreatic cancer (LSAM-PTX). TV reduction and increases in immune effector cells occurred following IT LSAM-DTX and IT LSAM-PTX together with anti-mCTLA-4 and anti-mPD-1, respectively. Synergistic benefits from combinatorial therapy in a 4T1-Luc breast cancer model included reduction of metastasis with IT LSAM-DTX + anti-mCTLA-4. IT LSAM-PTX and LSAM-DTX are tumoricidal, immune enhancing, and may improve solid tumor response to immune checkpoint inhibitors without additional systemic toxicity.
    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
    目的:实体恶性肿瘤的经典近距离放射治疗是一种侵入性方法,通常会导致剂量分布不均,同时需要在一定时间后手术去除密封的放射性种子源。为了规避这些问题,我们报道了作为新型纳米级近距离放射治疗剂的内在放射性标记和阿拉伯树胶糖蛋白功能化的[169Yb]Yb2O3纳米种子的合成,可以通过肿瘤内注射直接给药用于肿瘤治疗。
    方法:169Yb(T½=32天)是通过在核反应堆中对富集的(在168Yb中为15.2%)Yb2O3靶进行中子辐照而产生的,放射化学转化为[169Yb]YbCl3并用于纳米颗粒(NP)合成。通过在阿拉伯树胶糖蛋白培养基中控制Yb3离子的水解来合成固有的放射性标记的NP。体内SPECT/CT成像,放射自显影,在肿瘤内注射放射性标记的NP后,对携带B16F10肿瘤的C57BL/6小鼠进行了生物分布研究。进行系统肿瘤消退研究和组织病理学分析以证明在相同小鼠模型中的治疗功效。
    结果:纳米制剂是具有高胶体和放射化学稳定性的澄清溶液。通过SPECT/CT成像和放射自显影研究观察到放射性标记的纳米制剂在肿瘤块中的均匀分布和保留。在肿瘤消退研究中,与对照组相比,用不同剂量的放射性标记的NP显着阻止了肿瘤的生长,并且在〜27.8MBq剂量下观察到了最佳的治疗效果。在组织病理学分析中,有丝分裂细胞的损失在治疗组的肿瘤组织中是明显的,而肾脏没有明显损伤,肺,观察肝脏组织形态。
    结论:这些结果有望使纳米级近距离放射治疗成为无法切除的实体癌的临床实用治疗方式。
    OBJECTIVE: Classical brachytherapy of solid malignant tumors is an invasive procedure which often results in an uneven dose distribution, while requiring surgical removal of sealed radioactive seed sources after a certain period of time. To circumvent these issues, we report the synthesis of intrinsically radiolabeled and gum Arabic glycoprotein functionalized [169Yb]Yb2O3 nanoseeds as a novel nanoscale brachytherapy agent, which could directly be administered via intratumoral injection for tumor therapy.
    METHODS: 169Yb (T½ = 32 days) was produced by neutron irradiation of enriched (15.2% in 168Yb) Yb2O3 target in a nuclear reactor, radiochemically converted to [169Yb]YbCl3 and used for nanoparticle (NP) synthesis. Intrinsically radiolabeled NP were synthesized by controlled hydrolysis of Yb3+ ions in gum Arabic glycoprotein medium. In vivo SPECT/CT imaging, autoradiography, and biodistribution studies were performed after intratumoral injection of radiolabeled NP in B16F10 tumor bearing C57BL/6 mice. Systematic tumor regression studies and histopathological analyses were performed to demonstrate therapeutic efficacy in the same mice model.
    RESULTS: The nanoformulation was a clear solution having high colloidal and radiochemical stability. Uniform distribution and retention of the radiolabeled nanoformulation in the tumor mass were observed via SPECT/CT imaging and autoradiography studies. In a tumor regression study, tumor growth was significantly arrested with different doses of radiolabeled NP compared to the control and the best treatment effect was observed with ~ 27.8 MBq dose. In histopathological analysis, loss of mitotic cells was apparent in tumor tissue of treated groups, whereas no significant damage in kidney, lungs, and liver tissue morphology was observed.
    CONCLUSIONS: These results hold promise for nanoscale brachytherapy to become a clinically practical treatment modality for unresectable solid cancers.
    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
    植入是一种现代的化疗药物给药方法,由于给药频率低,具有高度的针对性和更好的患者耐受性。植入物能够控制释放,这使得它们成为输注化疗的可行替代方案,让患者享受更好的生活质量,而无需长期住院。与皮下植入相比,瘤内植入在靶向和副作用方面具有许多显著优势,但就临床试验而言,对化疗这一领域的了解仍然很少。同时,对于肿瘤内给药的植入物和注射剂形式的药物有更多已知的发展。经典肿瘤内植入物的缺点是需要手术干预以安装系统,并且一些专家指出肿瘤破裂的风险增加。新一代植入物是在各种刺激的影响下,由于相变(溶胶-凝胶转变)而在肿瘤中形成的原位植入物系统。在这些系统中,有些系统对某些类型的恶性肿瘤具有高度选择性。这种系统是注射的,具有肿瘤内注射的所有优点,但是由于相变发生在原位,它们形成允许长期释放化疗剂的储库形式。
    Implantation is a modern method of administering chemotherapeutic agents, with a highly targeted effect and better patient tolerance due to the low frequency of administration. Implants are capable of controlled release, which makes them a viable alternative to infusional chemotherapy, allowing patients to enjoy a better quality of life without the need for prolonged hospitalization. Compared to subcutaneous implantation, intratumoral implantation has a number of significant advantages in terms of targeting and side effects, but this area of chemotherapy is still poorly understood in terms of clinical trials. At the same time, there are more known developments of drugs in the form of implants and injections for intratumoral administration. The disadvantages of classical intratumoral implants are the need for surgical intervention to install the system and the increased risk of tumor rupture noted by some specialists. The new generation of implants are in situ implants-systems formed in the tumor due to a phase transition (sol-gel transition) under the influence of various stimuli. Among this systems some are highly selective for a certain type of malignant neoplasm. Such systems are injected and have all the advantages of intratumoral injections, but due to the phase transition occurring in situ, they form depot forms that allow the long-term release of chemotherapeutic agents.
    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
    介入治疗越来越多地用于肝细胞癌(HCC)的临床试验。索拉非尼是肝癌的一线治疗药物,然而,化学抗性不变地发生并影响治疗的有效性。在之前的研究中,开发了用于HCC的去甲斑皮素脂质体乳液混合(NLEH)递送系统。本研究旨在检查NLEH和索拉非尼联合瘤内注射治疗HCC的疗效。索拉非尼联合NLEH通过协同上调caspase-9激活细胞凋亡通路,促进细胞毒性,凋亡(64.57%),和G2/M细胞周期阻滞(48.96%)。去甲斑驳素可以通过抵消索拉非尼诱导的Akt磷酸化来减轻索拉非尼耐药性。此外,瘤内注射NLEH在24h内显示出肿瘤中的持续积累,并且没有分布到其他主要器官。NLEH与口服索拉非尼的瘤内注射在体内显示出最有效的肿瘤生长抑制作用(77.91%)。H&E染色结果以及肾和肝功能测试的指标证明了该组合疗法的安全性。总的来说,这些结果表明,瘤内注射NLEH联合口服索拉非尼治疗是肝癌合理的潜在治疗选择.
    Interventional therapies are increasingly used in clinical trials for hepatocellular carcinoma (HCC). Sorafenib is the front-line remedy for HCC, however, chemoresistance occurs immutably and affects the effectiveness of treatment. In a previous study, a norcantharidin liposome emulsion hybrid (NLEH) delivery system for HCC was developed. This study aims to examine the therapeutic effects of the combination of intratumoral injection of NLEH and sorafenib in treating HCC. Sorafenib combined with NLEH activated the apoptosis pathway by synergistically upregulating caspase-9, promoting cytotoxicity, apoptosis (64.57%), and G2/M cell cycle arrest (48.96%). Norcantharidin could alleviate sorafenib resistance by counteracting sorafenib-induced phosphorylation of Akt. Additionally, intratumoral injection of NLEH exhibited a sustained accumulation in the tumor within 24 h and didn\'t distribute to other major organs. Intratumoral injection of NLEH in combination with oral sorafenib displayed the most potent tumor growth inhibitory effect (77.91%) in vivo. H&E staining results and the indicators of the renal and liver function tests demonstrated the safety of this combination therapy. Overall, these results showed that intratumoral injection of NLEH in combination with oral sorafenib treatment represented a rational potential therapeutic option for HCC.
    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
    光动力疗法(PDT)是黑色素瘤的潜在治疗策略。作为第二代光敏剂,酞菁锌(ZnPc)在抗肿瘤PDT,例如在红色和近红外区域的强烈吸收,高的光和化学稳定性,等。然而,ZnPc水溶性差,由于分子间的π-π相互作用,易于聚集,这限制了它的应用。在这项研究中,筛选了各种溶剂和表面活性剂,用于溶解ZnPc,制备ZnPc@SDC-TPGS胶束和热敏原位凝胶。在测试了热敏凝胶对PDT的细胞毒性作用后,评估了通过瘤内注射对小鼠PDT的抗肿瘤作用,包括体重,和肿瘤重量,体积和形态。研究了ZnPc原位凝胶诱导的细胞死亡途径以及活性氧产生量与肿瘤细胞凋亡率的关系。结果表明,N-甲基吡咯烷酮(NMP)与2%SDC混合,含2%TPGS和2%SDC的水溶液用于合成ZnPc@SDC-TPGS胶束和热敏原位凝胶。热敏凝胶的细胞毒性作用显示ZnPc@SDC-TPGS原位凝胶具有良好的抑瘤作用,空白凝胶没有毒性。照射下含有3μgZnPc的肿瘤内注射原位凝胶在黑色素瘤小鼠中显示出良好的肿瘤抑制作用。细胞凋亡已被确立为细胞死亡的主要途径,活性氧(ROS)的产生在ZnPc@SDC-TPGS原位凝胶诱导的细胞凋亡中起着至关重要的作用。总之,肿瘤内注射ZnPc@SDC-TPGS热敏原位凝胶为黑色素瘤提供了一种有希望的局部治疗选择.
    Photodynamic therapy (PDT) is a potential treatment strategy for melanoma. As a second-generation photosensitizer, Zinc phthalocyanine (ZnPc) has many advantages for anti-tumor PDTs, such as strong absorption in the red and near infrared regions, high photo and chemical stability, etc. However, ZnPc has a poor water solubility and is apt to aggregate due to the π-π interaction between molecules, which limits its applications. In this study, various solvents and surfactants were screened for dissolving ZnPc and preparing ZnPc@SDC-TPGS micelle and thermosensitive in situ gel. After the cytotoxic effects of thermosensitive gels on PDT were tested, the antitumor effects on PDT of them in mice by intratumoral injection were evaluated, including body weight, and tumor weight, volume and morphology. The cell death pathway and the relationship of reactive oxygen species yield with apoptotic rate of tumor cells induced by ZnPc in situ gel were investigated. The results were that N-methyl-pyrrolidone (NMP) mixed with 2 % SDC and aqueous solution containing 2 % TPGS and 2 % SDC were used to synthesize ZnPc@SDC-TPGS micelle and the thermosensitive in situ gel. The cytotoxic effects of thermosensitive gels showed good tumor suppression of ZnPc@SDC-TPGS in situ gel and no toxicity of the blank gel. Intratumoral injection in situ gel containing 3 µg ZnPc under irradiation demonstrated good tumor inhibition in mice with melanoma. Apoptosis has been established as the primary pathway of cell death, and the production of reactive oxygen species (ROS) plays a crucial role in cellular apoptosis induced by ZnPc@SDC-TPGS in situ gel. In conclusion, the intratumoral injection of ZnPc@SDC-TPGS thermosensitive in situ gel provides a promising local treatment option for melanoma.
    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
    头颈部鳞状细胞癌的死亡率很高。褪黑激素已被证明在不同类型的癌症中具有抑癌作用。然而,据报道,体内应用的结果不一致。因此,需要一种替代的给药途径来提高生物利用度并确定用于癌症治疗的褪黑激素的最佳剂量。另一方面,患者来源的肿瘤模型的使用改变了药物研究领域,因为它们反映了患者肿瘤组织的异质性.在本研究中,我们探讨了提高褪黑素在肿瘤中生物利用度的机制,并研究了其作为佐剂提高顺铂在异种移植细胞系和原代人HNSCC的治疗效果的潜力.我们分析了在Cal-27和SCC-9异种移植物以及患者来源的异种移植物中皮下或瘤内施用的两种不同褪黑素制剂的效果。还评估了褪黑素对肿瘤线粒体代谢的影响以及褪黑素对肿瘤细胞迁移的作用。与皮下褪黑激素获得的结果相反,肿瘤内注射褪黑素可显著抑制HNSCC来源的异种移植物的肿瘤进展,以及源自患者的异种移植物。有趣的是,瘤内注射褪黑素增强了CDDP的作用,降低Cal-27肿瘤生长。我们证明褪黑激素增加了肿瘤中ROS的产生和细胞凋亡,靶向线粒体。褪黑素还降低迁移能力和转移标志物。这些结果说明了肿瘤内褪黑素治疗的巨大临床潜力,并鼓励未来在癌症患者中进行临床试验以建立适当的临床褪黑素治疗。
    Head and neck squamous cell carcinoma present a high mortality rate. Melatonin has been shown to have oncostatic effects in different types of cancers. However, inconsistent results have been reported for in vivo applications. Consequently, an alternative administration route is needed to improve bioavailability and establish the optimal dosage of melatonin for cancer treatment. On the other hand, the use of patient-derived tumor models has transformed the field of drug research because they reflect the heterogeneity of patient tumor tissues. In the present study, we explore mechanisms for increasing melatonin bioavailability in tumors and investigate its potential as an adjuvant to improve the therapeutic efficacy of cisplatin in the setting of both xenotransplanted cell lines and primary human HNSCC. We analyzed the effect of two different formulations of melatonin administered subcutaneously or intratumorally in Cal-27 and SCC-9 xenografts and in patient-derived xenografts. Melatonin effects on tumor mitochondrial metabolism was also evaluated as well as melatonin actions on tumor cell migration. In contrast to the results obtained with the subcutaneous melatonin, intratumoral injection of melatonin drastically inhibited tumor progression in HNSCC-derived xenografts, as well as in patient-derived xenografts. Interestingly, intratumoral injection of melatonin potentiated CDDP effects, decreasing Cal-27 tumor growth. We demonstrated that melatonin increases ROS production and apoptosis in tumors, targeting mitochondria. Melatonin also reduces migration capacities and metastasis markers. These results illustrate the great clinical potential of intratumoral melatonin treatment and encourage a future clinical trial in cancer patients to establish a proper clinical melatonin treatment.
    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