- 无标题文档
查看论文信息

中文题名:

 自催化型纳米反应器诱导细胞焦亡和免疫反应抑制三阴性乳腺癌增殖和术后复发    

姓名:

 朱钰彤    

学号:

 20121213164    

保密级别:

 公开    

论文语种:

 chi    

学科代码:

 0831    

学科名称:

 工学 - 生物医学工程(可授工学、理学、医学学位)    

学生类型:

 硕士    

学位:

 理学硕士    

学校:

 西安电子科技大学    

院系:

 生命科学技术学院    

专业:

 生物医学工程    

研究方向:

 生物医学工程    

第一导师姓名:

 王忠良    

第一导师单位:

  西安电子科技大学    

完成日期:

 2023-05-20    

答辩日期:

 2023-05-27    

外文题名:

 Autocatalytic Nanoreactor Inhibits Triple-Negative Breast Cancer Proliferation and Postoperative Recurrence via Pyroptosis and Immune Response    

中文关键词:

 乳腺癌治疗 ; 细胞焦亡 ; 过氧化铜纳米反应器 ; 术后辅助治疗    

外文关键词:

 Breast cancer therapy ; Pyroptosis ; Copper peroxide nanoreactor ; Postoperative adjuvant therapy    

中文摘要:

三阴性乳腺癌具有恶性程度高、免疫应答弱、五年生存率低的特点,目前临床上手术切除后的主要术后辅助治疗方式是化疗以及放疗,手术切除原位肿瘤后预后差,易出现术后复发与转移,放化疗不仅增加了转移风险,强烈的副作用还会严重影响患者生活质量,急需更好的治疗策略支撑。

细胞焦亡是新发现的一种免疫原性的细胞程序性死亡方式,不仅可以直接杀伤肿瘤细胞还可以激活抗肿瘤免疫反应,因而在三阴性乳腺癌治疗中有着广泛的应用前景。在诱发肿瘤细胞焦亡的多种通路中,caspase-3相关通路由于其诱导细胞凋亡或细胞焦亡的双向性受到广泛关注。Caspase-3是典型的凋亡相关蛋白,当细胞内焦亡执行蛋白GSDME高表达时,胞内激活的caspase-3将切割GSDME并释放其N端结构域在细胞膜上穿孔,导致细胞肿胀、破裂并引发细胞焦亡。因此,如何激活caspase-3并提高肿瘤细胞中GSDME的表达是细胞焦亡依赖性肿瘤治疗的关键问题。本论文设计并合成了过氧化铜纳米反应器,从胞内氧化应激损伤激活caspase-3通路入手,利用临床去甲基化药物地西他滨(Decitabine,DAC/DEC)提高三阴性乳腺癌细胞中GSDME表达量,实现了细胞焦亡与肿瘤免疫应答的激活。具体研究内容及结果如下:

成功制备了可自催化产生过氧化氢(H2O2)的过氧化铜纳米反应器(Copper peroxide nanoreactor,CP),以高效产生活性氧(ROS)并且不受肿瘤处H2O2不足的制约,然后对CP性能进行了详细研究。证明CP在酸性环境/溶酶体中(pH 5.5)能够分解产生Cu2+和H2O2,进而发生类芬顿反应产生大量羟基自由基(•OH),显著提升胞内ROS水平。

通过生物信息学手段分析得出乳腺癌细胞中Gsdme高度甲基化,进一步通过实时荧光定量聚合酶链式反应(Quantitative real-time PCR,q-PCR)证明DAC有效的提高了小鼠三阴性乳腺癌细胞系4T1中Gsdme基因表达量,进而探究了DAC+CP共同给药后的细胞发生细胞焦亡且释放多种标志性因子。在此基础上,本论文利用小鼠三阴性乳腺癌细胞系4T1建立了小鼠皮下瘤模型进行抗肿瘤治疗,结果表明DAC+CP具有较好的体内抗肿瘤效果,直接杀伤肿瘤细胞的同时有效激活了小鼠在体抗肿瘤免疫反应,显著抑制了肿瘤增殖。

三阴性乳腺癌不仅治疗手段有限,手术切除后的高复发率同样威胁着患者的生存。术后复发的主要原因是微小肿瘤病灶和术后免疫抑制微环境促进残余细胞生长,因此本论文设计合成了适合术后局部缓释给药的纳米复合水凝胶CP@Gel,联合DAC在杀伤残余微小肿瘤细胞的同时逆转免疫抑制微环境,激活抗肿瘤免疫,作为三阴性乳腺癌的术后辅助治疗手段。CP@Gel是一种可注射型水凝胶,在体定点注射成胶后水凝胶缓慢解离,释放的CP纳米反应器诱导术后残余肿瘤病灶细胞的caspase-3活化,DAC提高GSDME蛋白表达,进一步触发GSDME依赖性细胞焦亡并实现抗肿瘤免疫。本论文利用带荧光素酶基因的4T1-Luc细胞系构建了小鼠肿瘤不完全切除手术模型,模拟临床中三阴性乳腺癌手术切除过程。实验结果表明DAC+CP@Gel的联合治疗可有效的抑制三阴性乳腺癌术后复发并有效地大幅度延长小鼠存活率。

综上所述,本论文成功构建了自催化型纳米反应器CP以及可注射型纳米复合水凝胶CP@Gel,设计了一种集去甲基化、自催化型纳米反应器和诱导细胞焦亡三位一体的三阴性乳腺癌治疗策略。CP作为自催化型纳米反应器,表现出良好的稳定性、水溶性以及分散性,可以在微酸环境中快速分解,发生类芬顿反应释放ROS。DAC作为临床用去甲基化药物,可以有效对三阴性乳腺癌细胞DNA去甲基化。本研究在细胞水平与动物水平上均证明了可以通过DAC+CP的双药法成功诱导乳腺癌细胞发生焦亡,有效激活小鼠的抗肿瘤免疫反应。同时,针对三阴性乳腺癌的术后复发问题,本研究设计了DAC+CP@Gel可注射型水凝胶载药体系在术后辅助治疗中表现出了良好的效果,成功抑制了小鼠三阴性乳腺癌的术后原位复发并激活了小鼠肿瘤免疫记忆。

本论文通过临床药物证明了在体水平中肿瘤细胞表观遗传的可编辑性,同时成功地利用凋亡诱导性药物引发细胞焦亡并激活机体抗肿瘤免疫反应,在三阴性乳腺癌原位肿瘤治疗与术后辅助治疗中表现出巨大的临床转化潜力,作为引发细胞焦亡的范例为其余凋亡诱导性治疗方案开辟了新的应用场景与肿瘤治疗思路。

外文摘要:

Triple-negative breast cancer (TNBC) has a high degree of malignancy, weak immune response, and low five-year survival rate. Currently, the main postoperative adjuvant treatment methods are chemotherapy and radiotherapy. Chemotherapy and radiotherapy not only increase the risk of metastasis, but also has strong side effects that seriously affect the life quality of patients. Surgical resection of tumor in situ has a poor prognosis and is prone to postoperative recurrence and metastasis. Better treatment strategies are urgently needed. Pyroptosis is a newly discovered immunogenic programmed cell death mode, which has broad application prospects in the treatment of TNBC. Among the various pathways that induce tumor cell pyroptosis, caspase-3-related pathways have received extensive attention due to their duality in inducing apoptosis or pyroptosis. Caspase-3 is a typical apoptosis-related protein. When the pyroptosis executive protein GSDME is highly expressed in the cell, the activated caspase-3 in the cell will cut GSDME and release its N-terminal domain to perforate the cell membrane, resulting in cell swelling and rupture and trigger cell death. Therefore, how to activate caspase-3 and increase the expression of GSDME in tumor cells is a key issue in the pyroptosis-dependent tumor treatment. In this project, a copper peroxide nanoreactor was designed and synthesized, starting from the activation of the caspase-3 pathway by intracellular oxidative stress damage, and using the clinical demethylation drug Decitabine (DAC/DEC) to improve TNBC The expression of GSDME in the cells realizes the activation of pyroptosis and tumor immune response.

 

(1) Successfully prepared copper peroxide nanoparticles (CP) that can autocatalyze the generation of hydrogen peroxide (H2O2) to efficiently generate reactive oxygen species (ROS) and not be affected by the lack of H2O2 in the tumor constraints, and then the properties of CP are studied in detail. CP can be decomposed to produce Cu2+ and H2O2 in an acidic environment/lysosome (pH 5.5) by •OH detection and immunofluorescent staining at the cellular level, and then a Fenton-like reaction between Cu2+ and H2O2 generates hydroxyl radicals (•OH), significantly increased the level of intracellular ROS, thereby efficiently activating the caspase-3 signaling pathway.

 

(2) Through bioinformatics analysis, it was found that Gsdme was highly methylated in breast cancer cells, and further q-PCR experiment proved that the methylation of Gsdme in the cancer cell line 4T1 is severe, and the expression of GSDME can be effectively increased by using DAC. After co-administration of DAC+CP, the cells undergo pyroptosis and release LDH, ATP, IL-1β, HMGB1 and other marker factors. Based on this, we used the mouse TNBC cell line 4T1 to establish a mouse subcutaneous tumor model for anti-tumor treatment. The results showed that DAC+CP has a good anti-tumor effect in vivo, directly killing tumor cells and at the same time the anti-tumor immune response in mice was effectively activated, and tumor proliferation was significantly inhibited.

 

(3) TNBC not only has limited treatment options, but also a high recurrence rate after surgical resection that threatens the survival of patients. The main reason for postoperative recurrence is that small tumor lesions and postoperative immunosuppressive microenvironment promote the growth of residual cells. Therefore, we designed and synthesized a nanocomposite hydrogel CP@Gel that suitable for postoperative local sustained drug release, combined with DAC to kill residual cells and simultaneously reverse the immunosuppressive microenvironment and activate anti-tumor immunity as a postoperative adjuvant therapy for TNBC. CP@Gel is an injectable hydrogel, which slowly dissociates after in vivo injection, and the released CP nanoparticles induce the activation of caspase-3 in postoperative residual tumor lesion cells, and DAC increases the expression of GSDME, further triggering GSDME-dependent pyroptosis to achieve anti-tumor immunity. We used the 4T1-Luc cell line with the luciferase gene to construct a mouse model of incomplete tumor resection, simulating the surgical resection process of TNBC in clinic. The experimental results showed that the combined treatment of DAC+CP@Gel can effectively inhibit the postoperative recurrence of TNBC and effectively prolong the survival rate of mice.

 

In summary, this work successfully constructed the autocatalytic nanoreactor CP and the injectable nanocomposite hydrogel CP@Gel, and designed a combination of demethylation, self-catalytic nanoreactor and induction of pyroptosis. An integrated treatment strategy for TNBC. As a self-catalytic nanoreactor, CP exhibits good stability, water solubility, dispersibility, and biological safety. It can be rapidly decomposed in a slightly acidic environment, and a Fenton-like reaction occurs to release ROS. As a demethylation drug for clinical use, DAC can effectively demethylate DNA in TNBC cells. This study proved that the double-drug method of DAC+CP can successfully induce pyroptosis in breast cancer cells, release a large amount of cellular content and immune signaling factors, and effectively activate the anti-tumor immune response at both the cellular level and the animal level. At the same time, for the postoperative recurrence of TNBC, we designed the DAC+CP@Gel injectable hydrogel drug-loading system, which successfully inhibited TNBC postoperative orthotopic recurrence and activated tumor immune memory in mice.

 

This study proved the editability of tumor cell epigenetics in vivo through clinical drugs, and used apoptosis-inducing drugs to trigger pyroptosis and activate the body's anti-tumor immune response successfully, which have shown great potential for clinical transformation in situ TNBC treatment and postoperative adjuvant therapy. As a paradigm of pyroptosis induction, it has opened new application scenarios and tumor treatment strategies for other apoptosis-inducing therapies.

参考文献:
[1] Kocarnik J M, Compton K, Frances E D, et al. Cancer Incidence, Mortality, Years of Life Lost, Years Lived With Disability, and Disability-Adjusted Life Years for 29 Cancer Groups From 2010 to 2019: A Systematic Analysis for the Global Burden of Disease Study 2019 [J]. JAMA Oncol, 2022, 8, 420-444.
[2] Xia C, Dong X, Li H, et al. Cancer Statistics in China and United States, 2022: Profiles, Trends, And Determinants [J]. Chin Med J (Engl), 2022, 135, 584-590.
[3] Haffty B G, Yang Q, Reiss M, et al. Locoregional Relapse and Distant Metastasis in Conservatively Managed Triple Negative Early-Stage Breast Cancer [J]. J Clin Oncol, 2006, 24, 5652-5657.
[4] Zhong Y Y, Shu S W, Yan G, et al. Clinical Characteristics and Prognosis of Triple-negative Breast Cancer: A Report of 305 Cases [J]. Chin J Cancer, 2008, 27, 561-565.
[5] Dent R, Trudeau M, Pritchard K I, et al. Triple-Negative Breast Cancer: Clinical Features and Patterns of Recurrence [J]. Clin Cancer Res, 2007, 13, 4429-4434.
[6] Wang C, Xu L, Liang C, et al. Immunological Responses Triggered by Photothermal Therapy with Carbon Nanotubes in Combination with Anti-CTLA-4 Therapy to Inhibit Cancer Metastasis [J]. Adv Mater, 2014, 26, 8154-8162.
[7] Ma Y, Li X, Li A, et al. H2S-Activable MOF Nanoparticle Photosensitizer for Effective Photodynamic Therapy against Cancer with Controllable Singlet-Oxygen Release [J]. Angew Chem Int Ed Engl, 2017, 56, 13752-13756.
[8] Li J, Anraku Y, Kataoka K. Self-Boosting Catalytic Nanoreactors Integrated with Triggerable Crosslinking Membrane Networks for Initiation of Immunogenic Cell Death by Pyroptosis [J]. Angew Chem Int Ed Engl, 2020, 59, 13526-13530.
[9] Liu Y, Zhen W, Wang Y, et al. Na2S2O8 Nanoparticles Trigger Antitumor Immunotherapy through Reactive Oxygen Species Storm and Surge of Tumor Osmolarity [J]. J Am Chem Soc, 2020, 142, 21751-21757.
[10] Dong Z, Feng L, Hao Y, et al. Synthesis of CaCO3-Based Nanomedicine for Enhanced Sonodynamic Therapy via Amplification of Tumor Oxidative Stress [J]. Chem, 2020, 6, 1391-1407.
[11] Yang X, Gao L, Wei Y, et al. Photothermal Hydrogel Platform for Prevention of Post-Surgical Tumor Recurrence and Improving Breast Reconstruction [J]. J Nanobiotechnol, 2021, 19, 307.
[12] Zhang R, Wang L, Wang X, et al. Acid-Induced in Vivo Assembly of Gold Nanoparticles for Enhanced Photoacoustic Imaging-Guided Photothermal Therapy of Tumors [J]. Adv Healthc Mater, 2020, 9, e2000394.
[13] He T, Yuan Y, Jiang C, et al. Light-Triggered Transformable Ferrous Ion Delivery System for Photothermal Primed Chemodynamic Therapy [J]. Angew Chem Int Ed Engl, 2021, 60, 6047-6054.
[14] Xing Y, Kang T, Luo X, et al. Coral-shaped Au Nanostructures for Selective Apoptosis Induction during Photothermal Therapy [J]. J Mater Chem B, 2019, 7, 6224-6231.
[15] Liu C, Cao Y, Cheng Y, et al. An Open Source and Reduce Expenditure ROS Generation Strategy for Chemodynamic/Photodynamic Synergistic Therapy [J]. Nat Commun, 2020, 11, 1735.
[16] Jia D, Gong L, Li Y, et al. {BiW8O30} Exerts Antitumor Effect by Triggering Pyroptosis and Upregulating Reactive Oxygen Species [J]. Angew Chem Int Ed Engl, 2021,60,21449.
[17] Zhou B, Zhang J Y, Liu X S, et al. Tom20 Senses Iron-Activated ROS Signaling to Promote Melanoma Cell Pyroptosis [J]. Cell Res, 2018, 28, 1171-1185.
[18] Yi J, Zhu J, Wu J, et al. Oncogenic Activation of PI3K-AKT-mTOR Signaling Suppresses Ferroptosis via SREBP-Mediated Lipogenesis [J]. Proc Natl Acad Sci U S A, 2020, 117, 31189-31197.
[19] Wang H, Liu C, Zhao Y, et al. Mitochondria Regulation In Ferroptosis [J]. Eur J Cell Biol, 2020, 99, 151058.
[20] Kroemer G, Galassi C, Zitvogel L, et al. Immunogenic Cell Stress And Death [J]. Nat Immunol, 2022, 23, 487-500.
[21] Liu J, Song X, Kuang F, et al. NUPR1 Is A Critical Repressor of Ferroptosis [J]. Nat Commun, 2021, 12, 647.
[22] Han Y, Sun W, Ren D, et al. SIRT1 Agonism Modulates Cardiac NLRP3 Inflammasome Through Pyruvate Dehydrogenase during Ischemia and Reperfusion [J]. Redox Biol, 2020, 34, 101538.
[23] Zhang Q, Luo Q, Liu Z, et al. Nano-ROS-Generating Approaches to Cancer Dynamic Therapy: Lessons from Nanoparticles [J]. Chem Eng J, 2023, 457, 141255.
[24] Lin L S, Huang T, Song J, et al. Synthesis of Copper Peroxide Nanodots for H2O2 Self-Supplying Chemodynamic Therapy [J]. J Am Chem Soc, 2019, 141, 9937-9945.
[25] Ding M, Kong X, Chen W, et al. Efficient Starvation Therapy with Three-Pathway Blocking in Combination with PTT/CDT for TME Reversal and Tumor Apoptosis [J]. J Ind Eng Chem, 2022, 110, 456-470.
[26] Strasser A, Vaux D L. Cell Death in the Origin and Treatment of Cancer [J]. Mol Cell, 2020, 78, 1045-1054.
[27] Carneiro B A, El-Deiry W S. Targeting Apoptosis in Cancer Therapy [J]. Nat Rev Clin Oncol, 2020, 17, 395-417.
[28] Huang J L, Jiang G, Song Q X, et al. Lipoprotein-Biomimetic Nanostructure Enables Efficient Targeting Delivery of Sirna to Ras-Activated Glioblastoma Cells Via Macropinocytosis [J]. Nat Commun, 2017, 8, 15144.
[29] Jorgensen I, Rayamajhi M, Miao E A. Programmed Cell Death as A Defence Against Infection [J]. Nat Rev Immunol, 2017, 17, 151-164.
[30] Foley K, Kim V, Jaffee E, et al. Current Progress in Immunotherapy for Pancreatic Cancer [J]. Cancer Lett, 2016, 381, 244-251.
[31] Zychlinsky A. Shigella Flexneri Induces Apoptosis in Infected Macrophages [J]. Nature, 1992, 358, 167-169.
[32] Shi J, Zhao Y, Wang K, et al. Cleavage of GSDMD by Inflammatory Caspases Determines Pyroptotic Cell Death [J]. Nature, 2015, 526, 660-665.
[33] Rao Z, Zhu Y, Yang P, et al. Pyroptosis in Inflammatory Diseases and Cancer [J]. Theranostics, 2022, 12, 4310-4329.
[34] Shi J, Gao W, Shao F. Pyroptosis: Gasdermin-Mediated Programmed Necrotic Cell Death [J]. Trends Biochem Sci, 2017, 42, 245-254.
[35] Frank D, Vince J E. Pyroptosis Versus Necroptosis: Similarities, Differences, and Crosstalk [J]. Cell Death Differ, 2019, 26, 99-114.
[36] Xi G, Gao J, Wan B, et al. GSDMD Is Required for Effector CD8+ T Cell Responses to Lung Cancer Cells [J]. Int Immunopharmacol, 2019, 74, 105713.
[37] Zhou Z, He H, Wang K, et al. Granzyme A from Cytotoxic Lymphocytes Cleaves GSDMB to Trigger Pyroptosis in Target Cells [J]. Science, 2020, 368, eaaz7548.
[38] Broz P, Pelegrin P, Shao F. The Gasdermins, A Protein Family Executing Cell Death and Inflammation [J]. Nat Rev Immunol, 2020, 20, 143-157.
[39] Wang Y, Gao W, Shi X, et al. Chemotherapy Drugs Induce Pyroptosis Through Caspase-3 Cleavage of A Gasdermin [J]. Nature, 2017, 547, 99-103.
[40] Hou J, Zhao R, Xia W, et al. PD-L1-Mediated Gasdermin C Expression Switches Apoptosis to Pyroptosis in Cancer Cells and Facilitates Tumour Necrosis [J]. Nat Cell Biol, 2020, 22, 1264-1275.
[41] Fan J X, Deng R H, Wang H, et al. Epigenetics-Based Tumor Cells Pyroptosis for Enhancing the Immunological Effect of Chemotherapeutic Nanocarriers [J]. Nano Lett, 2019, 19, 8049-8058.
[42] Jiang M, Qi L, Li L, et al. The Caspase-3/GSDME Signal Pathway as a Switch between Apoptosis and Pyroptosis in Cancer [J]. Cell Death Discov, 2020, 6, 112.
[43] Wang Q, Wang Y, Ding J, et al. A Bioorthogonal System Reveals Antitumour Immune Function of Pyroptosis [J]. Nature, 2020, 579, 421-426.
[44] Seo S H, Han H D, Noh K H, et al. Chitosan Hydrogel Containing GMCSF and A Cancer Drug Exerts Synergistic Anti-Tumor Effects via the Induction of CD8+T Cell-Mediated Anti-Tumor Immunity [J]. Clin Exp Metastas, 2009, 26, 179-187.
[45] Lu C, Guo C, Chen H, et al. A Novel Chimeric PD1-NKG2D-41BB Receptor Enhances Antitumor Activity of NK92 Cells Against Human Lung Cancer H1299 Cells by Triggering Pyroptosis [J]. Mol Immunol, 2020, 122, 200-206.
[46] Lu H, Zhang S, Wu J, et al. Molecular Targeted Therapies Elicit Concurrent Apoptotic and GSDME-Dependent Pyroptotic Tumor Cell Death [J]. Clin Cancer Res, 2018, 24, 6066-6077.
[47] Li Q, Chen L, Dong Z, et al. Piperlongumine Analogue L50377 Induces Pyroptosis via ROS Mediated NF-KappaB Suppression in Non-Small-Cell Lung Cancer [J]. Chem Biol Interact, 2019, 313, 108820.
[48] Deng B B, Jiao B P, Liu Y J, et al. BIX-01294 Enhanced Chemotherapy Effect in Gastric Cancer by Inducing GSDME-Mediated Pyroptosis [J]. Cell Biol Int, 2020, 44, 1890-1899.
[49] Hou J, Zhao R, Xia W, et al. PD-L1-Mediated Gasdermin C Expression Switches Apoptosis to Pyroptosis in Cancer Cells and Facilitates Tumour Necrosis [J]. Nat Cell Biol, 2020, 22, 1264-1275.
[50] Wu M, Wang Y, Yang D, et al. A PLK1 Kinase Inhibitor Enhances the Chemosensitivity of Cisplatin by Inducing Pyroptosis in Oesophageal Squamous Cell Carcinoma [J]. EBioMedicine, 2019, 41, 244-255.
[51] Zhang B, Zhu W Y, Tian H, et al. Alpinumisoflavone Triggers GSDME-Dependent Pyroptosis in Esophageal Squamous Cell Carcinomas [J]. Anat Rec (Hoboken), 2021, 304, 323-332.
[52] Dan W, Sheng W, Guo C Y, and Xiao Y C. Cell Death Mediated by the Pyroptosis Pathway with the Aid of Nanotechnology Prospects for Cancer Therapy [J]. Angew Chem Int Edit, 2020, 60, 8018-8034.
[53] Zhou S, Shang Q, Ji J, et al. A Nanoplatform to Amplify Apoptosis-to-Pyroptosis Immunotherapy via Immunomodulation of Myeloid-Derived Suppressor Cells [J]. ACS Appl Mater Interfaces, 2021, 13, 47407-47417.
[54] Wang L, Lu D, Huo M, et al. Oligomycin A Induces Apoptosis-to-Pyroptosis Switch against Melanoma with Sensitized Immunotherapy [J]. Adv Funct Mater, 2021, 32, 2106332.
[55] Wang Z, Wu Z, Liu Y, et al. New Development in CAR-T Cell Therapy [J]. J Hematol Oncol, 2017, 10, 53.
[56] Gottschalk A, Sharma S, Ford J, et al. Review Article: The Role of The Perioperative Period in Recurrence after Cancer Surgery [J]. Anesth Analg, 2010, 110, 1636-1643.
[57] Jia Y P, Shi K, Yang F, et al. Multifunctional Nanoparticle Loaded Injectable Thermoresponsive Hydrogel as NIR Controlled Release Platform for Local Photothermal Immunotherapy to Prevent Breast Cancer Postoperative Recurrence and Metastases [J]. Adv Funct Mater, 2020, 30, 2001059.
[58] Wu D, Wei C, Li Y, et al. Pyroptosis, a New Breakthrough in Cancer Treatment [J]. Front Oncol, 2021, 11, 698811.
[59] Chao Y, Chen Q, Liu Z. Smart Injectable Hydrogels for Cancer Immunotherapy [J]. Adv Funct Mater, 2019, 30, 1902785.
[60] Billiet T, Vandenhaute M, Schelfhout J, et al. A Review of Trends and Limitations in Hydrogel-Rapid Prototyping for Tissue Engineering [J]. Biomaterials, 2012, 33, 6020-6041.
[61] Sivakumaran D, Maitland D, Hoare T. Injectable Microgel-Hydrogel Composites for Prolonged Small-Molecule Drug Delivery [J]. Biomacromolecules, 2011, 12, 4112-4120.
[62] Lee K Y, Mooney D J. Alginate: Properties and biomedical applications [J]. Prog Polym Sci, 2012, 37, 106-126.
[63] Zhao Z, Li Q, Qin X, et al. An Injectable Hydrogel Reshaping Adenosinergic Axis for Cancer Therapy [J]. Adv Funct Mater, 2022, 32, 2200801.
[64] Lin C C, Metters A T. Hydrogels in Controlled Release Formulations: Network Design and Mathematical Modeling [J]. Adv Drug Deliver Rev, 2006, 58, 1379-1408.
[65] Hoffman A S. Hydrogels for Biomedical Applications [J]. Adv Drug Deliver Rev, 2012, 64, 18-23.
[66] Zhou J J, Li X H, He P Y, et al. Implantable Versatile Oxidized Bacterial Cellulose Membrane for Postoperative HNSCC Treatment via Photothermal-Boosted Immunotherapy [J]. Nano Research, 2022, 16, 951-963.
[67] Dandan M J L, Ru J W, Yuke L, et al. Postsurgical Wound Management and Prevention of Triple-Negative Breast Cancer Recurrence with A Pryoptosis-Inducing, Photopolymerizable Hydrogel [J]. J Control Release, 2023, 356, 205-218.
[68] Zhang J, Chen C, Li A, et al. Immunostimulant Hydrogel for The Inhibition of Malignant Glioma Relapse Post-Resection [J]. Nat Nanotech, 2021, 16, 538-548.
[69] Zhao H, Song Q, Zheng C, et al. Implantable Bioresponsive Nanoarray Enhances Postsurgical Immunotherapy by Activating Pyroptosis and Remodeling Tumor Microenvironment [J]. Adv Funct Mater, 2020, 30, 2005747.
[70] Li L, Jiang M, Qi L, et al. Pyroptosis, A New Bridge to Tumor Immunity [J]. Cancer Sci, 2021, 112, 3979-3994.
[71] Ploetz E, Zimpel A, Cauda V, et al. Metal-Organic Framework Nanoparticles Induce Pyroptosis in Cells Controlled by the Extracellular pH [J]. Adv Mater, 2020, 32, 1907267.
[72] Zhang Z, Zhang Y, Xia S, et al. Gasdermin E Suppresses Tumour Growth by Activating Anti-Tumour Immunity [J]. Nature, 2020, 579, 415-420.
[73] Lin L S, Huang T, Song J, et al. Synthesis of Copper Peroxide Nanodots for H2O2 Self-Supplying Chemodynamic Therapy [J]. J Am Chem Soc, 2019, 141, 9937-9945.
中图分类号:

 R31    

开放日期:

 2023-12-23    

无标题文档

   建议浏览器: 谷歌 火狐 360请用极速模式,双核浏览器请用极速模式