Photodynamic Therapy

光动力疗法
  • 文章类型: Journal Article
    光动力疗法(PDT)与化学动力疗法(CDT)相结合已被证明是提高癌症治疗效率的有希望的策略。因为两种模式之间产生的协同治疗作用。在这里,我们报道了一种基于三元NiCoTi-层状双氢氧化物(NiCoTi-LDH)纳米片的无机纳米剂,可实现高效的光动力/化学动力协同治疗。NiCoTi-LDH纳米片在酸性环境下表现出氧空位促进的电子-空穴分离和光生空穴诱导的不依赖O2的活性氧(ROS)的产生,实现原位pH响应性PDT。此外,由于光生电子引起的Co3和Co2之间的有效转换,NiCoTi-LDH纳米片通过Fenton反应催化H2O2释放羟基自由基(·OH),导致CDT。激光辐照增强了NiCoTi-LDH纳米片的催化能力,以促进ROS的产生,导致在pH6.5时比TiO2纳米颗粒更好的性能。体外和体内实验结果最终表明,NiCoTi-LDH纳米片加上辐射导致有效的细胞凋亡和对肿瘤生长的显着抑制。这项研究报告了一种新型的pH响应无机纳米试剂,具有氧空位促进的光动力/化学动力协同性能,为选择性消除肿瘤提供了一种潜在的有吸引力的临床策略。
    Combining photodynamic therapy (PDT) with chemodynamic therapy (CDT) has been proven to be a promising strategy to improve the treatment efficiency of cancer, because of the synergistic therapeutic effect arising between the two modalities. Herein, we report an inorganic nanoagent based on ternary NiCoTi-layered double hydroxide (NiCoTi-LDH) nanosheets to realize highly efficient photodynamic/chemodynamic synergistic therapy. The NiCoTi-LDH nanosheets exhibit oxygen vacancy-promoted electron-hole separation and photogenerated hole-induced O2-independent reactive oxygen species (ROS) generation under acidic circumstances, realizing in situ pH-responsive PDT. Moreover, due to the effective conversion between Co3+ and Co2+ caused by photogenerated electrons, the NiCoTi-LDH nanosheets catalyze the release of hydroxyl radicals (·OH) from H2O2 through Fenton reactions, resulting in CDT. Laser irradiation enhances the catalyzed ability of the NiCoTi-LDH nanosheets to promote the ROS generation, resulting in a better performance than TiO2 nanoparticles at pH 6.5. In vitro and in vivo experimental results show conclusively that NiCoTi-LDH nanosheets plus irradiation lead to efficient cell apoptosis and significant inhibition of tumor growth. This study reports a new pH-responsive inorganic nanoagent with oxygen vacancy-promoted photodynamic/chemodynamic synergistic performance, offering a potentially appealing clinical strategy for selective tumor elimination.
    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
    理想的肿瘤治疗策略包括可以增强肿瘤微环境的免疫原性同时消除原发性肿瘤的治疗方法。胆酸修饰的铱(III)(Ir3)光敏剂,靶向内质网(ER),据报道,对三阴性乳腺癌(MDA-MB-231)具有有效的I型和II型光动力治疗作用。该光敏剂通过光动力学手段诱导由gasderminE(GSDME)介导的焦化性细胞死亡,并增强肿瘤免疫疗法。机制研究表明,复合物Ir3在光照条件下诱导MDA-MB-231乳腺癌细胞中损伤相关分子模式(DAMPs)的特征。这些包括细胞表面钙网蛋白(CRT)外翻,细胞外高迁移率族蛋白1(HMGB1)和ATP释放,伴有内质网应激和活性氧(ROS)增加。因此,复合物Ir3在光照条件下促进树突状细胞成熟和抗原呈递,体内完全激活T细胞依赖性免疫反应,并最终消除远处的肿瘤,同时破坏原发肿瘤。总之,金属配合物介导的免疫调节和靶向干预代表了肿瘤治疗的新方法。这为联合靶向治疗和免疫治疗的发展提供了有效的策略。
    An ideal tumor treatment strategy involves therapeutic approaches that can enhance the immunogenicity of the tumor microenvironment while simultaneously eliminating the primary tumor. A cholic acid-modified iridium(III) (Ir3) photosensitizer, targeted to the endoplasmic reticulum (ER), has been reported to exhibit potent type I and type II photodynamic therapeutic effects against triple-negative breast cancer (MDA-MB-231). This photosensitizer induces pyroptotic cell death mediated by gasdermin E (GSDME) through photodynamic means and enhances tumor immunotherapy. Mechanistic studies have revealed that complex Ir3 induces characteristics of damage-related molecular patterns (DAMPs) in MDA-MB-231 breast cancer cells under light conditions. These include cell-surface calreticulin (CRT) eversion, extracellular high mobility group box 1 (HMGB1) and ATP release, accompanied by ER stress and increased reactive oxygen species (ROS). Consequently, complex Ir3 promotes dendritic cell maturation and antigen presentation under light conditions, fully activates T cell-dependent immune response in vivo, and ultimately eliminates distant tumors while destroying primary tumors. In conclusion, immune regulation and targeted intervention mediated by metal complexes represent a new and promising approach to tumor therapy. This provides an effective strategy for the development of combined targeted therapy and 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
    治疗诱导的免疫原性细胞死亡(ICD)和免疫检查点阻断的组合可以提供相互增强的策略来逆转肿瘤的不良免疫原性和免疫逃逸行为。在这项工作中,嵌合肽工程免疫刺激剂(ER-PPB)被制造用于针对转移性肿瘤的内质网(ER)靶向光动力免疫疗法。其中,两亲性嵌合肽(ER-PP)由ER靶向肽FFKDEL组成,亲水PEG8接头和光敏剂原卟啉IX(PpIX),可以与PD-1/PD-L1阻断剂(BMS-1)组装以制备ER-PPB。被动靶向肿瘤组织后,ER-PPB将选择性地积累在ER中。接下来,ER-PPB的局部PDT会产生大量的ROS破坏原发肿瘤细胞,同时增加ER应力以启动强大的ICD级联。此外,BMS-1的伴随递送可以通过PD-1/PD-L1阻断阻止肿瘤细胞的免疫逃逸,从而协同激活免疫系统来对抗转移性肿瘤。体外和体内结果表明ER-PPB具有强大的免疫激活和转移性肿瘤抑制特征,这可能为时空控制的转移性肿瘤治疗提供有希望的策略。
    The combination of therapy-induced immunogenic cell death (ICD) and immune checkpoint blockade can provide a mutually reinforced strategy to reverse the poor immunogenicity and immune escape behavior of tumors. In this work, a chimeric peptide-engineered immunostimulant (ER-PPB) is fabricated for endoplasmic reticulum (ER)-targeted photodynamic immunotherapy against metastatic tumors. Among which, the amphiphilic chimeric peptide (ER-PP) is composed of ER-targeting peptide FFKDEL, hydrophilic PEG8 linker and photosensitizer protoporphyrin IX (PpIX), which could be assembled with a PD-1/PD-L1 blocker (BMS-1) to prepare ER-PPB. After passively targeting at tumor tissues, ER-PPB will selectively accumulate in the ER. Next, the localized PDT of ER-PPB will produce a lot of ROS to destroy the primary tumor cells, while increasing the ER stress to initiate a robust ICD cascade. Moreover, the concomitant delivery of BMS-1 can impede the immune escape of tumor cells through PD-1/PD-L1 blockade, thus synergistically activating the immune system to combat metastatic tumors. In vitro and in vivo results demonstrate the robust immune activation and metastatic tumor inhibition characteristics of ER-PPB, which may offer a promising strategy for spatiotemporally controlled metastatic tumor 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
    光动力疗法(PDT)是一种完善的治疗方式,通常用单波长照射进行,对于不同的肿瘤位置和大小,这可能并不总是最佳的。为了解决这个问题,正在探索吸收波长为550至760nm的光敏剂。在这里,合成了一系列5,15-二芳基四苯并卟啉(Ar2TBP)。所有化合物在550-700nm(尤其是在〜668nm)显示出明显的吸收,强烈的荧光,单线态氧的高效生成和良好的光动力抗肿瘤作用。值得注意的是,化合物I3(5,15-双[(4-羧基甲氧基)苯基]四苯并卟啉)在红光照射下对Eca-109细胞系显示出优异的细胞毒性,IC50值为0.45μM,光疗指数为25.8。流式细胞仪显示,I3可以诱导明显的细胞凋亡。体内研究表明,化合物I3在单次给药和光照射下,在肿瘤部位选择性积累并表现出突出的PDT效应,具有抗肿瘤活性。并显示出比临床光敏剂Verteporfin更高的效率。这些发现强调了I3作为一种强大的治疗药物的巨大前景,提供实时荧光成像功能,并作为一种有效的光敏剂,用于肿瘤的个性化和精确的光动力治疗。
    Photodynamic therapy (PDT) is a well-established treatment modality, typically conducted with single-wavelength irradiation, which may not always be optimal for varying tumor locations and sizes. To address this, photosensitizers with absorption wavelengths ranging from 550 to 760 nm are being explored. Herein, a series of 5,15-diaryltetrabenzoporphyrins (Ar2TBPs) were synthesized. All compounds displayed obvious absorption at 550-700 nm (especially at ∼668 nm), intense fluorescence, efficient generation of singlet oxygen and good photodynamic antitumor effects. Notably, compound I3 (5,15-bis[(4-carboxymethoxy)phenyl]tetrabenzoporphyrin) showed excellent cytotoxicity against Eca-109 cell line upon red light irradiation, with an IC50 value of 0.45 μM, and phototherapeutic index of 25.8. Flow cytometry revealed that I3 could induce distinct cell apoptosis. In vivo studies revealed that compound I3 selectively accumulated at tumor site and exhibited outstanding PDT effect with antitumor activity under single-time administration and light irradiation, and revealed more efficiency than the clinical photosensitizer Verteporfin. These findings underscore the considerable promise of I3 as a robust theranostic agent, offering capabilities in real-time fluorescence imaging and serving as a potent photosensitizer for personalized and precise photodynamic therapy of tumors.
    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
    目的:本研究旨在分析一种新的手术模式联合光动力疗法治疗复发性呼吸性乳头状瘤病(RRP)的安全性和有效性。
    方法:回顾南京明基医疗中心选择综合手术联合光动力治疗的RRP患者的病例资料,从2021年1月到2023年5月。通过比较手术前后的年度手术次数和Derkay评分来评估该计划的有效性。
    结果:共23例RRP患者纳入研究。治疗后,复发率为65.2%(15/23),平均复发时间94.3±50.8天。复发时的平均Derkay评分明显低于治疗前的平均Derkay评分(P<0.001)。治疗前平均年复发率为2.2±1.3,治疗后为1.5±1.5,差异无统计学意义(P=0.16)。然而,亚组分析显示,治疗后成人发病RRP的年复发率显著降低(P=0.01).最常见的不良反应为轻度咽痛(11/23)。新发声带粘连3例。无患者出现严重呼吸道相关不良反应,麻醉相关不良反应,或全身性光毒性反应.
    结论:结论:这项研究表明,手术联合光动力疗法(PDT)可能是治疗RRP的一种安全有效的选择,尤其是成人复发呼吸道乳头状瘤病(AORRP)患者。
    OBJECTIVE: This study aims to analyze the safety and effectiveness of a new model of surgery combined with Photodynamic therapy for treating Recurrent Respiratory Papillomatosis (RRP).
    METHODS: Review the case data of patients with RRP who opted for comprehensive surgery combined with Photodynamic therapy at the Nanjing BenQ Medical Center, from January 2021 to May 2023. The efficacy of this program was evaluated by comparing the annual number of surgeries and Derkay scores before and after the surgery.
    RESULTS: A total of 23 RRP patients were included in the study. After treatment, the recurrence rate was 65.2 % (15/23), with an average recurrence time of 94.3 ± 50.8 days. The average Derkay score at the time of recurrence was significantly lower than the average pre-treatment Derkay score (P < 0.001). The average annual recurrence rate before treatment was 2.2 ± 1.3, compared to 1.5 ± 1.5 after treatment, with no significant difference (P = 0.16). However, subgroup analysis revealed a significant decrease in the annual recurrence rate of adult-onset RRP after treatment (P = 0.01). The most common adverse reaction was mild pharyngeal pain (11/23). There were 3 cases of new-onset vocal cord adhesions. No patients experienced serious respiratory-related adverse reactions, anesthesia-related adverse reactions, or systemic phototoxic reactions.
    CONCLUSIONS: In conclusion, this study indicates that surgery combined with Photodynamic therapy (PDT) might be a safe and effective option for treating RRP, especially in patients with Adult-Onset Recurrent Respiratory Papillomatosis (AORRP).
    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:肝细胞癌(HCC)是一种以缺氧环境而闻名的高度恶性肿瘤,这有助于对抗癌药物索拉非尼(SF)的抗性。解决HCC中的SF耐药性需要创新的策略来改善肿瘤氧合并有效地提供治疗。
    结果:在我们的研究中,我们探讨了KPNA4在介导缺氧诱导的肝癌SF抵抗中的作用。我们开发了能够携带氧气的血红蛋白纳米簇(Hb-NC),负载吲哚菁绿(ICG)和SF,名为HPRG@SF。体外,HPRG@SF靶向肝癌细胞,缓解缺氧,抑制KPNA4表达,并增强了PDT对缺氧的细胞毒性,SF抗性HCC细胞。体内实验支持这些发现,表明HPRG@SF有效改善了肿瘤微环境内的氧合,并通过联合光动力疗法(PDT)抵抗了SF抵抗。
    结论:Hb-NC与ICG和SF的组合,形成HPRG@SF,提出了通过改善缺氧和使用PDT来克服肝细胞癌耐药性的有效策略。这种方法不仅针对耐药性背后的低氧条件,而且还提供了协同抗癌作用。强调其临床应用于治疗耐药肝癌的潜力。
    BACKGROUND: Hepatocellular carcinoma (HCC) is a highly malignant tumor known for its hypoxic environment, which contributes to resistance against the anticancer drug Sorafenib (SF). Addressing SF resistance in HCC requires innovative strategies to improve tumor oxygenation and effectively deliver therapeutics.
    RESULTS: In our study, we explored the role of KPNA4 in mediating hypoxia-induced SF resistance in HCC. We developed hemoglobin nanoclusters (Hb-NCs) capable of carrying oxygen, loaded with indocyanine green (ICG) and SF, named HPRG@SF. In vitro, HPRG@SF targeted HCC cells, alleviated hypoxia, suppressed KPNA4 expression, and enhanced the cytotoxicity of PDT against hypoxic, SF-resistant HCC cells. In vivo experiments supported these findings, showing that HPRG@SF effectively improved the oxygenation within the tumor microenvironment and countered SF resistance through combined photodynamic therapy (PDT).
    CONCLUSIONS: The combination of Hb-NCs with ICG and SF, forming HPRG@SF, presents a potent strategy to overcome drug resistance in hepatocellular carcinoma by improving hypoxia and employing PDT. This approach not only targets the hypoxic conditions that underlie resistance but also provides a synergistic anticancer effect, highlighting its potential for clinical applications in treating resistant HCC.
    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
    由活性氧(ROS)响应性药物递送系统介导的化疗可以潜在地减轻化疗药物的毒副作用并显着增强其治疗功效。然而,在肿瘤部位实现精确的靶向药物递送和对ROS响应性药物释放的实时控制仍然是一个艰巨的挑战。因此,本研究旨在描述一种具有特定肿瘤靶向能力的ROS反应性药物递送系统,用于在荧光(FL)和磁共振(MR)双峰成像指导下减轻化疗诱导的毒性,同时增强治疗效果.
    吲哚菁绿(ICG),阿霉素(DOX)前药pB-DOX和超顺磁性氧化铁(SPIO,通过双乳化方法将Fe3O4)包封在聚(乳酸-羟基乙酸共聚物)(PLGA)中,以制备ICG/pB-DOX/Fe3O4/PLGA纳米颗粒(IBFPNPs)。通过碳二亚胺方法用乳腺珠蛋白抗体(mAb)官能化IBFPNP的表面以构建靶向乳腺癌的mAb/IBFPNP(MIBFPNP)。此后,在体外和体内评估了MIBFPNP的FL和MR双峰成像能力。最后,研究了基于MIBFPNP的联合光动力疗法(PDT)和化疗疗效评估。
    多功能MIBFPNP对乳腺癌表现出显著的靶向功效。FL和MR双峰成像清楚地显示了体内靶向MIBFPNPs的分布。在近红外激光照射下,加载ICG的MIBFPNP有效地生成了PDT的ROS,实现精确的肿瘤消融。同时,它通过切割其敏感部分来触发pB-DOX的激活,从而恢复DOX活性并实现ROS反应性靶向化疗。此外,MIBFPNPs联合PDT和化疗提高双峰成像指导下的肿瘤消融效率。
    MIBFPNP构成了一种新型的双模态成像引导药物递送系统,用于靶向治疗乳腺癌,并提供精确和可控的联合治疗选择。
    UNASSIGNED: Chemotherapy mediated by Reactive oxygen species (ROS)-responsive drug delivery systems can potentially mitigate the toxic side effects of chemotherapeutic drugs and significantly enhance their therapeutic efficacy. However, achieving precise targeted drug delivery and real-time control of ROS-responsive drug release at tumor sites remains a formidable challenge. Therefore, this study aimed to describe a ROS-responsive drug delivery system with specific tumor targeting capabilities for mitigating chemotherapy-induced toxicity while enhancing therapeutic efficacy under guidance of Fluorescence (FL) and Magnetic resonance (MR) bimodal imaging.
    UNASSIGNED: Indocyanine green (ICG), Doxorubicin (DOX) prodrug pB-DOX and Superparamagnetic iron oxide (SPIO, Fe3O4) were encapsulated in poly(lactic-co-glycolic acid) (PLGA) by double emulsification method to prepare ICG/ pB-DOX/ Fe3O4/ PLGA nanoparticles (IBFP NPs). The surface of IBFP NPs was functionalized with mammaglobin antibodies (mAbs) by carbodiimide method to construct the breast cancer-targeting mAbs/ IBFP NPs (MIBFP NPs). Thereafter, FL and MR bimodal imaging ability of MIBFP NPs was evaluated in vitro and in vivo. Finally, the combined photodynamic therapy (PDT) and chemotherapy efficacy evaluation based on MIBFP NPs was studied.
    UNASSIGNED: The multifunctional MIBFP NPs exhibited significant targeting efficacy for breast cancer. FL and MR bimodal imaging clearly displayed the distribution of the targeting MIBFP NPs in vivo. Upon near-infrared laser irradiation, the MIBFP NPs loaded with ICG effectively generated ROS for PDT, enabling precise tumor ablation. Simultaneously, it triggered activation of the pB-DOX by cleaving its sensitive moiety, thereby restoring DOX activity and achieving ROS-responsive targeted chemotherapy. Furthermore, the MIBFP NPs combined PDT and chemotherapy to enhance the efficiency of tumor ablation under guidance of bimodal imaging.
    UNASSIGNED: MIBFP NPs constitute a novel dual-modality imaging-guided drug delivery system for targeted breast cancer therapy and offer precise and controlled combined treatment options.
    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
    5-氨基乙酰丙酸(5-ALA)是卟啉IX(PpIX)的前药。5-ALA的缺点包括稳定性差,快速消除,生物利用度差,和微弱的细胞渗透,这大大降低了基于5-ALA的光动力疗法(PDT)的临床效果。目前,使用金纳米颗粒(AuNP)作为载体,通过Au-硫和离子键负载CSNIDARAC(CC9)靶向肽和5-ALA,构建了一种新型靶向纳米系统,分别,然后通过自组装包裹在聚乳酸乙醇酸(PLGA)NPs中,以提高抗肿瘤效果并减少副作用。ALA/CC9@AuNPs-PLGANP的成功制备使用紫外-可见光进行验证,傅里叶变换红外光谱,和X射线光电子能谱。分析显示球形度良好,粒径约为140nm,Zeta电位为10.11mV,和在弱酸性环境中缓慢控制释放的特性。共聚焦显微镜显示,NPs通过主动内化CC9并避免RAW264.7细胞的吞噬作用而靶向NCL-H460细胞,和实时荧光成像揭示了荷瘤小鼠的肿瘤靶向性。与免费的5-ALA相比,纳米系统通过增加PpIX和活性氧的产生以诱导线粒体途径凋亡而显示出增强的抗癌活性。在三维培养的细胞中始终观察到抗肿瘤功效,因为肿瘤球的完整性丧失。在异种移植肿瘤模型中,通过降低的生长速率和增加的肿瘤凋亡证明了更有效的抗肿瘤功效。组织学分析表明,该系统没有毒性,降低5-ALA的肝毒性。因此,ALA/CC9@AuNPs-PLGANPs通过载波级联提供5-ALA,通过被动增强的渗透性和保留作用以及主动靶向对肿瘤积累和PDT具有优异的效果。这种癌症治疗的创新策略在实施之前需要更多的临床试验。
    5-Aminolevulinic acid (5-ALA) is a prodrug of porphyrin IX (PpIX). Disadvantages of 5-ALA include poor stability, rapid elimination, poor bioavailability, and weak cell penetration, which greatly reduce the clinical effect of 5-ALA based photodynamic therapy (PDT). Presently, a novel targeting nanosystem was constructed using gold nanoparticles (AuNPs) as carriers loaded with a CSNIDARAC (CC9)-targeting peptide and 5-ALA via Au-sulphur and ionic bonds, respectively, and then wrapped in polylactic glycolic acid (PLGA) NPs via self-assembly to improve the antitumor effects and reduce the side effect. The successful preparation of ALA/CC9@ AuNPs-PLGA NPs was verified using ultraviolet-visible, Fourier transform infrared spectroscopy, and X-ray photoelectron spectroscopy. The analyses revealed good sphericity with a particle size of approximately140 nm, Zeta potential of 10.11 mV, and slow-controlled release characteristic in a weak acid environment. Confocal microscopy revealed targeting of NCL-H460 cells by NPs by actively internalising CC9 and avoiding the phagocytic action of RAW264.7 cells, and live fluorescence imaging revealed targeting of tumours in tumour-bearing mice. Compared to free 5-ALA, the nanosystem displayed amplified anticancer activity by increasing production of PpIX and reactive oxygen species to induce mitochondrial pathway apoptosis. Antitumor efficacy was consistently observed in three-dimensionally cultured cells as the loss of integrity of tumour balls. More potent anti-tumour efficacy was demonstrated in xenograft tumour models by decreased growth rate and increased tumour apoptosis. Histological analysis showed that this system was not toxic, with lowered liver toxicity of 5-ALA. Thus, ALA/CC9@AuNPs-PLGA NPs deliver 5-ALA via a carrier cascade, with excellent effects on tumour accumulation and PDT through passive enhanced permeability and retention action and active targeting. This innovative strategy for cancer therapy requires more clinical trials before being implemented.
    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
    光热,光动力和声动力癌症治疗提供了精确的肿瘤消融和减少副作用的机会。干扰素基因的环状鸟苷酸腺苷酸合成酶刺激物(cGAS-STING)途径被认为是刺激患者免疫系统并实现持续免疫应答的潜在靶标。结合光热,使用cGAS-STING激动剂的光动力和声动力疗法代表了一种新开发的癌症治疗方法,在其对免疫系统的影响方面表现出明显的创新。最近的综述集中在各种材料及其在癌症治疗中的功能上。在这次审查中,我们专注于光热的分子机制,光动力和声动力癌症治疗以及cGAS-STING激动剂在治疗癌症中的相关作用。
    Photothermal, photodynamic and sonodynamic cancer therapies offer opportunities for precise tumor ablation and reduce side effects. The cyclic guanylate adenylate synthase-stimulator of interferon genes (cGAS-STING) pathway has been considered a potential target to stimulate the immune system in patients and achieve a sustained immune response. Combining photothermal, photodynamic and sonodynamic therapies with cGAS-STING agonists represents a newly developed cancer treatment demonstrating noticeable innovation in its impact on the immune system. Recent reviews have concentrated on diverse materials and their function in cancer therapy. In this review, we focus on the molecular mechanism of photothermal, photodynamic and sonodynamic cancer therapies and the connected role of cGAS-STING agonists in treating 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
    前列腺癌是一种具有挑战性的疾病,因为它通常被描述为免疫学上的“冷”肿瘤,导致临床环境中当前免疫治疗方法的次优结果。光动力疗法(PDT)利用光敏剂(PS)产生的活性氧来破坏细胞内氧化还原平衡。这个过程诱导线粒体和细胞核的DNA损伤,激活免疫原性细胞死亡(ICD)和cGAS-STING途径的过程。最终,这种级联事件导致抗肿瘤免疫反应的启动。然而,现有的PS面临挑战,包括次优的肿瘤靶向,聚集诱导猝灭,肿瘤区域的氧气水平不足。为此,设计了一种通用的仿生纳米平台,用于同时递送聚集诱导的发射PSTPAQ-Py-PF6和紫杉醇(PTX)。纳米平台的细胞膜伪装导致其在肿瘤靶向和细胞内化方面的显着能力。激光照射后,TPAQ-Py-PF6与PTX联合使用显示出明显且增强的协同抗肿瘤作用。此外,纳米平台具有启动cGAS-STING途径的能力,导致细胞因子的产生。由ICD诱导的损伤相关分子模式的存在与这些上述细胞因子协作导致树突状细胞成熟的募集和促进。因此,这引发了针对肿瘤的全身免疫反应。总之,这种有前途的策略突出了多功能仿生纳米平台的使用,联合化疗,PDT,和免疫疗法,以增强抗肿瘤治疗的有效性。
    Prostate cancer presents as a challenging disease, as it is often characterized as an immunologically \"cold\" tumor, leading to suboptimal outcomes with current immunotherapeutic approaches in clinical settings. Photodynamic therapy (PDT) harnesses reactive oxygen species generated by photosensitizers (PSs) to disrupt the intracellular redox equilibrium. This process induces DNA damage in both the mitochondria and nucleus, activating the process of immunogenic cell death (ICD) and the cGAS-STING pathway. Ultimately, this cascade of events leads to the initiation of antitumor immune responses. Nevertheless, existing PSs face challenges, including suboptimal tumor targeting, aggregation-induced quenching, and insufficient oxygen levels in the tumor regions. To this end, a versatile bionic nanoplatform has been designed for the simultaneous delivery of the aggregation-induced emission PS TPAQ-Py-PF6 and paclitaxel (PTX). The cell membrane camouflage of the nanoplatform leads to its remarkable abilities in tumor targeting and cellular internalization. Upon laser irradiation, the utilization of TPAQ-Py-PF6 in conjunction with PTX showcases a notable and enhanced synergistic antitumor impact. Additionally, the nanoplatform has the capability of initiating the cGAS-STING pathway, leading to the generation of cytokines. The presence of damage-associated molecular patterns induced by ICD collaborates with these aforementioned cytokines lead to the recruitment and facilitation of dendritic cell maturation. Consequently, this elicits a systemic immune response against tumors. In summary, this promising strategy highlights the use of a multifunctional biomimetic nanoplatform, combining chemotherapy, PDT, and immunotherapy to enhance the effectiveness of antitumor 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