ACS Publications. Most Trusted. Most Cited. Most Read
My Activity
CONTENT TYPES

In Situ Manipulation of Dendritic Cells by an Autophagy-Regulative Nanoactivator Enables Effective Cancer Immunotherapy

  • Yi Wang
    Yi Wang
    CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, P.R. China
    Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
    More by Yi Wang
  • Yao-Xin Lin
    Yao-Xin Lin
    CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, P.R. China
    Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
    More by Yao-Xin Lin
  • Jie Wang
    Jie Wang
    CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, P.R. China
    Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
    More by Jie Wang
  • Sheng-Lin Qiao
    Sheng-Lin Qiao
    CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, P.R. China
    Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
    More by Sheng-Lin Qiao
  • Yu-Ying Liu
    Yu-Ying Liu
    National Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, P.R. China
    More by Yu-Ying Liu
  • Wen-Qian Dong
    Wen-Qian Dong
    National Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, P.R. China
    More by Wen-Qian Dong
  • Junqing Wang
    Junqing Wang
    Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
    More by Junqing Wang
  • Hong-Wei An
    Hong-Wei An
    CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, P.R. China
    Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100149, P.R. China
    More by Hong-Wei An
  • Chao Yang
    Chao Yang
    CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, P.R. China
    More by Chao Yang
  • Muhetaerjiang Mamuti
    Muhetaerjiang Mamuti
    CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, P.R. China
    Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
  • Lei Wang
    Lei Wang
    CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, P.R. China
    More by Lei Wang
  • Bo Huang
    Bo Huang
    National Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, P.R. China
    More by Bo Huang
  • , and 
  • Hao Wang*
    Hao Wang
    CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, P.R. China
    Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
    *E-mail: [email protected]
    More by Hao Wang
Cite this: ACS Nano 2019, 13, 7, 7568–7577
Publication Date (Web):July 1, 2019
https://doi.org/10.1021/acsnano.9b00143
Copyright © 2019 American Chemical Society

    Article Views

    4086

    Altmetric

    -

    Citations

    54
    LEARN ABOUT THESE METRICS
    Other access options
    Supporting Info (7)»

    Abstract

    Abstract Image

    Cellular immunotherapeutics aim to employ immune cells as anticancer agents. Ex vivo engineering of dendritic cells (DCs), the initial role of an immune response, benefits tumor elimination by boosting specific antitumor responses. However, directly activating DCs in vivo is less efficient and therefore quite challenging. Here, we designed a nanoactivator that manufactures DCs through autophagy upregulating in vivo directly, which lead to a high-efficiency antigen presention of DCs and antigen-specific T cells generation. The nanoactivator significantly enhances tumor antigen cross-presentation and subsequent T cell priming. Consequently, in vivo experiments show that the nanoactivators successfully reduce tumor growth and prolong murine survival. Taken together, these results indicate in situ DCs manipulation by autophagy induction is a promising strategy for antigen presentation enhancement and tumor elimination.

    Read this article

    To access this article, please review the available access options below.

    Get instant access

    Purchase Access

    Read this article for 48 hours. Check out below using your ACS ID or as a guest.

    Recommended

    Access through Your Institution

    You may have access to this article through your institution.

    Your institution does not have access to this content. You can change your affiliated institution below.

    Supporting Information

    ARTICLE SECTIONS
    Jump To

    The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acsnano.9b00143.

    • Data file S1: The MS-ESI spectra of B peptide (TIF)

    • Data file S2: The HPLC spectra of B peptide (TIF)

    • Data file S3: The MS-ESI spectra of OVA peptide (TIF)

    • Data file S4: The HPLC spectra of OVA peptide (TIF)

    • Data file S5: The MS-ESI spectra of B(S) peptide (TIF)

    • Data file S6: The HPLC spectra of B(S) peptide (TIF)

    • Materials, methods of transfection, RNA extraction and laser scanning confocal microscopy imaging, characterization of peptide and copolymers, and toxicity assessment of nanoactivators in vitro and in vivo (PDF)

    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

    Cited By

    This article is cited by 54 publications.

    1. Yangkai Zhou, Jing Yuan, Ke Xu, Shilin Li, Ying Liu. Nanotechnology Reprogramming Metabolism for Enhanced Tumor Immunotherapy. ACS Nano 2024, 18 (3) , 1846-1864. https://doi.org/10.1021/acsnano.3c11260
    2. Tingting He, Lu Wang, Shuangquan Gou, Lu Lu, Genhua Liu, Kui Wang, Yulu Yang, Qiaojian Duan, Wenbo Geng, Peng Zhao, Zhong Luo, Kaiyong Cai. Enhanced Immunogenic Cell Death and Antigen Presentation via Engineered Bifidobacterium bifidum to Boost Chemo-immunotherapy. ACS Nano 2023, 17 (11) , 9953-9971. https://doi.org/10.1021/acsnano.2c11474
    3. Jingwen Jiang, Yanjuan Huang, Zishan Zeng, Chunshun Zhao. Harnessing Engineered Immune Cells and Bacteria as Drug Carriers for Cancer Immunotherapy. ACS Nano 2023, 17 (2) , 843-884. https://doi.org/10.1021/acsnano.2c07607
    4. Quanwei Sun, Xiaohui Hou, Jinming Yang, Mengmeng Zhang, Ye Yang, Yang Liu, Wei Shen, Dengke Yin. Heparin-Coated Photosensitive Metal–Organic Frameworks as Drug Delivery Nanoplatforms of Autophagy Inhibitors for Sensitized Photodynamic Therapy against Breast Cancer. ACS Applied Materials & Interfaces 2021, 13 (46) , 55577-55590. https://doi.org/10.1021/acsami.1c18055
    5. Wolfgang J. Parak, (Associate Editor)Tanja Weil, (Editorial Advisory Board and Associate Editor of the Journal of the American Chemical Society)Paul S. Weiss (Editor-in-Chief). A Virtual Issue on Nanomedicine. ACS Nano 2021, 15 (10) , 15397-15401. https://doi.org/10.1021/acsnano.1c09106
    6. Shixian Lv, Meilyn Sylvestre, Alexander N. Prossnitz, Lucy F. Yang, Suzie H. Pun. Design of Polymeric Carriers for Intracellular Peptide Delivery in Oncology Applications. Chemical Reviews 2021, 121 (18) , 11653-11698. https://doi.org/10.1021/acs.chemrev.0c00963
    7. Jie Wang, Muhetaerjiang Mamuti, Hao Wang. Therapeutic Vaccines for Cancer Immunotherapy. ACS Biomaterials Science & Engineering 2020, 6 (11) , 6036-6052. https://doi.org/10.1021/acsbiomaterials.0c01201
    8. Yongyan Deng, Pengyu Song, Xiaohui Chen, Yue Huang, Liangjie Hong, Qiao Jin, Jian Ji. 3-Bromopyruvate-Conjugated Nanoplatform-Induced Pro-Death Autophagy for Enhanced Photodynamic Therapy against Hypoxic Tumor. ACS Nano 2020, 14 (8) , 9711-9727. https://doi.org/10.1021/acsnano.0c01350
    9. Wen Deng, Haojie Shang, Yonghua Tong, Xiao Liu, Qiu Huang, Yu He, Jian Wu, Xiaozhuo Ba, Zhiqiang Chen, Yuan Chen, Kun Tang. The application of nanoparticles-based ferroptosis, pyroptosis and autophagy in cancer immunotherapy. Journal of Nanobiotechnology 2024, 22 (1) https://doi.org/10.1186/s12951-024-02297-8
    10. Yingying Hou, Min Chen, Yuan Bian, Yuan Hu, Junlan Chuan, Lei Zhong, Yuxuan Zhu, Rongsheng Tong. Insights into vaccines for elderly individuals: from the impacts of immunosenescence to delivery strategies. npj Vaccines 2024, 9 (1) https://doi.org/10.1038/s41541-024-00874-4
    11. Yiwei Peng, Yiliang Yang, Zhenzhen Yang, Datong Gao, Jiajia Li, Qi Sun, Yitian Du, Meng Lin, Xianrong Qi. Bionic immunoactivator copresenting autophagy promoting and costimulatory molecules for synergistic cancer immunotherapy. Nano Research 2024, 17 (3) , 1710-1724. https://doi.org/10.1007/s12274-023-5933-2
    12. Yunmeng Liu, Yaxin Wang, Jincheng Zhang, Qikai Peng, Xingdong Wang, Xiyue Xiao, Kai Shi. Nanotherapeutics targeting autophagy regulation for improved cancer therapy. Acta Pharmaceutica Sinica B 2024, 44 https://doi.org/10.1016/j.apsb.2024.03.019
    13. Lulu Xue, Ajay S. Thatte, David Mai, Rebecca M. Haley, Ningqiang Gong, Xuexiang Han, Karin Wang, Neil C. Sheppard, Carl H. June, Michael J. Mitchell. Responsive biomaterials: optimizing control of cancer immunotherapy. Nature Reviews Materials 2024, 9 (2) , 100-118. https://doi.org/10.1038/s41578-023-00617-2
    14. Xinyi Zhang, Mengya Zhang, Hengqing Cui, Tinglin Zhang, Lili Wu, Can Xu, Chuan Yin, Jie Gao. Autophagy-modulating biomembrane nanostructures: A robust anticancer weapon by modulating the inner and outer cancer environment. Journal of Controlled Release 2024, 366 , 85-103. https://doi.org/10.1016/j.jconrel.2023.12.032
    15. Yingying Li, Xiyou Du, Xinru Kong, Yuelin Fang, Zhijing He, Dongzhu Liu, Hang Wu, Jianbo Ji, Xiaoye Yang, Lei Ye, Guangxi Zhai. A pro-death autophagy-based nanoplatform for enhancing antitumour efficacy with improved immune responses. European Journal of Medicinal Chemistry 2024, 263 , 115952. https://doi.org/10.1016/j.ejmech.2023.115952
    16. Yongjuan Li, Yichen Guo, Kaixin Zhang, Rongrong Zhu, Xiaoyuan Chen, Zhenzhong Zhang, Weijing Yang. Cell Death Pathway Regulation by Functional Nanomedicines for Robust Antitumor Immunity. Advanced Science 2024, 11 (3) https://doi.org/10.1002/advs.202306580
    17. Caixia Yang, Yuan Ding, Zhengwei Mao, Weilin Wang. Nanoplatform-Mediated Autophagy Regulation and Combined Anti-Tumor Therapy for Resistant Tumors. International Journal of Nanomedicine 2024, Volume 19 , 917-944. https://doi.org/10.2147/IJN.S445578
    18. Jinhua Li, Huaijuan Zhou, Chun Liu, Shuailong Zhang, Ran Du, Yulin Deng, Xuenong Zou. Biomembrane‐inspired design of medical micro/nanorobots: From cytomembrane stealth cloaks to cellularized Trojan horses. Aggregate 2023, 4 (5) https://doi.org/10.1002/agt2.359
    19. Qianqian Sun, Weilin Chen, Man Wang, Pan Zheng, Minghong Gao, Fengling Song, Chunxia Li. A “Chase and Block” Strategy for Enhanced Cancer Therapy with Hypoxia‐Promoted Photodynamic Therapy and Autophagy Inhibition Based on Upconversion Nanocomposites. Advanced Healthcare Materials 2023, 12 (27) https://doi.org/10.1002/adhm.202301087
    20. Ziqi Wang, Changhao Zhao, Yaowei Li, Jiaqi Wang, Dayong Hou, Lu Wang, Yueze Wang, Xunwei Wang, Xiao Liu, Hao Wang, Wanhai Xu. Photostable Cascade‐Activatable Peptide Self‐Assembly on a Cancer Cell Membrane for High‐Performance Identification of Human Bladder Cancer. Advanced Materials 2023, 35 (35) https://doi.org/10.1002/adma.202210732
    21. Yicheng Ye, Hao Tian, Jiamiao Jiang, Weichang Huang, Ruotian Zhang, Huaan Li, Lu Liu, Junbin Gao, Haixin Tan, Meihuan Liu, Fei Peng, Yingfeng Tu. Magnetically Actuated Biodegradable Nanorobots for Active Immunotherapy. Advanced Science 2023, 10 (25) https://doi.org/10.1002/advs.202300540
    22. Chang Wang, Siyu Wang, Diana D. Kang, Yizhou Dong. Biomaterials for in situ cell therapy. BMEMat 2023, 1 (3) https://doi.org/10.1002/bmm2.12039
    23. Jin Li, Xianyanling Yi, Liangren Liu, Xiaohui Wang, Jianzhong Ai. Advances in tumor nanotechnology: theragnostic implications in tumors via targeting regulated cell death. Apoptosis 2023, 28 (7-8) , 1198-1215. https://doi.org/10.1007/s10495-023-01851-3
    24. Aihua Wu, Afeng Yang, Qinli Tong, Guoguang Wei, Sihang Zhang, Sheng Yu, Chen Zhang, Jiaojiao Xu, Wei Lu. A rationally designed cancer vaccine based on NIR-II fluorescence image-guided light-triggered remote control of antigen cross-presentation and autophagy. Acta Pharmaceutica Sinica B 2023, 13 (7) , 3121-3136. https://doi.org/10.1016/j.apsb.2022.11.027
    25. Ke Yang, Bowen Yu, Wei Liu, Zequn Zhang, Li Huang, Shaojing Zhao, Benhua Wang, Jianing Yi, Jun Yuan, Yingping Zou, Changwei Lin, Xiangzhi Song, Minhuan Lan. All-in-one phototheranostics based on BTP-4F-DMO nanoparticles for NIR-II fluorescence/photoacoustic dual-mode imaging and combinational therapy. Chinese Chemical Letters 2023, 34 (6) , 107889. https://doi.org/10.1016/j.cclet.2022.107889
    26. Feng Xiao, Yao Chen, Jie Qi, Qiaofeng Yao, Jianping Xie, Xingyu Jiang. Multi‐Targeted Peptide‐Modified Gold Nanoclusters for Treating Solid Tumors in the Liver. Advanced Materials 2023, 35 (20) https://doi.org/10.1002/adma.202210412
    27. Xuehua Long, Jianqin Yan, Zhuoran Zhang, Jing Chang, Bin He, Yong Sun, Yan Liang. Autophagy-targeted nanoparticles for effective cancer treatment: advances and outlook. NPG Asia Materials 2022, 14 (1) https://doi.org/10.1038/s41427-022-00422-3
    28. Jaiwoo Lee, Dongyoon Kim, Quoc-Viet Le, Yu-Kyoung Oh. Nanotherapeutics for immune network modulation in tumor microenvironments. Seminars in Cancer Biology 2022, 86 , 1066-1087. https://doi.org/10.1016/j.semcancer.2021.11.005
    29. Xuan Wang, Peng Hua, Chengwei He, Meiwan Chen. Non-apoptotic cell death-based cancer therapy: Molecular mechanism, pharmacological modulators, and nanomedicine. Acta Pharmaceutica Sinica B 2022, 12 (9) , 3567-3593. https://doi.org/10.1016/j.apsb.2022.03.020
    30. Mei-Yu Lv, Wu-Yi Xiao, Yue-Peng Zhang, Ling-Ling Jin, Zhi-Heng Li, Zhixin Lei, Dong-Bing Cheng, Shou-De Jin. In situ self-assembled peptide enables effective cancer immunotherapy by blockage of CD47. Colloids and Surfaces B: Biointerfaces 2022, 217 , 112655. https://doi.org/10.1016/j.colsurfb.2022.112655
    31. Ji Qi, Shaorui Jia, Xiaoying Kang, Xinying Wu, Yuning Hong, Ke Shan, Xianglong Kong, Zhiming Wang, Dan Ding. Semiconducting Polymer Nanoparticles with Surface‐Mimicking Protein Secondary Structure as Lysosome‐Targeting Chimaeras for Self‐Synergistic Cancer Immunotherapy. Advanced Materials 2022, 34 (31) https://doi.org/10.1002/adma.202203309
    32. Hanchun Yao, Xiaobao Gong, Meilin Geng, Songchao Duan, Pan Qiao, Fangfang Sun, Zhihui Zhu, Bin Du. Cascade nanozymes based on the “butterfly effect” for enhanced starvation therapy through the regulation of autophagy. Biomaterials Science 2022, 10 (14) , 4008-4022. https://doi.org/10.1039/D2BM00595F
    33. Xianbin Ma, Shu-Jin Li, Yuantong Liu, Tian Zhang, Peng Xue, Yuejun Kang, Zhi-Jun Sun, Zhigang Xu. Bioengineered nanogels for cancer immunotherapy. Chemical Society Reviews 2022, 51 (12) , 5136-5174. https://doi.org/10.1039/D2CS00247G
    34. Ying-Hua Guan, Na Wang, Zhen-Wei Deng, Xi-Guang Chen, Ya Liu. Exploiting autophagy-regulative nanomaterials for activation of dendritic cells enables reinforced cancer immunotherapy. Biomaterials 2022, 282 , 121434. https://doi.org/10.1016/j.biomaterials.2022.121434
    35. Yingying Li, Shan Gao, Xiyou Du, Jianbo Ji, Yanwei Xi, Guangxi Zhai. Advances in autophagy as a target in the treatment of tumours. Journal of Drug Targeting 2022, 30 (2) , 166-187. https://doi.org/10.1080/1061186X.2021.1961792
    36. Jie Wang, Yi Wang, Shenglin Qiao, Muhetaerjiang Mamuti, Hongwei An, Hao Wang. In situ phase transitional polymeric vaccines for improved immunotherapy. National Science Review 2022, 9 (2) https://doi.org/10.1093/nsr/nwab159
    37. Yanfeng Zhou, Chang Liu, Haiyun Song. Innate Immunomodulatory Nanodevices for Cancer Therapy: A Review. Journal of Biomedical Nanotechnology 2022, 18 (2) , 293-318. https://doi.org/10.1166/jbn.2022.3241
    38. Shuojiong Pan, Tianyu Li, Yizheng Tan, Huaping Xu. Selenium-containing nanoparticles synergistically enhance Pemetrexed&NK cell-based chemoimmunotherapy. Biomaterials 2022, 280 , 121321. https://doi.org/10.1016/j.biomaterials.2021.121321
    39. Qinjun Chen, Tao Sun, Chen Jiang. Recent Advancements in Nanomedicine for ‘Cold’ Tumor Immunotherapy. Nano-Micro Letters 2021, 13 (1) https://doi.org/10.1007/s40820-021-00622-6
    40. Weijuan Zou, Junnian Hao, Jianrong Wu, Xiaojun Cai, Bing Hu, Zhigang Wang, Yuanyi Zheng. Biodegradable reduce expenditure bioreactor for augmented sonodynamic therapy via regulating tumor hypoxia and inducing pro-death autophagy. Journal of Nanobiotechnology 2021, 19 (1) https://doi.org/10.1186/s12951-021-01166-y
    41. Yalan Duan, Xiaoqing Tian, Qian Liu, Jianhua Jin, Juanjuan Shi, Yongzhong Hou. Role of autophagy on cancer immune escape. Cell Communication and Signaling 2021, 19 (1) https://doi.org/10.1186/s12964-021-00769-0
    42. Meng-Yu Zhang, Chen Huo, Jian-Yu Liu, Zhuang-E. Shi, Wen-Di Zhang, Jia-Jia Qu, Yue-Liang Yue, Yi-Qing Qu. Identification of a Five Autophagy Subtype-Related Gene Expression Pattern for Improving the Prognosis of Lung Adenocarcinoma. Frontiers in Cell and Developmental Biology 2021, 9 https://doi.org/10.3389/fcell.2021.756911
    43. Pengying Wu, Mingting Zhu, Yan Li, Zhen Ya, Yabo Yang, Yuchen Yuan, Wei Dong, Shifang Guo, Shukuan Lu, Lei Zhang, Yujin Zong, Mingxi Wan. Cascade‐Amplifying Synergistic Therapy for Intracranial Glioma via Endogenous Reactive Oxygen Species‐Triggered “All‐in‐One” Nanoplatform. Advanced Functional Materials 2021, 31 (46) https://doi.org/10.1002/adfm.202105786
    44. Eder Lilia Romero, Maria Jose Morilla. Preclinical autophagy modulatory nanomedicines: big challenges, slow advances. Expert Opinion on Drug Delivery 2021, 18 (10) , 1415-1434. https://doi.org/10.1080/17425247.2021.1933428
    45. Sahib Zada, Jin Seok Hwang, Mahmoud Ahmed, Trang Huyen Lai, Trang Minh Pham, Omar Elashkar, Deok Ryong Kim. Cross talk between autophagy and oncogenic signaling pathways and implications for cancer therapy. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer 2021, 1876 (1) , 188565. https://doi.org/10.1016/j.bbcan.2021.188565
    46. Ting Jiang, Xisha Chen, Xingcong Ren, Jin-Ming Yang, Yan Cheng. Emerging role of autophagy in anti-tumor immunity: Implications for the modulation of immunotherapy resistance. Drug Resistance Updates 2021, 56 , 100752. https://doi.org/10.1016/j.drup.2021.100752
    47. Wonchan Park, Keum Yong Seong, Hye Hyeon Han, Seung Yun Yang, Sei Kwang Hahn. Dissolving microneedles delivering cancer cell membrane coated nanoparticles for cancer immunotherapy. RSC Advances 2021, 11 (17) , 10393-10399. https://doi.org/10.1039/D1RA00747E
    48. Mingming Song, Chang Liu, Siyu Chen, Wenxiang Zhang. Nanocarrier-Based Drug Delivery for Melanoma Therapeutics. International Journal of Molecular Sciences 2021, 22 (4) , 1873. https://doi.org/10.3390/ijms22041873
    49. Alana Serrano Campelo de Souza, Letícia Boslooper Gonçalves, Ana Paula Lepique, Patrícia Savio de Araujo-Souza. The Role of Autophagy in Tumor Immunology—Complex Mechanisms That May Be Explored Therapeutically. Frontiers in Oncology 2020, 10 https://doi.org/10.3389/fonc.2020.603661
    50. Ruo-Chen Guo, Xue-Hao Zhang, Lei Ji, Zi-Jin Wei, Zhong-Yu Duan, Zeng-Ying Qiao, Hao Wang. Recent progress of therapeutic peptide based nanomaterials: from synthesis and self-assembly to cancer treatment. Biomaterials Science 2020, 8 (22) , 6175-6189. https://doi.org/10.1039/D0BM01358G
    51. Rui Chang, Xuehai Yan. Supramolecular Immunotherapy of Cancer Based on the Self‐Assembling Peptide Design. Small Structures 2020, 1 (2) https://doi.org/10.1002/sstr.202000068
    52. Emmanuel Naveen Raj, Yu‐Wei Lin, Chien‐Hung Chen, Kuang‐Kai Liu, Jui‐I Chao. Selective Autophagy Pathway of Nanoparticles and Nanodrugs: Drug Delivery and Pathophysiological Effects. Advanced Therapeutics 2020, 3 (9) https://doi.org/10.1002/adtp.202000085
    53. Qiuhua Tan, Yongjian Liu, Xiaoyi Deng, Jiajia Chen, Ping-Ju Tsai, Pei-Hsuan Chen, Manxiang Ye, Jiao Guo, Zhengquan Su. Autophagy: a promising process for the treatment of acetaminophen-induced liver injury. Archives of Toxicology 2020, 94 (9) , 2925-2938. https://doi.org/10.1007/s00204-020-02780-9
    54. Chen Qian, Li-Jun Yang, Hong Cui. Recent Advances in Nanotechnology for Dendritic Cell-Based Immunotherapy. Frontiers in Pharmacology 2020, 11 https://doi.org/10.3389/fphar.2020.00960

    Pair your accounts.

    Export articles to Mendeley

    Get article recommendations from ACS based on references in your Mendeley library.

    Pair your accounts.

    Export articles to Mendeley

    Get article recommendations from ACS based on references in your Mendeley library.

    You’ve supercharged your research process with ACS and Mendeley!

    STEP 1:
    Click to create an ACS ID

    Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

    Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

    Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

    MENDELEY PAIRING EXPIRED
    Your Mendeley pairing has expired. Please reconnect