Synergistic immunotherapy

  • 文章类型: Journal Article
    利用抗PD-L1阻断的免疫疗法在临床乳腺癌治疗中取得了巨大的成功,但经常受到有限的免疫反应的阻碍。越来越多的证据表明,免疫原性细胞死亡(ICD)最近成为扩大肿瘤免疫原性和有效引发系统性抗肿瘤免疫的有希望的策略。然而,开发简单但通用的,高效但低毒,生物安全,和临床上可获得的转化ICD诱导物仍然是巨大的需求并且是高度期望的。在这里,通过整合光热疗法(PTT)纳米平台介孔聚多巴胺(MPDA),有目的地开发了一种多功能ICD诱导剂A6-MPDA@PAL,CDK4/6抑制剂palbociclib(PAL),和CD44特异性靶向A6肽以一种简单的方式增强抗PD-L1治疗的免疫抗肿瘤功效。值得注意的是,光诱导的纳米平台表现出多种有利的治疗特征,确保了优越和生物安全的PTT/化疗疗效.加上更强的累积ICD诱导,A6-MPDA@PAL的单次给药可以通过吸引T淋巴细胞的肿瘤内浸润和同时逆转免疫抑制肿瘤微环境,在抗PD-L1阻断的辅助下,引发强大的全身抗肿瘤免疫和远视效应,治疗实现了辉煌的协同免疫治疗与有效的肿瘤消融。这项研究提出了一种简单而智能的ICD诱导剂,为加强抗PD-L1治疗乳腺癌开辟了有吸引力的临床可能性。
    Immunotherapy utilizing anti-PD-L1 blockade has achieved dramatic success in clinical breast cancer management but is often hampered by the limited immune response. Increasing evidence shows that immunogenic cell death (ICD) recently arises as a promising strategy for enlarging tumor immunogenicity and eliciting systemic anti-tumor immunity effectively. However, developing simple but versatile, highly efficient but low-toxic, biosafe, and clinically available transformed ICD inducers remains a huge demand and is highly desirable. Herein, a multifunctional ICD inducer is purposefully developed A6-MPDA@PAL by integrating photothermal therapy (PTT) nanoplatforms mesoporous polydopamine (MPDA), CDK4/6 inhibitor palbociclib (PAL), and CD44-specific targeting A6 peptide in a simple way for augmenting the immune antitumor efficacy of anti-PD-L1 therapy. Remarkably, the light-inducible nanoplatforms exhibit multiple favorable therapeutic features ensuring a superior and biosafe PTT/chemotherapy efficacy. Together with stronger accumulative ICD induction, single administration of A6-MPDA@PAL can trigger robust systemic antitumor immunity and abscopal effect with the assistance of anti-PD-L1 blockade by fascinating the intratumoral infiltration of T lymphocytes and reversing the immunosuppressive tumor microenvironment simultaneously, therapy achieving brilliant synergistic immunotherapy with effective tumor ablation. This study presents a simple and smart ICD inducer opening up attractive clinical possibilities for reinforcing the anti-PD-L1 therapy against 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
    如今,针对三阴性乳腺癌(TNBC)的有效免疫疗法由于具有免疫抑制性的肿瘤微环境而仍然具有挑战性.免疫检查点抑制剂主要用于恢复肿瘤特异性免疫细胞的活性,然而,由于肿瘤浸润性CD8+T细胞的数量有限和免疫药物向肿瘤组织的低效递送,其治疗效果甚微。为了解决这些问题,我们在此构建了一个定制的溶解微针共封装TLR7/8激动剂R848和免疫检查点抑制剂aPD-1,称为αNP-RNP@DMN,并将其制造为透皮给药系统。这个精心设计的微针贴片,具有高效的肿瘤给药能力,能够成熟肿瘤浸润的树突状细胞(TIDC),并在R848的帮助下进一步促进CD8T细胞向肿瘤组织的浸润。此外,aPD-1的引入阻断了程序性细胞死亡蛋白1/程序性细胞死亡配体1(PD-1/PD-L1)免疫检查点,协同逆转TNBC的免疫抑制微环境。体内治疗结果表明,αNP-RNP@DMN不仅显著延长了4T1荷瘤小鼠的生存时间,还能抑制肿瘤术后复发和肺转移,这意味着这种有效的药物递送系统在增强浅表肿瘤的免疫治疗中的巨大潜力。重要声明:有限数量的肿瘤浸润性CD8+T细胞和免疫药物向肿瘤组织的低效递送阻碍了三阴性乳腺癌(TNBC)的有效免疫治疗。在这里,开发并制造了共封装TLR7/8激动剂R848和免疫检查点抑制剂aPD-1的溶解微针作为透皮给药系统.这种量身定制的微针贴片不仅以安全无痛的方式促进了药物在肿瘤部位的积累,但也解除了肿瘤浸润性树突状细胞(TIDC)的免疫抑制状态。活化的TIDC进一步增强了T细胞向肿瘤组织的浸润,从而成功地提高了aPD-1的疗效。这项研究表明,这种精心设计的微针贴片可以作为一种有效的药物递送系统,用于增强TNBC的免疫治疗。
    Nowadays, effective immunotherapy against triple-negative breast cancer (TNBC) remains challenging due to the immunosuppressive tumor microenvironment. Immune checkpoint inhibitor is mostly employed to restore the activity of tumor-specific immune cells, which however brings little therapeutic outcome owing to the limited number of tumor-infiltrating CD8+ T cells and the inefficient delivery of immune drugs to the tumor tissue. Aiming to solve these problems, we herein constructed a tailor-made dissolving microneedle co-encapsulating the TLR7/8 agonist R848 and the immune checkpoint inhibitor aPD-1, termed αNP-RNP@DMN, and fabricated it as a transdermal drug delivery system. This well-designed microneedle patch, endowed with efficient tumor drug delivery ability, was able to mature tumor-infiltrating dendritic cells (TIDCs) and further promote the infiltration of CD8+ T cells into the tumor tissue with the aid of R848. Moreover, the introduction of aPD-1 blocked the programmed cell death protein 1/programmed cell death ligand 1(PD-1/PD-L1) immune checkpoints, synergistically reversing the immunosuppressive microenvironment of TNBC. In vivo therapeutic results demonstrated that αNP-RNP@DMN not only significantly prolonged the survival time of 4T1 tumor-bearing mice, but also inhibited tumor recurrence and lung metastasis after surgery, implying the great potential of this effective drug delivery system for enhanced immunotherapy of superficial tumors. STATEMENT OF SIGNIFICANCE: The limited number of tumor-infiltrating CD8+ T cells and the inefficient delivery of immune drugs to the tumor tissue hinder the effective immunotherapy of triple-negative breast cancer (TNBC). Herein, a dissolving microneedle co-encapsulating TLR7/8 agonist R848 and immune checkpoint inhibitor aPD-1 was developed and fabricated as a transdermal drug delivery system. This tailor-made microneedle patch not only promoted drug accumulation in tumor sites in a safe and painless manner, but also lifted the immune-suppressive state of tumor-infiltrating dendritic cells (TIDCs). The activated TIDCs further enhanced T-cell infiltration into the tumor tissue, thus successfully boosting the therapeutic efficacy of aPD-1. This study demonstrated that this well-designed microneedle patch could be served as an effective drug delivery system for enhanced immunotherapy of TNBC.
    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
    趋化联合光疗的治疗效果仍然受到容易的铜流出的阻碍,非特异性积累和有限的光穿透深度。这里,首先通过双供体工程合成了高性能的NIR-II半导体聚合物。然后通过Cu(II)介导的NIR-II超小聚合物点与化疗药物DOX的协同自组装制备了仿生角化放大器(PCD@CM),然后伪装肿瘤细胞膜.同源靶向递送至肿瘤细胞后,肿瘤微环境(TME)中过度表达的GSH通过将Cu(II)还原为Cu(I)触发放大器的分解和治疗成分的释放,这使得NIR-II荧光/光声成像引导的NIR-II光热治疗(PTT)和化疗成为可能。释放的Cu(I)诱导脂质化线粒体蛋白的聚集,伴随着铁硫蛋白的损失,导致严重的蛋白毒性应激和最终的角化。NIR-IIPTT和GSH耗竭使肿瘤细胞对角化更敏感。扩增的角化致敏引起显著的免疫监视,触发免疫原性细胞死亡(ICD)以促进细胞毒性T淋巴细胞浸润以及aPD-L1介导的免疫检查点阻断。这项工作提出了一种新的策略,以开发具有同源靶向和抗肿瘤免疫反应能力的NIR-II光疗增强的角化增敏系统。
    The therapeutic efficacy of cuproptosis combined with phototheranostics is still hindered by easy copper efflux, nonspecific accumulation and limited light penetration depth. Here, a high-performance NIR-II semiconductor polymer was first synthesized through dual-donor engineering. Then a biomimetic cuproptosis amplifier (PCD@CM) was prepared by Cu(II)-mediated coordinative self-assembly of NIR-II ultrasmall polymer dots and the chemotherapeutic drug DOX, followed by camouflaging of tumor cell membranes. After homologous targeting delivery to tumor cells, overexpressed GSH in the tumor microenvironment (TME) triggers the disassembly of the amplifier and the release of therapeutic components through the reduction of Cu(II) to Cu(I), which enable NIR-II fluorescence/photoacoustic imaging-guided NIR-II photothermal therapy (PTT) and chemotherapy. The released Cu(I) induces the aggregation of lipoylated mitochondrial proteins accompanied by the loss of iron-sulfur proteins, leading to severe proteotoxic stress and eventually cuproptosis. NIR-II PTT and GSH depletion render tumor cells more sensitive to cuproptosis. The amplified cuproptosis sensitization provokes significant immune surveillance, triggering the immunogenic cell death (ICD) to promote cytotoxic T lymphocyte infiltration together with aPD-L1-mediated immune checkpoint blockade. This work proposes a new strategy to develop cuproptosis sensitization systems enhanced by NIR-II phototheranostics with homologous targeting and anti-tumor immune response capabilities.
    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
    用于癌症免疫疗法的纳米技术干预是具有巨大潜力的快速发展的范例。自组装纳米材料为其不可降解的对应物提供了更安全的替代品,并且在受控药物递送和光疗方面具有更好的功能以激活免疫原性细胞死亡。在这篇评论中,我们讨论了几类基于聚合物的自组装纳米材料,脂质,肽,水凝胶,金属有机框架,和共价有机框架,具有激活全身免疫反应并将“冷”免疫抑制肿瘤块转化为“热”抗肿瘤免疫细胞丰富的微环境的能力。这些材料的独特方面得到了支撑,并讨论了它们的组合免疫治疗作用机制。还强调了与他们的临床翻译相关的未来挑战。
    Nanotechnological interventions for cancer immunotherapy are a rapidly evolving paradigm with immense potential. Self-assembled nanobiomaterials present safer alternatives to their nondegradable counterparts and pose better functionalities in terms of controlled drug delivery and phototherapy to activate immunogenic cell death. In this Review, we discuss several classes of self-assembled nanobiomaterials based on polymers, lipids, peptides, hydrogel, metal organic frameworks, and covalent-organic frameworks with the ability to activate systemic immune response and convert a \"cold\" immunosuppressive tumor mass to a \"hot\" antitumor immune cell rich microenvironment. The unique aspects of these materials are underpinned, and their mechanisms of combinatorial immunotherapeutic action are discussed. Future challenges associated with their clinical translation are also highlighted.
    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
    免疫检查点阻断(ICB)和免疫原性细胞死亡(ICD)的协同免疫疗法在各种癌症中显示出显著的治疗功效。然而,由于程序性死亡-配体1(PD-L1)的再循环机制和诱导ICD的化疗药物的全身毒性,患者对这些联合治疗的应答率和不良结局较低.在这里,我们提出了多合一乙二醇壳聚糖纳米颗粒(CNPs),该纳米颗粒可将抗PD-L1肽(PP)和多柔比星(DOX)递送至靶向肿瘤组织,以实现安全和更有效的协同免疫疗法.PP-CNPs,通过将α-形式PP(NYSKPTDRQYHF)与CNPs缀合制备,形成稳定的纳米颗粒,促进与靶向肿瘤细胞表面PD-L1蛋白的多价结合,与抗PD-L1抗体相比,导致有效的溶酶体PD-L1降解,诱导内吞PD-L1的再循环。因此,PP-CNP阻止CT26结肠荷瘤小鼠亚细胞PD-L1再循环并最终破坏免疫逃逸机制。此外,ICD诱导器,将DOX加载到PP-CNPs(DOX-PP-CNPs)中,用于协同ICD和ICB治疗,在目标肿瘤组织中诱导大量损伤相关分子模式(DAMPs),在正常组织中毒性最小。当DOX-PP-CNPs静脉注射到CT26结肠荷瘤小鼠体内时,PP和DOX通过纳米粒子衍生的被动和主动靶向有效地递送到肿瘤组织,最终诱导溶酶体PD-L1降解和大量ICD,通过强烈的抗肿瘤免疫应答导致肿瘤完全消退的高比率(CR:60%)。总的来说,这项研究证明了使用多合一纳米颗粒将PP和DOX递送至靶向肿瘤组织的协同免疫疗法的优异疗效.
    Synergistic immunotherapy of immune checkpoint blockade (ICB) and immunogenic cell death (ICD) has shown remarkable therapeutic efficacy in various cancers. However, patients show low response rates and undesirable outcomes to these combination therapies owing to the recycling mechanism of programmed death-ligand 1 (PD-L1) and the systemic toxicity of ICD-inducing chemotherapeutic drugs. Herein, we propose all-in-one glycol chitosan nanoparticles (CNPs) that can deliver anti-PD-L1 peptide (PP) and doxorubicin (DOX) to targeted tumor tissues for a safe and more effective synergistic immunotherapy. The PP-CNPs, which are prepared by conjugating ᴅ-form PP (NYSKPTDRQYHF) to CNPs, form stable nanoparticles that promote multivalent binding with PD-L1 proteins on the targeted tumor cell surface, resulting in effective lysosomal PD-L1 degradation in contrast with anti-PD-L1 antibody, which induces recycling of endocytosed PD-L1. Consequently, PP-CNPs prevent subcellular PD-L1 recycling and eventually destruct immune escape mechanism in CT26 colon tumor-bearing mice. Moreover, the ICD inducer, DOX is loaded into PP-CNPs (DOX-PP-CNPs) for synergistic ICD and ICB therapy, inducing a large number of damage-associated molecular patterns (DAMPs) in targeted tumor tissues with minimal toxicity in normal tissues. When the DOX-PP-CNPs are intravenously injected into CT26 colon tumor-bearing mice, PP and DOX are efficiently delivered to the tumor tissues via nanoparticle-derived passive and active targeting, which eventually induce both lysosomal PD-L1 degradation and substantial ICD, resulting in a high rate of complete tumor regression (CR: 60%) by a strong antitumor immune response. Collectively, this study demonstrates the superior efficacy of synergistic immunotherapy using all-in-one nanoparticles to deliver PP and DOX to targeted tumor tissues.
    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
    Cancer immunotherapy has achieved promising clinical progress over the recent years for its potential to treat metastatic tumors and inhibit their recurrences effectively. However, low patient response rates and dose-limiting toxicity remain as major dilemmas for immunotherapy. Stimuli-responsive nanoparticles (srNPs) combined with immunotherapy offer the possibility to amplify anti-tumor immune responses, where the weak acidity, high concentration of glutathione, overexpressions of enzymes, and reactive oxygen species, and external stimuli in tumors act as triggers for controlled drug release. This review highlights the design of srNPs based on tumor microenvironment and/or external stimuli to combine with different anti-tumor drugs, especially the immunoregulatory agents, which eventually realize synergistic immunotherapy of malignant primary or metastatic tumors and acquire a long-term immune memory to prevent tumor recurrence. The authors hope that this review can provide theoretical guidance for the construction and clinical transformation of smart srNPs for controlled drug delivery in synergistic cancer 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
    肿瘤术后复发是导致治疗失败的主要原因。然而,复发的初始阶段不容易发现,后期很难治愈。为了提高术后患者的生活质量,开发了一种有效的协同免疫疗法,以实现对手术后肿瘤复发的早期诊断和治疗,同时。在本文中,制备了两种基于金纳米棒(AuNRs)平台的治疗剂。一种试剂中的AuNRs和量子点(QDs)用于检测癌胚抗原(CEA),使用荧光共振能量转移(FRET)技术来指示原位复发的发生,而另一种药物中的AuNRs用于光热治疗(PTT),与抗PDL1介导的免疫治疗一起缓解肿瘤转移的过程。一系列试验表明,这种协同免疫疗法可以诱导肿瘤细胞死亡和CD3+/CD4+T淋巴细胞和CD3+/CD8+T淋巴细胞的产生增加。此外,与单一免疫疗法相比,协同免疫疗法分泌的免疫因子(IL-2,IL-6和IFN-γ)更多.这种协同的免疫治疗策略可以同时用于肿瘤术后复发的诊断和治疗。为基础和临床研究提供了新的视角。
    Tumor recurrence after surgery is the main cause of treatment failure. However, the initial stage of recurrence is not easy to detect, and it is difficult to cure in the late stage. In order to improve the life quality of postoperative patients, an efficient synergistic immunotherapy was developed to achieve early diagnosis and treatment of post-surgical tumor recurrence, simultaneously. In this paper, two kinds of theranostic agents based on gold nanorods (AuNRs) platform were prepared. AuNRs and quantum dots (QDs) in one agent was used for the detection of carcinoembryonic antigen (CEA), using fluorescence resonance energy transfer (FRET) technology to indicate the occurrence of in situ recurrence, while AuNRs in the other agent was used for photothermal therapy (PTT), together with anti-PDL1 mediated immunotherapy to alleviate the process of tumor metastasis. A series of assays indicated that this synergistic immunotherapy could induce tumor cell death and the increased generation of CD3+/CD4+ T-lymphocytes and CD3+/CD8+ T-lymphocytes. Besides, more immune factors (IL-2, IL-6, and IFN-γ) produced by synergistic immunotherapy were secreted than mono-immunotherapy. This cooperative immunotherapy strategy could be utilized for diagnosis and treatment of postoperative tumor recurrence at the same time, providing a new perspective for basic and clinical research.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    已经开发了多种疗法并用于治疗结肠癌,然而,高死亡率仍然很高,仍然迫切需要更有效的策略。在这项研究中,一种由先天免疫激活剂黄芪甲苷(As)和光动力疗法(PDT)试剂氯e6(Ce6)(((As+Ce6)@MSNs-PEG)组成的免疫治疗方法,是为结肠癌治疗而开发的。我们表明(AsCe6)@MSNs-PEG可以有效激活NK细胞并抑制肿瘤细胞的增殖。它还可以有效地到达肿瘤部位,诱导免疫细胞渗入肿瘤,并增强体内自然杀伤细胞和CD8+T细胞的细胞毒性。没有明显的副作用,(AsCe6)@MSNs-PEG治疗可显着抑制肿瘤生长并延长荷瘤小鼠的寿命。进一步的结果表明,处理(As+Ce6)@MSNs-PEG导致增强的IFN分泌的免疫细胞和增加的T-box转录因子(T-bet),T细胞高度表达。因此,(As+Ce6)@MSNs-PEG可作为与纳米草药药物和PDT联合使用的有效且安全的平台,为结肠癌治疗提供新的疗法。
    A variety of therapies have been developed and used for the treatment of colon cancer, however, the high mortality rate remains high and more effective strategies are still in urgent needs. In this study, an immunotherapy approach that is composed of innate immune activator Astragaloside III (As) and the photodynamic therapy (PDT) reagent chlorine e6 (Ce6) ((As + Ce6)@MSNs-PEG), was developed for colon cancer treatment. We showed that (As + Ce6)@MSNs-PEG could effectively activate NK cells and inhibit the proliferation of tumor cells in vitro. It could also effectively reach tumor sites, induce infiltration of immune cells into the tumor, and enhance the cytotoxicity of natural killer cells and CD8+ T cells in vivo. Without obvious side effects, (As + Ce6)@MSNs-PEG treatment significantly inhibited tumor growth and extended the lifespan of tumor-bearing mice. Further results revealed that treatment of (As + Ce6)@MSNs-PEG led to enhanced IFN secretion by immune cells and increased T-box transcription factor (T-bet), which is highly expressed by T cells. Therefore, (As + Ce6)@MSNs-PEG may serve as an effective and safe platform for combinatory use with nano-herb medicine and PDT to provide a new therapy for colon cancer treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Given that vascular endothelial growth factor (VEGF) and placental growth factor (PIGF), over-expressed in breast cancer cells and M2-like tumor-associated macrophages (M2-TAMs) within tumor microenvironment (TME), work synergistically and independently in mediating tumor progression and immunosuppression, combinatorial immune-based approaches targeting them are expected to be a potent therapeutic modality for patients. Here, polyethylene glycol (PEG) and mannose doubly modified trimethyl chitosan (PEG = MT) along with citraconic anhydride grafted poly (allylamine hydrochloride) (PC)-based nanoparticles (NPs) (PEG = MT/PC NPs) with dual pH-responsiveness were developed to deliver VEGF siRNA (siVEGF)/PIGF siRNA (siPIGF) to both M2-TAMs and breast cancer cells for antitumor immunotherapy. With prolonged blood circulation and intelligent pH-sensitivity, PEG = MT/PC NPs were highly accumulated in tumor tissues and then internalized in M2-TAMs and breast cancer cells via mannose-mediated active targeting and passive targeting, respectively. With the charge-reversal of PC, PEG = MT/PC NPs presented effective endosomal/lysosomal escape and intracellular siRNA release, resulting in efficient gene silencing. Due to the synergism between siVEGF and siPIGF in anti-proliferation of tumor cells and reversal of the TME from pro-oncogenic to anti-tumoral, PEG = MT/PC/siVEGF/siPIGF NPs (PEG = MT/PC/siV-P NPs) exerted robust suppression of breast tumor growth and lung metastasis. This combination strategy may provide a promising alternative for 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
    Patients with glioblastoma multiforme (GBM) exhibit a deficient anti-tumor immune response. Both arms of the immune system were shown to be hampered in GBM, namely the local cellular immunity mediated by the Th1 subset of helper T cells and the systemic humoral immunity mediated by the Th2 subset of helper T cells. Immunotherapy is rapidly becoming one of the pillars of anti-cancer therapy. GBM has not received similar clinical successes as of yet, which may be attributed to its relative inaccessibility (the blood-brain barrier (BBB)), its poor immunogenicity, few characterized cancer antigens, or any of the many other immune mechanisms known to be hampered. Focused ultrasound (FUS) is emerging as a promising treatment approach. The effects of FUS on the tissue are not merely thermal. Mounting evidence suggests that in addition to thermal ablation, FUS induces mechanical acoustic cavitation and immunomodulation plays a key role in boosting the host anti-tumor immune responses. We separately discuss the different pertinent immunosuppressive mechanisms harnessed by GBM and the immunomodulatory effects of FUS. The effect of FUS and microbubbles in disrupting the BBB and introducing antigens and drugs to the tumor milieu is discussed. The FUS-induced pro-inflammatory cytokines secretion and stress response, the FUS-induced change in the intra-tumoral immune-cells populations, the FUS-induced augmentation of dendritic cells activity, and the FUS-induced increased cytotoxic cells potency are all discussed. We next attempt at offering a conceptual synopsis of the synergistic treatment of GBM utilizing FUS and immunotherapy. In conclusion, it is increasingly apparent that no single treatment modality will triumph on GBM. The reviewed FUS-induced immunomodulation effects can be harnessed to current and developing immunotherapy approaches. Together, these may overcome GBM-induced immune-evasion and generate a clinically relevant anti-tumor immune response.
    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