Thermosensitive liposomes

热敏脂质体
  • 文章类型: Journal Article
    已经开发了癌症药物多柔比星的脂质体制剂以解决在常规制剂中施用该药物导致的严重副作用。其中,热敏脂质体多柔比星与局部肿瘤部位的轻度热疗结合时,可增强肿瘤靶向性和有效的药物释放。利用局部热疗的放射增敏益处,放射治疗与热激活脂质体系统的整合被认为可以增强抗肿瘤功效。本研究探索了一种联合热敏脂质体阿霉素的协同治疗策略,轻度高热,和放射治疗,使用原位小鼠乳腺癌模型。序贯多模式治疗方案,结合低剂量化疗和放疗,与以升高的剂量应用单药治疗相比,实质上推迟了原发性肿瘤生长的进展。还观察到未加热的远处病变的改善。此外,联合治疗的毒性与热敏脂质体治疗或低剂量单独放疗的毒性相当.这些结果强调了多模式治疗策略在改善治疗效果同时减少乳腺癌治疗中的不良反应的潜力。为热敏脂质体阿霉素应用的未来改进提供有价值的见解。
    Liposome formulations of the cancer drug doxorubicin have been developed to address the severe side effects that result from administration of this drug in a conventional formulation. Among them, thermosensitive liposomal doxorubicin presents enhanced tumor targeting and efficient drug release when combined with mild hyperthermia localized to the tumor site. Exploiting the radiosensitizing benefits of localized thermal therapy, the integration of radiation therapy with the thermally activated liposomal system is posited to amplify the anti-tumor efficacy. This study explored a synergistic therapeutic strategy that combines thermosensitive liposomal doxorubicin, mild hyperthermia, and radiotherapy, using an orthotopic murine model of breast cancer. The protocol of sequential multi-modal treatment, incorporating low-dose chemotherapy and radiotherapy, substantially postponed the progression of primary tumor growth in comparison to the application of monotherapy at elevated dosages. Improvements in unheated distant lesions were also observed. Furthermore, the toxicity associated with the combination treatment was comparable to that of either thermosensitive liposome treatment or radiation alone at low doses. These outcomes underscore the potential of multi-modal therapeutic strategies to refine treatment efficacy while concurrently diminishing adverse effects in the management of breast cancer, providing valuable insight for the future refinement of thermosensitive liposomal doxorubicin applications.
    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
    本研究设计了多功能脂质体聚多巴胺纳米颗粒(MPM@Lipo),联合化疗,光热疗法(PTT)和氧富集可以清除过度增殖的炎症细胞并改善类风湿性关节炎(RA)治疗的缺氧微环境。MPM@Lipo显著清除细胞内活性氧,缓解关节缺氧,因此有助于M1巨噬细胞复极化为M2表型。此外,MPM@Lipo可在炎症关节处积聚,抑制炎症因子的产生,并在体内保护软骨,在大鼠佐剂诱导的关节炎模型中有效缓解RA进展。此外,在激光照射下,MPM@Lipo可以提高温度,不仅可以明显消除过度增殖的炎症细胞,而且可以加速甲氨蝶呤和氧气的产生。产生优异的RA治疗效果。总的来说,使用协同化疗/PTT/氧富集疗法治疗RA是一种有效的潜在策略.
    Amultifunctional liposomal polydopamine nanoparticle (MPM@Lipo) was designed in this study, to combine chemotherapy, photothermal therapy (PTT) and oxygen enrichment to clear hyperproliferating inflammatory cells and improve the hypoxic microenvironment for rheumatoid arthritis (RA) treatment. MPM@Lipo significantly scavenged intracellular reactive oxygen species and relieved joint hypoxia, thus contributing to the repolarization of M1 macrophages into M2 phenotype. Furthermore, MPM@Lipo could accumulate at inflammatory joints, inhibit the production of inflammatory factors, and protect cartilage in vivo, effectively alleviating RA progression in a rat adjuvant-induced arthritis model. Moreover, upon laser irradiation, MPM@Lipo can elevate the temperature to not only significantly obliterate excessively proliferating inflammatory cells but also accelerate the production of methotrexate and oxygen, resulting in excellent RA treatment effects. Overall, the use of synergistic chemotherapy/PTT/oxygen enrichment therapy to treat RA is a powerful potential strategy.
    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
    不同疗法的组合是癌症治疗的有吸引力的方法。然而,同步不同的疗法以最大化治疗效果是一个挑战。在这项工作中,通过将磁性Fe3O4纳米颗粒和负载多柔比星(Dox)的热敏脂质体与生物可降解聚合物杂交,制备了一种能够同步磁性热疗和化疗的智能复合支架.交变磁场(AMF)的照射不仅可以提高磁性热疗的支架温度,而且可以触发化疗的Dox释放。通过打开和关闭AMF,使磁热和化疗的两个功能同步。通过体外细胞培养和体内动物实验证实了复合支架的协同抗癌作用。复合支架能在AMF照射下有效清除乳腺癌细胞。此外,该支架可以支持间充质干细胞增殖和成脂分化,用于抗癌治疗后的脂肪组织重建。体内再生实验表明,复合支架能有效保持其结构完整性,促进支架内正常细胞的浸润和增殖。复合支架具有多功能,是一种新型的乳腺癌治疗平台。
    Combination of different therapies is an attractive approach for cancer therapy. However, it is a challenge to synchronize different therapies for maximization of therapeutic effects. In this work, a smart composite scaffold that could synchronize magnetic hyperthermia and chemotherapy was prepared by hybridization of magnetic Fe3O4 nanoparticles and doxorubicin (Dox)-loaded thermosensitive liposomes with biodegradable polymers. Irradiation of alternating magnetic field (AMF) could not only increase the scaffold temperature for magnetic hyperthermia but also trigger the release of Dox for chemotherapy. The two functions of magnetic hyperthermia and chemotherapy were synchronized by switching AMF on and off. The synergistic anticancer effects of the composite scaffold were confirmed by in vitro cell culture and in vivo animal experiments. The composite scaffold could efficiently eliminate breast cancer cells under AMF irradiation. Moreover, the scaffold could support proliferation and adipogenic differentiation of mesenchymal stem cells for adipose tissue reconstruction after anticancer treatment. In vivo regeneration experiments showed that the composite scaffolds could effectively maintain their structural integrity and facilitate the infiltration and proliferation of normal cells within the scaffolds. The composite scaffold possesses multi-functions and is attractive as a novel platform for efficient breast cancer therapy.
    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
    热敏脂质体与局部轻度热疗相结合可以改善药物向实体瘤部位的递送。出于这个原因,已经设计了一系列化疗药物的热敏脂质体制剂。我们的小组先前开发并表征了热休克蛋白90抑制剂alvespimycin的热敏脂质体制剂,作为与ThermoDox相当的热敏脂质体制剂的伴随治疗剂(即,ThermoDXR),目的是提高阿霉素的治疗指数,因为该组合在一组包括MDA-MB-231在内的人类乳腺癌细胞系中具有高度协同作用(Dunne等人。,2019)。本文提供的数据进一步描述了多柔比星(DXR)和阿维维霉素(ALV)组合在体外和体内的作用。具体来说,在小鼠乳腺癌4T1细胞中的联合作用以及这种热激活化疗组合在免疫功能低下(MDA-MB-231荷瘤雌性SCID小鼠)和免疫功能低下(4T1荷瘤雌性BALB/c小鼠)乳腺癌模型中的体内功效。
    Thermosensitive liposomes in combination with localized mild hyperthermia can improve the delivery of drug to solid tumor sites. For this reason, thermosensitive liposome formulations of a range of chemotherapy drugs have been designed. Our group previously developed and characterized a thermosensitive liposome formulation of the heat shock protein 90 inhibitor alvespimycin as a companion therapeutic to a thermosensitive liposome formulation equivalent in composition to ThermoDox (i.e., ThermoDXR), with the goal of increasing the therapeutic index of doxorubicin as the combination was revealed to be highly synergistic in a panel of human breast cancer cell lines including MDA-MB-231 (Dunne et al., 2019). The data presented here further describes the effect of the doxorubicin (DXR) and alvespimycin (ALV) combination in vitro and in vivo. Specifically, the combination effect in mouse breast cancer 4T1 cells and the in vivo efficacy of this heat-activated chemotherapy combination in both immunocompromised (MDA-MB-231 tumor bearing female SCID mice) and immunocompetent (4T1 tumor bearing female BALB/c mice) models of breast cancer.
    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
    双去甲氧基姜黄素(BDMC)是从姜科植物中分离出的主要活性成分,其中它具有优异的抗肿瘤作用。然而,不溶于水限制了其临床应用。在这里,我们报道了一种微流控芯片装置,可以将BDMC加载到脂质双层中,形成BDMC热敏脂质体(BDMCTSL)。选择天然活性成分甘草甜素作为表面活性剂,以提高BDMC的溶解度。BDMCTSL的颗粒尺寸小,均匀的尺寸分布,并增强了体外培养释放。通过3-(4,5-二甲基噻唑-2-基)-2,5-二苯基四唑溴化物方法研究了BDMCTSL对人肝细胞癌的抗肿瘤作用,活/死染色,和流式细胞术。这些结果表明,所配制的脂质体具有很强的癌细胞抑制作用,并呈现剂量依赖性的迁移抑制作用。进一步的机制研究表明,BDMCTSL联合轻度局部热疗可显著上调B细胞淋巴瘤2相关X蛋白水平,降低B细胞淋巴瘤2蛋白水平,从而诱导细胞凋亡。通过微流体设备制造的BDMCTSL在轻度局部高温下分解,能有效增强原料不溶性物质的抗肿瘤作用,促进脂质体的翻译。
    Bisdemethoxycurcumin (BDMC) is the main active ingredient that is isolated from Zingiberaceae plants, wherein it has excellent anti-tumor effects. However, insolubility in water limits its clinical application. Herein, we reported a microfluidic chip device that can load BDMC into the lipid bilayer to form BDMC thermosensitive liposome (BDMC TSL). The natural active ingredient glycyrrhizin was selected as the surfactant to improve solubility of BDMC. Particles of BDMC TSL had small size, homogenous size distribution, and enhanced cultimulative release in vitro. The anti-tumor effect of BDMC TSL on human hepatocellular carcinomas was investigated via 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method, live/dead staining, and flowcytometry. These results showed that the formulated liposome had a strong cancer cell inhibitory, and presented a dose-dependent inhibitory effect on migration. Further mechanistic studies showed that BDMC TSL combined with mild local hyperthermia could significantly upregulate B cell lymphoma 2 associated X protein levels and decrease B cell lymphoma 2 protein levels, thereby inducing cell apoptosis. The BDMC TSL that was fabricated via microfluidic device were decomposed under mild local hyperthermia, which could beneficially enhance the anti-tumor effect of raw insoluble materials and promote translation of liposome.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • DOI:
    文章类型: Journal Article
    温度敏感性脂质体制剂是改善药物治疗指数且毒性最小的有前途的工具。这项研究的目的是研究伴随递送含有热敏脂质体(TSLs)的顺铂(Cis)和多柔比星(Dox)的潜力,并在体外和体内进行轻度高温抗癌。聚乙二醇涂层DPPC/DSPC,热敏和DSPC,制备并表征掺入Cis和Dox的非热敏脂质体。采用常规差示扫描量热法(DSC)和傅里叶变换红外光谱(FT-IR)技术研究了药物-磷脂的相互作用和相容性。在高温条件下,在苯并[a]芘(BaP)诱导的纤维肉瘤中评估了这些制剂的化疗功效。测得所制备的热敏脂质体的尺寸直径为120±10nm。DSC数据显示DSPC+Dox和DSPC+Cis曲线的变化,同时比较纯DSPC和药物。然而,FITR分别和混合物中的磷脂和药物谱相同。数据显示Cis-Dox-TSL的更高效力,因为在高热条件下的该组动物中记录到84%的肿瘤生长抑制。卡普兰-梅尔曲线显示,高温下Cis-Dox-TSL和无高温下Cis-Dox-NTSL处理组的动物存活率分别为100%和80%,分别。然而,Cis-TSL和Dox-TSL表现出50%的存活率,而用Dox-NTSL和Cis-NTSL治疗的动物组仅记录了20%的存活率。流式细胞术分析显示Cis-Dox-NTSL增强了肿瘤细胞凋亡的诱导,记录为18%。不出所料,Cis-Dox-TSL显示出巨大的潜力,因为39%的细胞被测量为凋亡细胞,与Cis-Dox-NTSL相比,Dox-TSL和Cis-TSL。通过流式细胞术进行的细胞凋亡分析清楚地表明在施用Cis-Dox-TSL制剂时治疗期间热疗的影响。最后,通过共聚焦显微镜对肿瘤组织进行的免疫组织化学分析显示,在Sham-NTSL和Sham-TSL的载体治疗的动物中,pAkt的表达增加了几倍。然而,Cis-Dox-TSL在Akt的表达中表现出极大的降低,下降了11倍。本研究的结果指导了在高温条件下伴随递送阿霉素和顺铂的热敏脂质体在开发治疗癌症的新型治疗策略中的作用。
    The temperature sensitive liposomal formulations are a promising tool to improve the therapeutic index of the drugs with minimal toxicity. The aim of this study was to investigate the potential of concomitant delivery of cisplatin (Cis) and doxorubicin (Dox) containing thermosensitive liposomes (TSLs) with mild hyperthermia against cancer in vitro and in vivo. The polyethylene glycol coated DPPC/DSPC, thermosensitive and DSPC, non-thermosensitive liposomes incorporating Cis and Dox were prepared and characterized. A conventional Differential Scanning Calorimetry (DSC) technique and Fourier Transform Infrared Spectroscopy (FT-IR) were applied to study drug-phospholipid interaction and compatibility. The chemotherapeutic efficacy of these formulations was evaluated in benzo[a]pyrene (BaP) induced fibrosarcoma under hyperthermic condition. The size diameter of prepared thermosensitive liposomes was measured to be 120 ± 10 nm. The DSC data exhibited the changes in the curves of DSPC + Dox and DSPC + Cis while comparing the pure DSPC and drugs. However, the FITR showed same spectrum of phospholipids and drugs individually and in the mixture as well. The data showed higher efficacy of Cis-Dox-TSL as 84% inhibition in tumor growth was recorded in this group of animals in hyperthermic condition. The Kaplan-Meir curve revealed, 100% and 80% survival of the animals in the groups treated with Cis-Dox-TSL under hyperthermia and Cis-Dox-NTSL without hyperthermia, respectively. However, Cis-TSL as well as Dox-TSL exhibited 50% survival, while only 20% survival was recorded in the groups of animals treated with Dox-NTSL and Cis-NTSL. The flow cytometry analysis revealed that Cis-Dox-NTSL augments the induction of apoptosis in the tumor cells which was recorded as 18%. As expected, Cis-Dox-TSL showed great potential as 39% of cells were measured as apoptotic cells, significantly very high in comparison to Cis-Dox-NTSL, Dox-TSL and Cis-TSL as well. The apoptotic analysis of the cells by flow cytometry clearly indicated the effect of hyperthermia during the treatment while Cis-Dox-TSL formulation was administered. Finally, the immunohistochemical analysis of the tumor tissues by confocal microscopy exhibited several fold increases in the expression of pAkt in the animals treated with vehicles in Sham-NTSL as well as Sham-TSL. However, Cis-Dox-TSL showed great reduction in the expression of Akt, as it declined by 11-fold. The results of the present study directed the role of concomitant delivery doxorubicin and cisplatin containing thermosensitive liposomes under hyperthermic conditions for the development of a novel therapeutic strategy for the treatment of cancer.
    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
    慢性骨髓炎是由影响骨膜的细菌感染引起的炎症性骨骼疾病,骨头,还有骨髓.耐甲氧西林金黄色葡萄球菌(MRSA)是最常见的病原体。在坏死骨上形成的细菌生物膜对于治疗MRSA感染的骨髓炎是相当大的挑战。这里,我们开发了一种多合一阳离子热敏纳米治疗剂(TLCA),用于治疗MRSA感染的骨髓炎.制备的TLCA颗粒带正电,尺寸<230nm,这使得它们能够有效地扩散到生物膜中。纳米治疗剂的正电荷准确地靶向生物膜,并随后在近红外(NIR)光照射下调节药物释放,从而有效地发挥NIR光驱动光热灭菌和化疗的协同作用。超过80%的抗生素在50°C时突然释放,使生物膜分散高达90%。当应用于MRSA感染的骨髓炎时,在808nm激光照射引起的50°C的局部温度下,它不仅消除了细菌,控制了感染,而且抑制了骨组织的炎症反应,显著降低TNF-α,IL-1β,和IL-6水平。总之,我们构建了一体化抗菌治疗模式,为慢性骨髓炎的局部治疗提供了新的有效策略.
    Chronic osteomyelitis is an inflammatory skeletal disease caused by a bacterial infection that affects the periosteum, bone, and bone marrow. Methicillin-resistant Staphylococcus aureus (MRSA) is the most common causative agent. The bacterial biofilm formed on the necrotic bone is a considerable challenge to treating MRSA-infected osteomyelitis. Here, we developed an all-in-one cationic thermosensitive nanotherapeutic (TLCA) for treating MRSA-infected osteomyelitis. The prepared TLCA particles were positively charged and <230 nm in size, which allowed them to diffuse effectively into the biofilm. The positive charges of the nanotherapeutic accurately targeted the biofilm, and it subsequently regulated the drug release under near-infrared (NIR) light irradiation, thereby efficiently exerting the synergistic effect of NIR light-driven photothermal sterilization and chemotherapy. More than 80% of the antibiotics were abruptly released at 50 °C, which dispersed the biofilm by up to 90%. When applied to MRSA-infected osteomyelitis, with a localized temperature of 50 °C induced by 808 nm laser irradiation, it not only eliminated the bacteria and controlled infection but also inhibited the bone tissue inflammatory response, significantly reducing TNF-α, IL-1β, and IL-6 levels. In conclusion, we constructed an all-in-one antimicrobial treatment modality that provides a new and effective strategy for the topical treatment of chronic osteomyelitis.
    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
    俄歇电子(AE)是由诸如I-125(125I)的放射性核素发射的非常低能量的电子。该能量在较小的距离(<0.5μm)上沉积,导致高线性能量转移,对癌细胞造成致命损害是有效的。因此,发射AE的放射治疗剂具有用于癌症治疗的巨大潜力。在这项研究中,开发了包封125I标记的多柔比星(DOX)衍生物的热敏脂质体(TSLs),用于俄歇电子疗法,靶向癌细胞的DNA.放射性碘化的DOX衍生物[125I]5在癌细胞的细胞核中高度积累,并通过AE显示出对结肠26癌细胞的有效细胞毒性。随后,[125I]5以高封装效率加载到TSL中。通过加热实现了从TSL中有效释放[125I]5,而在没有加热的情况下观察到释放减少。此外,封装[125I]5的TSLs在42°C持续1小时在细胞核中显示出高摄取。我们认为[125I]5通过在42°C加热而释放并积累在细胞的细胞核中。这些结果表明封装[125I]5的TSL和热疗的组合是有效的癌症疗法。
    Auger electrons (AEs) are very low-energy electrons emitted by radionuclides such as I-125 (125I). This energy is deposited across a small distance (<0.5 μm), resulting in high linear energy transfer that is potent for causing lethal damage to cancer cells. Thus, AE-emitting radiotherapeutic agents have great potential for cancer treatment. In this study, thermosensitive liposomes (TSLs) encapsulating 125I-labeled doxorubicin (DOX) derivatives were developed for Auger electron therapy, targeting the DNA of cancer cells. A radioiodinated DOX derivative [125I]5 highly accumulated in the nuclei of cancer cells and showed potent cytotoxicity against Colon 26 cancer cells by AEs. Subsequently, [125I]5 was loaded into the TSLs with high encapsulation efficiency. Potent release of [125I]5 from TSLs was achieved with heating, whereas a decreased release was observed without heating. Furthermore, TSLs encapsulating [125I]5 showed a high uptake in the nuclei at 42 °C for 1 h. We supposed that [125I]5 was released by heating at 42 °C and accumulated in the nuclei in the cells. These results suggest that the combination of TSLs encapsulating [125I]5 and hyperthermia is an effective cancer therapy.
    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
    热敏脂质体(TSL)是响应热疗而释放包封的药物的触发纳米颗粒。结合局部热疗,TSL使局部区域药物递送到具有降低的全身毒性的肿瘤。最近的TSL制剂基于血管内触发释放,其中药物释放发生在微脉管系统内。因此,这种交付策略不需要增强的渗透性和保留(EPR)。与基于EPR的传统纳米颗粒药物递送系统相比,具有被动或主动肿瘤靶向(通常为5%ID/g肿瘤),TSL可以取得优越的肿瘤药物摄取(10%ID/g肿瘤)。在过去的四十年中,在临床前和临床研究中,许多TSL制剂已经与各种药物和热疗装置组合。这里,我们回顾了TSL的特性如何决定了药物的释放,并讨论了TSL快速释放药物的优势。我们展示了通过组织快速提取选择药物的好处,细胞吸收迅速。此外,回顾了热疗设备的最佳特性,并讨论了肿瘤生物学和癌细胞特性的影响。因此,这篇综述提供了如何通过优化TSL的组合来改善TSL给药的指南,药物,和热疗方法。所讨论的许多概念适用于各种其他触发的药物递送系统。
    Thermosensitive liposomes (TSL) are triggered nanoparticles that release the encapsulated drug in response to hyperthermia. Combined with localized hyperthermia, TSL enabled loco-regional drug delivery to tumors with reduced systemic toxicities. More recent TSL formulations are based on intravascular triggered release, where drug release occurs within the microvasculature. Thus, this delivery strategy does not require enhanced permeability and retention (EPR). Compared to traditional nanoparticle drug delivery systems based on EPR with passive or active tumor targeting (typically <5%ID/g tumor), TSL can achieve superior tumor drug uptake (>10%ID/g tumor). Numerous TSL formulations have been combined with various drugs and hyperthermia devices in preclinical and clinical studies over the last four decades. Here, we review how the properties of TSL dictate delivery and discuss the advantages of rapid drug release from TSL. We show the benefits of selecting a drug with rapid extraction by tissue, and with quick cellular uptake. Furthermore, the optimal characteristics of hyperthermia devices are reviewed, and impact of tumor biology and cancer cell characteristics are discussed. Thus, this review provides guidelines on how to improve drug delivery with TSL by optimizing the combination of TSL, drug, and hyperthermia method. Many of the concepts discussed are applicable to a variety of other triggered drug delivery systems.
    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
    Chemotherapy plays an important role in debulking tumors in advance of surgery and/or radiotherapy, tackling residual disease, and treating metastatic disease. In recent years many promising advanced drug delivery strategies have emerged that offer more targeted delivery approaches to chemotherapy treatment. For example, thermosensitive liposome-mediated drug delivery in combination with localized mild hyperthermia can increase local drug concentrations resulting in a reduction in systemic toxicity and an improvement in local disease control. However, the majority of solid tumor-associated deaths are due to metastatic spread. A therapeutic approach focused on a localized target area harbors the risk of overlooking and undertreating potential metastatic spread. Previous studies reported systemic, albeit limited, anti-tumor effects following treatment with thermosensitive liposomal chemotherapy and localized mild hyperthermia. This work explores the systemic treatment capabilities of a thermosensitive liposome formulation of the vinca alkaloid vinorelbine in combination with mild hyperthermia in an immunocompetent murine model of rhabdomyosarcoma. This treatment approach was found to be highly effective at heated, primary tumor sites. However, it demonstrated limited anti-tumor effects in secondary, distant tumors. As a result, the addition of immune checkpoint inhibition therapy was pursued to further enhance the systemic anti-tumor effect of this treatment approach. Once combined with immune checkpoint inhibition therapy, a significant improvement in systemic treatment capability was achieved. We believe this is one of the first studies to demonstrate that a triple combination of thermosensitive liposomes, localized mild hyperthermia, and immune checkpoint inhibition therapy can enhance the systemic treatment capabilities of thermosensitive liposomes.
    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