51
|
Ruiz-Molina D, Mao X, Alfonso-Triguero P, Lorenzo J, Bruna J, Yuste VJ, Candiota AP, Novio F. Advances in Preclinical/Clinical Glioblastoma Treatment: Can Nanoparticles Be of Help? Cancers (Basel) 2022; 14:4960. [PMID: 36230883 PMCID: PMC9563739 DOI: 10.3390/cancers14194960] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma multiforme (GB) is the most aggressive and frequent primary malignant tumor in the central nervous system (CNS), with unsatisfactory and challenging treatment nowadays. Current standard of care includes surgical resection followed by chemotherapy and radiotherapy. However, these treatments do not much improve the overall survival of GB patients, which is still below two years (the 5-year survival rate is below 7%). Despite various approaches having been followed to increase the release of anticancer drugs into the brain, few of them demonstrated a significant success, as the blood brain barrier (BBB) still restricts its uptake, thus limiting the therapeutic options. Therefore, enormous efforts are being devoted to the development of novel nanomedicines with the ability to cross the BBB and specifically target the cancer cells. In this context, the use of nanoparticles represents a promising non-invasive route, allowing to evade BBB and reducing systemic concentration of drugs and, hence, side effects. In this review, we revise with a critical view the different families of nanoparticles and approaches followed so far with this aim.
Collapse
Affiliation(s)
- Daniel Ruiz-Molina
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
| | - Xiaoman Mao
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
| | - Paula Alfonso-Triguero
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Julia Lorenzo
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Jordi Bruna
- Neuro-Oncology Unit, Bellvitge University Hospital-ICO (IDIBELL), Avinguda de la Gran Via de l’Hospitalet, 199-203, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Victor J. Yuste
- Instituto de Neurociencias. Universitat Autònoma de Barcelona (UAB), Campus UAB, 08193 Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Campus UAB, 08193 Cerdanyola del Vallès, Spain
| | - Ana Paula Candiota
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08193 Cerdanyola del Vallès, Spain
| | - Fernando Novio
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Departament de Química, Universitat Autònoma de Barcelona (UAB), Campus UAB, 08193 Cerdanyola del Vallès, Spain
| |
Collapse
|
52
|
Jin L, Yang D, Song Y, Li D, Xu W, Zhu Y, Xu CF, Lu Y, Yang X. In Situ Programming of Nanovaccines for Lymph Node-Targeted Delivery and Cancer Immunotherapy. ACS NANO 2022; 16:15226-15236. [PMID: 36018240 DOI: 10.1021/acsnano.2c06560] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
In situ cancer vaccines consisting of antigens and adjuvants are a promising cancer treatment modality; however, the convenient manufacture of vaccines in vivo and their efficient delivery to lymph nodes (LNs) remains a major challenge. Herein, we outline a facile approach to simultaneously achieve the in situ programming of vaccines via two synergetic nanomedicines, Tu-NPFN and Ln-NPR848. Tu-NPFN (∼100 nm) generated a large number of antigens under an alternating magnetic field, and Ln-NPR848 (∼35 nm) encapsulating adjuvant R848 captured a portion of generated antigens for the manufacture of nanovaccines in situ and LN-targeted delivery, which significantly promoted the uptake and maturation of dendritic cells to initiate potent anticancer immune responses. Notably, combined with an anti-CTLA4 antibody (aCTLA-4), this therapy completely eradicated distant tumors in some mice and exerted a long-term immune memory effect on tumor metastasis. This study provides a generalizable strategy for in situ cancer vaccination.
Collapse
Affiliation(s)
- Liangjie Jin
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, P. R. China
| | - Dongmei Yang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, Guangdong, P. R. China
| | - Yonghong Song
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei 230009, Anhui, P. R. China
| | - Dongdong Li
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, P. R. China
| | - Weijia Xu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, Guangdong, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, and Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, Guangdong, P. R. China
| | - Yueqiang Zhu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, Guangdong, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, and Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, Guangdong, P. R. China
| | - Cong-Fei Xu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, Guangdong, P. R. China
| | - Yang Lu
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei 230009, Anhui, P. R. China
| | - Xianzhu Yang
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, P. R. China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, Guangdong, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, and Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, Guangdong, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology; Guangzhou 510006, P. R. China
| |
Collapse
|
53
|
Wang C, Li Y, Tian Y, Ma W, Sun Y. Effects of polymer carriers on the occurrence and development of autophagy in drug delivery. NANOSCALE ADVANCES 2022; 4:3676-3688. [PMID: 36133340 PMCID: PMC9470016 DOI: 10.1039/d2na00355d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/18/2022] [Indexed: 06/16/2023]
Abstract
Autophagy is an evolutionarily conserved catabolic process that can degrade cytoplasmic materials and recycle energy to maintain metabolite homeostasis in cells. Autophagy is closely related to various physiological or pathological processes. Macromolecular materials are widely used in drug delivery systems and disease treatments due to their intrinsic effects, such as altered pharmacokinetics and biodistribution. Interaction of autophagic flux or the signal pathway with macromolecules may cause autophagy inhibition or autophagy cell death. This review covers autophagy regulation pathways and macromolecular materials (including functional micelles, biodegradable and pH-sensitive polymers, biomacromolecules, dendrimers, coordination polymers, and hybrid nanoparticles) mediated autophagy modulation.
Collapse
Affiliation(s)
- Changduo Wang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University Qingdao 266000 China +86-532-82991203
| | - Yang Li
- Department of Pharmacy, Qingdao Municipal Hospital Qingdao 266000 China
| | - Yu Tian
- Department of Pharmaceutics, School of Pharmacy, Qingdao University Qingdao 266000 China +86-532-82991203
| | - Wenyuan Ma
- Department of Pharmaceutics, School of Pharmacy, Qingdao University Qingdao 266000 China +86-532-82991203
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University Qingdao 266000 China +86-532-82991203
| |
Collapse
|
54
|
Marei HE. Multimodal targeting of glioma with functionalized nanoparticles. Cancer Cell Int 2022; 22:265. [PMID: 35999629 PMCID: PMC9396820 DOI: 10.1186/s12935-022-02687-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/16/2022] [Indexed: 11/10/2022] Open
Abstract
The most common and aggressive primitive intracranial tumor of the central nervous system is the glioma. The blood–brain barrier (BBB) has proven to be a significant obstacle to the effective treatment of glioma. To effectively treat glioma, different ways have been used to cross the BBB to deliver drugs to the brain. Drug delivery through nanocarriers proves to be an effective and non-invasive technique for the treatment of glioma and has great potential in the treatment of glioma. In this review, we will provide an overview of nanocarrier-mediated drug delivery and related glioma therapy. Nanocarrier-mediated drug delivery techniques to cross the BBB (liposomes, micelles, inorganic systems, polymeric nanoparticles, nanogel system, and biomimetic nanoparticles) are explored. Finally, the use of nanotherapeutic approaches in the treatment of glioblastoma including chemotherapy, radiotherapy, photothermal therapy, gene therapy, glioma genome editing, immunotherapy, chimeric antigen receptor (CAR) T-cells, immune checkpoint modulators, immune photothermal therapy, vaccine-based immunotherapy, and combination therapy is summarized. Furthermore, this article offers various views on the clinical applicability of nanomedicine.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35116, Egypt.
| |
Collapse
|
55
|
Irradiation combined with PD-L1 -/- and autophagy inhibition enhances the antitumor effect of lung cancer via cGAS-STING-mediated T cell activation. iScience 2022; 25:104690. [PMID: 35847556 PMCID: PMC9283938 DOI: 10.1016/j.isci.2022.104690] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/03/2022] [Accepted: 06/27/2022] [Indexed: 01/07/2023] Open
Abstract
Radiotherapy combined with immune checkpoint blockade has gradually revealed the superiority in the antitumor therapy; however, the contribution of host PD-L1 remains elusive. In this study, we found that the activation of CD8+ T cells was strikingly increased in both irradiated PD-L1-expressing primary tumor and distant non-irradiated syngeneic tumor in PD-L1-deficient mouse host, and thus enhanced radiation-induced antitumor abscopal effect (ATAE) by activating cGAS-STING pathway. Notably, the autophagy inhibitors distinctively promoted dsDNA aggregation in the cytoplasm and increased the release of cGAS-STING-regulated IFN-β from irradiated cells, which further activated bystander CD8+ T cells to release IFN-γ and contributed to ATAE. These findings revealed a signaling cascade loop that the cytokines released from irradiated tumor recruit CD8+ T cells that in turn act on the tumor cells with amplified immune responses in PD-L1-deficient host, indicating a potential sandwich therapy strategy of RT combined with PD-L1 blockage and autophagy inhibition.
Collapse
|
56
|
Li C, Li Y, Li G, Wu S. Functional Nanoparticles for Enhanced Cancer Therapy. Pharmaceutics 2022; 14:pharmaceutics14081682. [PMID: 36015307 PMCID: PMC9412412 DOI: 10.3390/pharmaceutics14081682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/03/2022] [Accepted: 08/10/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer is the leading cause of death in people worldwide. The conventional therapeutic approach is mainly based on chemotherapy, which has a series of side effects. Compared with traditional chemotherapy drugs, nanoparticle-based delivery of anti-cancer drugs possesses a few attractive features. The application of nanotechnology in an interdisciplinary manner in the biomedical field has led to functional nanoparticles achieving much progress in cancer therapy. Nanoparticles have been involved in the diagnosis and targeted and personalized treatment of cancer. For example, different nano-drug strategies, including endogenous and exogenous stimuli-responsive, surface conjugation, and macromolecular encapsulation for nano-drug systems, have successfully prevented tumor procession. The future for functional nanoparticles is bright and promising due to the fast development of nanotechnology. However, there are still some challenges and limitations that need to be considered. Based on the above contents, the present article analyzes the progress in developing functional nanoparticles in cancer therapy. Research gaps and promising strategies for the clinical application are discussed.
Collapse
Affiliation(s)
- Chenchen Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen 518000, China
| | - Yuqing Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen 518000, China
| | - Guangzhi Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen 518000, China
- Correspondence: (G.L.); (S.W.)
| | - Song Wu
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen 518000, China
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, China
- Correspondence: (G.L.); (S.W.)
| |
Collapse
|
57
|
Wang X, Li C, Wang Y, Chen H, Zhang X, Luo C, Zhou W, Li L, Teng L, Yu H, Wang J. Smart drug delivery systems for precise cancer therapy. Acta Pharm Sin B 2022; 12:4098-4121. [DOI: 10.1016/j.apsb.2022.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/25/2022] [Accepted: 08/08/2022] [Indexed: 11/28/2022] Open
|
58
|
Hu L, Huang S, Chen G, Li B, Li T, Lin M, Huang Y, Xiao Z, Shuai X, Su Z. Nanodrugs Incorporating LDHA siRNA Inhibit M2-like Polarization of TAMs and Amplify Autophagy to Assist Oxaliplatin Chemotherapy against Colorectal Cancer. ACS APPLIED MATERIALS & INTERFACES 2022; 14:31625-31633. [PMID: 35796429 DOI: 10.1021/acsami.2c05841] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Oxaliplatin (OXA) is a first-line chemotherapeutic agent for treating colorectal cancer (CC). However, the chemotherapeutic effect of OXA on CC is limited by the M2-like polarization of tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) and protective autophagy of tumor cells. Here, a cationic polymer APEG-PAsp(PEI) (PAPEI) was prepared to deliver small-interfering RNA (siRNA) to silence the lactate dehydrogenase A (LDHA) gene (LDHA-siRNA) to enhance the chemotherapeutic effect of OXA on CC. The PAPEI/LDHA-siRNA nanocomplex effectively silenced the LDHA gene to inhibit the secretion of lactic acid from tumor cells, resulting in inhibition of the M2-like polarization of TAMs. In addition, the nanocomplex also amplified OXA-induced autophagy and transformed protective autophagy into autophagic death. Consequently, the combination treatment of OXA and PAPEI/LDHA-siRNA showed a dramatically increased chemotherapeutic effect on CC compared with the OXA-alone treatment, which also suggested its attractive potential for treating CC-like immune "cold" tumors.
Collapse
Affiliation(s)
- Lijun Hu
- Department of Ultrasound, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, P. R. China
| | - Sicong Huang
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, P. R. China
| | - Gengjia Chen
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, P. R. China
| | - Bo Li
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, P. R. China
| | - Tan Li
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, P. R. China
| | - Minzhao Lin
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, P. R. China
| | - Yongquan Huang
- Department of Ultrasound, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, P. R. China
| | - Zecong Xiao
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, P. R. China
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Zhongzhen Su
- Department of Ultrasound, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai 519000, P. R. China
| |
Collapse
|
59
|
Neganova ME, Aleksandrova YR, Sukocheva OA, Klochkov SG. Benefits and limitations of nanomedicine treatment of brain cancers and age-dependent neurodegenerative disorders. Semin Cancer Biol 2022; 86:805-833. [PMID: 35779712 DOI: 10.1016/j.semcancer.2022.06.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/25/2022] [Accepted: 06/25/2022] [Indexed: 02/07/2023]
Abstract
The treatment of central nervous system (CNS) malignancies, including brain cancers, is limited by a number of obstructions, including the blood-brain barrier (BBB), the heterogeneity and high invasiveness of tumors, the inaccessibility of tissues for early diagnosis and effective surgery, and anti-cancer drug resistance. Therapies employing nanomedicine have been shown to facilitate drug penetration across the BBB and maintain biodistribution and accumulation of therapeutic agents at the desired target site. The application of lipid-, polymer-, or metal-based nanocarriers represents an advanced drug delivery system for a growing group of anti-cancer chemicals. The nanocarrier surface is designed to contain an active ligand (cancer cell marker or antibody)-binding structure which can be modified to target specific cancer cells. Glioblastoma, ependymoma, neuroblastoma, medulloblastoma, and primary CNS lymphomas were recently targeted by easily absorbed nanocarriers. The metal- (such as transferrin drug-loaded systems), polymer- (nanocapsules and nanospheres), or lipid- (such as sulfatide-containing nanoliposomes)-based nano-vehicles were loaded with apoptosis- and/or ferroptosis-stimulating agents and demonstrated promising anti-cancer effects. This review aims to discuss effective nanomedicine approaches designed to overcome the current limitations in the therapy of brain cancers and age-dependent neurodegenerative disorders. To accent current obstacles for successful CNS-based cancer therapy, we discuss nanomedicine perspectives and limitations of nanodrug use associated with the specificity of nervous tissue characteristics and the effects nanocarriers have on cognition.
Collapse
Affiliation(s)
- Margarita E Neganova
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka, 142432, Russia
| | - Yulia R Aleksandrova
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka, 142432, Russia
| | - Olga A Sukocheva
- School of Health Sciences, Flinders University of South Australia, Bedford Park, SA 5042, Australia.
| | - Sergey G Klochkov
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka, 142432, Russia
| |
Collapse
|
60
|
Gao G, Jiang YW, Zhan W, Liu X, Tang R, Sun X, Deng Y, Xu L, Liang G. Trident Molecule with Nanobrush-Nanoparticle-Nanofiber Transition Property Spatially Suppresses Tumor Metastasis. J Am Chem Soc 2022; 144:11897-11910. [PMID: 35731698 DOI: 10.1021/jacs.2c05743] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Metastasis-induced high mortality of cancers urgently demands new approaches to simultaneously inhibit primary tumor metastasis and distant tumor growth. Herein, by rational design of a trident molecule Nap-Phe-Phe-Lys(SA-CPT)-Lys(SA-HCQ)-Tyr(H2PO3)-OH (Nap-CPT-HCQ-Yp) with three functional "spears" (i.e., a phosphotyrosine motif for enzymatic self-assembly, camptothecin (CPT) motif for chemotherapy, and hydroxychloroquine (HCQ) motif for autophagy inhibition) and nanobrush-nanoparticle-nanofiber transition property, we propose a novel strategy of intracellular enzymatic nanofiber formation and synergistic autophagy inhibition-enhanced chemotherapy and immunotherapy for spatial suppression of tumor metastasis. Under sequential alkaline phosphatase catalysis and carboxylesterase hydrolysis, Nap-CPT-HCQ-Yp undergoes nanobrush-nanoparticle-nanofiber transition, accompanied by the releases of CPT and HCQ. The formed intracellular nanofibers effectively inhibit the metastasis and invasion behaviors of cancer cells. Meanwhile, the released CPT and HCQ synergistically induce a prominent therapeutic effect through autophagy inhibition-enhanced chemotherapy. Furthermore, chemotherapy of Nap-CPT-HCQ-Yp enhances immunogenic cell death, resulting in the activation of toxic T-cells. Finally, a combination of checkpoint blockade therapy and Nap-CPT-HCQ-Yp-mediated chemotherapy elicits systemic antitumor immunity, thereby achieving efficient inhibitions of primary tumors as well as distant tumors in a breast tumor model. Our work offers a simple and feasible strategy for the design of "smart" multifunctional prodrugs to spatially suppress tumor metastasis.
Collapse
Affiliation(s)
- Ge Gao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yao-Wen Jiang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Wenjun Zhan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xiaoyang Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Runqun Tang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xianbao Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yu Deng
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Lingling Xu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Gaolin Liang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| |
Collapse
|
61
|
Novel Effects of Statins on Cancer via Autophagy. Pharmaceuticals (Basel) 2022; 15:ph15060648. [PMID: 35745567 PMCID: PMC9228383 DOI: 10.3390/ph15060648] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 02/05/2023] Open
Abstract
Cancer is one of the main causes of death globally. Most of the molecular mechanisms underlying cancer are marked by complex aberrations that activate the critical cell-signaling pathways that play a pivotal role in cell metabolism, tumor development, cytoskeletal reorganization, and metastasis. The phosphatidylinositol 3-kinase/protein kinase-B/mammalian target of the rapamycin (PI3K/AKT/mTOR) pathway is one of the main signaling pathways involved in carcinogenesis and metastasis. Autophagy, a cellular pathway that delivers cytoplasmic components to lysosomes for degradation, plays a dual role in cancer, as either a tumor promoter or a tumor suppressor, depending on the stage of the carcinogenesis. Statins are the group of drugs of choice to lower the level of low-density lipoprotein (LDL) cholesterol in the blood. Experimental and clinical data suggest the potential of statins in the treatment of cancer. In vitro and in vivo studies have demonstrated the molecular mechanisms through which statins inhibit the proliferation and metastasis of cancer cells in different types of cancer. The anticancer properties of statins have been shown to result in the suppression of tumor growth, the induction of apoptosis, and autophagy. This literature review shows the dual role of the autophagic process in cancer and the latest scientific evidence related to the inducing effect exerted by statins on autophagy, which could explain their anticancer potential.
Collapse
|
62
|
Aragon-Sanabria V, Aditya A, Zhang L, Chen F, Yoo B, Cao T, Madajewski B, Lee R, Turker MZ, Ma K, Monette S, Chen P, Wu J, Ruan S, Overholtzer M, Zanzonico P, Rudin CM, Brennan C, Wiesner U, Zhang L. Ultrasmall Nanoparticle Delivery of Doxorubicin Improves Therapeutic Index for High-Grade Glioma. Clin Cancer Res 2022; 28:2938-2952. [DOI: 10.1158/1078-0432.ccr-21-4053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/11/2022] [Accepted: 04/27/2022] [Indexed: 11/16/2022]
Abstract
Abstract
Purpose: Despite dramatic growth in the number of small molecule drugs developed to treat solid tumors, durable therapeutic options to control primary central nervous system malignancies are relatively scarce. Chemotherapeutic agents which appear biologically potent in model systems have often been found to be marginally effective at best when given systemically in clinical trials. This work presents for the first time an ultrasmall (< 8 nm) multimodal core-shell silica nanoparticle, Cornell prime dots (or C' dots), for the efficacious treatment of high-grade gliomas. Experimental Design: This work presents first-in-kind renally-clearable ultrasmall (< 8 nm) multimodal Cornell prime dots (or C' dots) with surface-conjugated doxorubicin via pH-sensitive linkers for the efficacious treatment in two different clinically relevant high-grade glioma models. Results: Optimal drug-per-particle ratios of as-developed nanoparticle-drug conjugates were established and used to obtain favorable pharmacokinetic profiles. The in vivo efficacy results showed significantly improved biological, therapeutic, and toxicological properties over the native drug after intravenous administration in platelet-derived growth factor-driven genetically engineered mouse model, and an epidermal growth factor expressing patient-derived xenograft (EGFR PDX) model. Conclusions: Ultrasmall C' dot-drug conjugates showed great translational potential over doxorubicin for improving the therapeutic outcome of patients with high-grade gliomas, even without a cancer-targeting moiety.
Collapse
Affiliation(s)
| | - Anusha Aditya
- Memorial Sloan Kettering Cancer Center, New York, United States
| | - Li Zhang
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Feng Chen
- Memorial Sloan Kettering Cancer Center, United States
| | | | - Tianye Cao
- Memorial Sloan Kettering Cancer Center, New York, United States
| | - Brian Madajewski
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | | | - Kai Ma
- Cornell University, Ithaca, NY, United States
| | - Sebastien Monette
- Memorial Sloan Kettering Cancer Center, The Rockefeller University, Weill Cornell Medicine, New York, New York, United States
| | - Peiming Chen
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jing Wu
- Hunter College, United States
| | - Shutian Ruan
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Pat Zanzonico
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Charles M. Rudin
- Memorial Sloan Kettering Cancer Center, New York, New York, United States
| | - Cameron Brennan
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Li Zhang
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
63
|
Qiao L, Yang H, Shao XX, Yin Q, Fu XJ, Wei Q. Research Progress on Nanoplatforms and Nanotherapeutic Strategies in Treating Glioma. Mol Pharm 2022; 19:1927-1951. [DOI: 10.1021/acs.molpharmaceut.1c00856] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Li Qiao
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Marine Traditional Chinese Medicine Research Center, Qingdao Academy of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Qingdao 266114, China
| | - Huishu Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xin-xin Shao
- Marine Traditional Chinese Medicine Research Center, Qingdao Academy of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Qingdao 266114, China
| | - Qiuyan Yin
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xian-Jun Fu
- Marine Traditional Chinese Medicine Research Center, Qingdao Academy of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Qingdao 266114, China
- Shandong Engineering and Technology Research Center of Traditional Chinese Medicine, Jinan 250355, China
| | - Qingcong Wei
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| |
Collapse
|
64
|
Tang S, Wen P, Li K, Deng J, Yang B. Tumor targetable and pH-sensitive polymer nanoparticles for simultaneously improve the Type 2 Diabetes Mellitus and malignant breast cancer. Bioengineered 2022; 13:9754-9765. [PMID: 35411835 PMCID: PMC9162024 DOI: 10.1080/21655979.2022.2060721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
In the recent study, we have developed novel tumor targetable and pH-sensitive PLGA nanoparticles co-loaded with camptothecin (CPT) and metformin (Metf) to simultaneously improve the Type 2 Diabetes Mellitus (T2DM) and malignant breast cancer. To improve the drug loading efficiency, the hydrophobic CPT was conjugated with PLGA polymer by the pH-sensitive hydrazone bonds (hyd). Then, the Metf was physically loaded into the hydrophobicity inner core of CPT-conjugated PLGA nanocomplex to form the dual drugs-loaded nanoparticles (NP/CPT-Metf). Furthermore, on the surface of NP/CPT-Metf was modified with tumor-homing CGKRK peptides to obtain the tumor targetable and pH-sensitive polymer nanoparticles (CNP/CPT-Metf). It was demonstrated that the developed CNP/CPT-Metf displayed sufficient sensitivity to the weak acidic tumor microenvironment. Besides, excellent ability of CNP/CPT-Metf to mediate accumulation of drugs in cells and tumor tissues finally in turn resulted in a signal enhanced anti-tumor effect. Furthermore, it was demonstrated as well that CNP/CPT-Metf was able of significantly alleviating the type 2 diabetes mellitus in diabetic mice. In summary, the developed multifunctional polymer nanoparticles might represent a promising strategy for simultaneously improve the T2DM and treat malignant breast cancer.
Collapse
Affiliation(s)
- Shi Tang
- Department of Breast Surgery, Dongguan City Maternal & Children Health Hospital, Dongguan, Guangdong, China
| | - Peiqi Wen
- Department of Breast Surgery, Dongguan City Maternal & Children Health Hospital, Dongguan, Guangdong, China
| | - Kaiheng Li
- Department of Breast Surgery, Dongguan City Maternal & Children Health Hospital, Dongguan, Guangdong, China
| | - Jiehua Deng
- Department of Breast Surgery, Dongguan City Maternal & Children Health Hospital, Dongguan, Guangdong, China
| | - Bo Yang
- Department of Breast Surgery, Dongguan City Maternal & Children Health Hospital, Dongguan, Guangdong, China
| |
Collapse
|
65
|
Lin Z, Zhang Z, Zheng H, Xu H, Wang Y, Chen C, Liu J, Yi G, Li Z, Wang X, Huang G. Molecular mechanism by which CDCP1 promotes proneural-mesenchymal transformation in primary glioblastoma. Cancer Cell Int 2022; 22:151. [PMID: 35410293 PMCID: PMC9003964 DOI: 10.1186/s12935-021-02373-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/28/2021] [Indexed: 11/10/2022] Open
Abstract
Background Compared with the proneural (PN) subtype of glioblastoma (GBM), the mesenchymal (MES) subtype is more invasive and immune evasive and is closely related to poor prognosis. Here, we used transcriptome data and experimental evidence to indicate that CUB domain-containing protein 1 (CDCP1) is a novel regulator that facilitates the transformation of PN-GBM to MES-GBM. Methods The mRNA expression data of CDCP1 in glioma were collected from the TCGA, CGGA and GEO databases, and in vitro experiments verified CDCP1 expression in glioma tissue samples. Independent prognostic analysis revealed the correlation of the CDCP1 expression level and patient survival. Bioinformatics analysis and experiments verified the biological function of CDCP1. Multivariate proportional hazards models and a PPI network were used to select key genes. A prognostic risk model for predicting the survival of glioma patients was constructed based on the selected genes. Results The results showed that the expression of CDCP1 increased with increasing tumor grade and that the overexpression of CDCP1 correlated with a poor prognosis. CDCP1 was highly expressed in MES-GBM but weakly expressed in PN-GBM. The risk model (considering CDCP1 combined with CD44 and ITGAM expression) could represent a tool for predicting survival and prognosis in glioma patients. Conclusions Our study indicates that CDCP1 plays an important role in facilitating the transformation of PN-GBM to MES-GBM. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02373-1.
Collapse
Affiliation(s)
- Zhiying Lin
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi, China.,Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China
| | - Zhu Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Haojie Zheng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Haiyan Xu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yajuan Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Chao Chen
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Junlu Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Guozhong Yi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhiyong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xiaoyan Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China. .,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Guanglong Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China. .,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
66
|
Wang X, Hua P, He C, Chen M. Non-apoptotic cell death-based cancer therapy: Molecular mechanism, pharmacological modulators, and nanomedicine. Acta Pharm Sin B 2022; 12:3567-3593. [PMID: 36176912 PMCID: PMC9513500 DOI: 10.1016/j.apsb.2022.03.020] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/25/2022] [Accepted: 02/16/2022] [Indexed: 02/08/2023] Open
Abstract
As an emerging cancer therapeutic target, non-apoptotic cell death such as ferroptosis, necroptosis and pyroptosis, etc., has revealed significant potential in cancer treatment for bypassing apoptosis to enhance the undermined therapeutic efficacy triggered by apoptosis resistance. A variety of anticancer drugs, synthesized compounds and natural products have been proven recently to induce non-apoptotic cell death and exhibit excellent anti-tumor effects. Moreover, the convergence of nanotechnology with functional materials and biomedicine science has provided tremendous opportunities to construct non-apoptotic cell death-based nanomedicine for innovative cancer therapy. Nanocarriers are not only employed in targeted delivery of non-apoptotic inducers, but also used as therapeutic components to induce non-apoptotic cell death to achieve efficient tumor treatment. This review first introduces the main characteristics, the mechanism and various pharmacological modulators of different non-apoptotic cell death forms, including ferroptosis, necroptosis, pyroptosis, autophagy, paraptosis, lysosomal-dependent cell death, and oncosis. Second, we comprehensively review the latest progresses of nanomedicine that induces various forms of non-apoptotic cell death and focus on the nanomedicine targeting different pathways and components. Furthermore, the combination therapies of non-apoptotic cell death with photothermal therapy, photodynamic therapy, immunotherapy and other modalities are summarized. Finally, the challenges and future perspectives in this regard are also discussed.
Collapse
|
67
|
Luan S, Xie R, Yang Y, Xiao X, Zhou J, Li X, Fang P, Zeng X, Yu X, Chen M, Gao H, Yuan Y. Acid-Responsive Aggregated Gold Nanoparticles for Radiosensitization and Synergistic Chemoradiotherapy in the Treatment of Esophageal Cancer. SMALL 2022; 18:e2200115. [PMID: 35261151 DOI: 10.1002/smll.202200115] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/17/2022] [Indexed: 02/05/2023]
Abstract
Radiotherapy and chemotherapy are limited by insufficient therapeutic efficacy of low-dose radiation and nonspecific drug biodistribution. Herein, an acid-responsive aggregated nanosystem (AuNPs-D-P-DA) loaded with doxorubicin (DOX) is designed for radiosensitization and synergistic chemoradiotherapy. In response to the acid microenvironment of esophageal cancer (EC), small-sized AuNPs-D-P-DA forms large-sized gold nanoparticle (AuNPs) aggregates in tumor tissues to hinder the backflow of AuNPs to the circulation, resulting in enhanced tumor accumulation and retention. Simultaneously, the AuNPs-based radiosensitization is significantly improved because of the high concentration and large size of intratumoral AuNPs, while DOX are delivered and released specifically into tumor cells triggered by the acid microenvironment for chemo-radio synergistic therapy. Acid-responsive AuNPs exacerbate radiation-induced DNA damage, cell apoptosis, cell cycle arrest, and low colony formation ability in vitro and enhance anti-tumor efficacy in vivo compared to un-responsive control. When combined with acid-responsive DOX, the therapeutic efficacy of the formulation is further improved by their synergistic effect. After the treatment of acid-responsive AuNPs plus radiotherapy, fatty acid metabolism is reprogrammed in xenograft models, which provides potential targets for further improvement of radiosensitization. In summary, the acid-responsive AuNPs-D-P-DA nanosystem leverages the radio- and chemotherapeutic synergies of AuNPs-sensitized X-ray irradiation and acid-responsive DOX in the treatment of EC.
Collapse
Affiliation(s)
- Siyuan Luan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Rou Xie
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Yushang Yang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Xin Xiao
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Jianfeng Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Xiaokun Li
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Pinhao Fang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Xiaoxi Zeng
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Xiangrong Yu
- Department of Radiology, Zhuhai People's Hospital, Jinan University, Zhuhai, 519000, P. R. China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, P. R. China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Yong Yuan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
68
|
Liu Z, Ji X, He D, Zhang R, Liu Q, Xin T. Nanoscale Drug Delivery Systems in Glioblastoma. NANOSCALE RESEARCH LETTERS 2022; 17:27. [PMID: 35171358 PMCID: PMC8850533 DOI: 10.1186/s11671-022-03668-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/09/2022] [Indexed: 05/13/2023]
Abstract
Glioblastoma is the most aggressive cerebral tumor in adults. However, the current pharmaceuticals in GBM treatment are mainly restricted to few chemotherapeutic drugs and have limited efficacy. Therefore, various nanoscale biomaterials that possess distinct structure and unique property were constructed as vehicles to precisely deliver molecules with potential therapeutic effect. In this review, nanoparticle drug delivery systems including CNTs, GBNs, C-dots, MOFs, Liposomes, MSNs, GNPs, PMs, Dendrimers and Nanogel were exemplified. The advantages and disadvantages of these nanoparticles in GBM treatment were illustrated.
Collapse
Affiliation(s)
- Zihao Liu
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China
| | - Xiaoshuai Ji
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250014, China
| | - Dong He
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China
| | - Rui Zhang
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China
| | - Qian Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Tao Xin
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250014, China.
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan, 250014, China.
- Department of Neurosurgery, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang Jiangxi, 330006, China.
| |
Collapse
|
69
|
Hu Y, Gao S, Khan AR, Yang X, Ji J, Xi Y, Zhai G. Tumor microenvironment-responsive size-switchable drug delivery nanosystems. Expert Opin Drug Deliv 2022; 19:221-234. [PMID: 35164610 DOI: 10.1080/17425247.2022.2042512] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Compared with ordinary chemotherapeutic drugs, the variable-size nanoparticles (NPs) have better therapeutic effects and fewer side effects. AREAS COVERED This review mainly summarizes the strategies used to construct smart, size-tunable nanocarriers based on characteristic factors of tumor microenvironment (TME) to dramatically increase the penetration and retention of drugs within tumors. EXPERT OPINION Nanosystems with changeable sizes based on the TME have been extensively studied in the past decade, and their permeability and retention have been greatly improved, making them a very promising treatment for tumors.
Collapse
Affiliation(s)
- Yue Hu
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Shan Gao
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Abdur Rauf Khan
- Government of Punjab, Specialized HealthCare and Medical Education Department, Lahore, Pakistan
| | - Xiaoye Yang
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Jianbo Ji
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Yanwei Xi
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Guangxi Zhai
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| |
Collapse
|
70
|
Pang X, Dong A, Deng L, Cheng Y, Deng H. 生物医用纳米材料在增强肿瘤细胞免疫原性死亡中的应用. CHINESE SCIENCE BULLETIN-CHINESE 2022. [DOI: 10.1360/tb-2021-0987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
71
|
Lakshmi BA, Kim YJ. Modernistic and Emerging Developments of Nanotechnology in Glioblastoma-Targeted Theranostic Applications. Int J Mol Sci 2022; 23:ijms23031641. [PMID: 35163563 PMCID: PMC8836088 DOI: 10.3390/ijms23031641] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/20/2022] [Accepted: 01/27/2022] [Indexed: 02/06/2023] Open
Abstract
Brain tumors such as glioblastoma are typically associated with an unstoppable cell proliferation with aggressive infiltration behavior and a shortened life span. Though treatment options such as chemotherapy and radiotherapy are available in combating glioblastoma, satisfactory therapeutics are still not available due to the high impermeability of the blood–brain barrier. To address these concerns, recently, multifarious theranostics based on nanotechnology have been developed, which can deal with diagnosis and therapy together. The multifunctional nanomaterials find a strategic path against glioblastoma by adjoining novel thermal and magnetic therapy approaches. Their convenient combination of specific features such as real-time tracking, in-depth tissue penetration, drug-loading capacity, and contrasting performance is of great demand in the clinical investigation of glioblastoma. The potential benefits of nanomaterials including specificity, surface tunability, biodegradability, non-toxicity, ligand functionalization, and near-infrared (NIR) and photoacoustic (PA) imaging are sufficient in developing effective theranostics. This review discusses the recent developments in nanotechnology toward the diagnosis, drug delivery, and therapy regarding glioblastoma.
Collapse
|
72
|
Mahhengam N, Kazemnezhad K, Setia Budi H, Ansari MJ, Olegovich Bokov D, Suksatan W, Thangavelu L, Siahmansouri H. Targeted therapy of tumor microenvironment by gold nanoparticles as a new therapeutic approach. J Drug Target 2022; 30:494-510. [DOI: 10.1080/1061186x.2022.2032095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Negah Mahhengam
- Faculty of General Medicine, Belarusian State Medical University, Minsk, Belarus.
| | - Kimia Kazemnezhad
- Faculty of General Medicine, Belarusian State Medical University, Minsk, Belarus.
| | - Hendrik Setia Budi
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya 60132, Indonesia.
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University,Al-kharj, Saudi Arabia.
| | - Dmitry Olegovich Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, 8 Trubetskaya St., bldg. 2, Moscow, 119991, Russian Federation.
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand.
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India.
| | - Homayoon Siahmansouri
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
73
|
Tang L, Zhang A, Zhang Z, Zhao Q, Li J, Mei Y, Yin Y, Wang W. Multifunctional inorganic nanomaterials for cancer photoimmunotherapy. Cancer Commun (Lond) 2022; 42:141-163. [PMID: 35001556 PMCID: PMC8822595 DOI: 10.1002/cac2.12255] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/24/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
Phototherapy and immunotherapy in combination is regarded as the ideal therapeutic modality to treat both primary and metastatic tumors. Immunotherapy uses different immunological approaches to stimulate the immune system to identify tumor cells for targeted elimination. Phototherapy destroys the primary tumors by light irradiation, which induces a series of immune responses through triggering immunogenic cancer cell death. Therefore, when integrating immunotherapy with phototherapy, a novel anti-cancer strategy called photoimmunotherapy (PIT) is emerging. This synergistic treatment modality can not only enhance the effectiveness of both therapies but also overcome their inherent limitations, opening a new era for the current anti-cancer therapy. Recently, the advancement of nanomaterials affords a platform for PIT. From all these nanomaterials, inorganic nanomaterials stand out as ideal mediators in PIT due to their unique physiochemical properties. Inorganic nanomaterials can not only serve as carriers to transport immunomodulatory agents in immunotherapy owing to their excellent drug-loading capacity but also function as photothermal agents or photosensitizers in phototherapy because of their great optical characteristics. In this review, the recent advances of multifunctional inorganic nanomaterial-mediated drug delivery and their contributions to cancer PIT will be highlighted.
Collapse
Affiliation(s)
- Lu Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China.,National Medical Products Administration Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China
| | - Aining Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China.,National Medical Products Administration Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China
| | - Ziyao Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China.,National Medical Products Administration Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China
| | - Qingqing Zhao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China.,National Medical Products Administration Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China
| | - Jing Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China.,National Medical Products Administration Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China
| | - Yijun Mei
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China.,National Medical Products Administration Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China
| | - Yue Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China.,National Medical Products Administration Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China.,National Medical Products Administration Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, Jiangsu, P. R. China
| |
Collapse
|
74
|
Wang X, Li Y, Lu J, Deng X, Wu Y. Engineering Nanoplatform for Combined Cancer Therapeutics via Complementary Autophagy Inhibition. Int J Mol Sci 2022; 23:657. [PMID: 35054843 PMCID: PMC8776236 DOI: 10.3390/ijms23020657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/01/2022] [Accepted: 01/05/2022] [Indexed: 12/21/2022] Open
Abstract
Despite advances in the development of tumor treatments, mortality from cancer continues to increase. Nanotechnology is expected to provide an innovative anti-cancer therapy, to combat challenges such as multidrug resistance and tumor recurrence. Nevertheless, tumors can greatly rely on autophagy as an alternative source for metabolites, and which desensitizes cancer cells to therapeutic stress, hindering the success of any current treatment paradigm. Autophagy is a conserved process by which cells turn over their own constituents to maintain cellular homeostasis. The multistep autophagic pathway provides potentially druggable targets to inhibit pro-survival autophagy under various therapeutic stimuli. In this review, we focus on autophagy inhibition based on functional nanoplatforms, which may be a potential strategy to increase therapeutic sensitivity in combinational cancer therapies, including chemotherapy, radiotherapy, phototherapy, sonodynamic therapy, and immunotherapy.
Collapse
Affiliation(s)
- Xuan Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; (X.W.); (J.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yunhao Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China;
| | - Jianqing Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; (X.W.); (J.L.)
| | - Xiongwei Deng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; (X.W.); (J.L.)
| | - Yan Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; (X.W.); (J.L.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
75
|
Zeng Q, Ma X, Song Y, Chen Q, Jiao Q, Zhou L. Targeting regulated cell death in tumor nanomedicines. Am J Cancer Res 2022; 12:817-841. [PMID: 34976215 PMCID: PMC8692918 DOI: 10.7150/thno.67932] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/21/2021] [Indexed: 12/17/2022] Open
Abstract
Nanomedicines hold great potential in anticancer therapy by modulating the biodistribution of nanomaterials and initiating targeted oxidative stress damage, but they are also limited by the inherent self-protection mechanism and the evolutionary treatment resistance of cancer cells. New emerging explorations of regulated cell death (RCD), including processes related to autophagy, ferroptosis, pyroptosis, and necroptosis, substantially contribute to the augmented therapeutic efficiency of tumors by increasing the sensitivity of cancer cells to apoptosis. Herein, paradigmatic studies of RCD-mediated synergistic tumor nanotherapeutics are introduced, such as regulating autophagy-enhanced photodynamic therapy (PDT), targeting ferroptosis-sensitized sonodynamic therapy (SDT), inducing necroptosis-augmented photothermal therapy (PTT), and initiating pyroptosis-collaborative chemodynamic therapy (CDT), and the coordination mechanisms are discussed in detail. Multiangle analyses addressing the present challenges and upcoming prospects of RCD-based nanomedicines have also been highlighted and prospected for their further strengthening and the broadening of their application scope. It is believed that up-and-coming coadjutant therapeutic methodologies based on RCDs will considerably impact precision nanomedicine for cancer.
Collapse
|
76
|
Peng S, Xiao F, Chen M, Gao H. Tumor-Microenvironment-Responsive Nanomedicine for Enhanced Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103836. [PMID: 34796689 PMCID: PMC8728817 DOI: 10.1002/advs.202103836] [Citation(s) in RCA: 165] [Impact Index Per Article: 82.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/20/2021] [Indexed: 05/07/2023]
Abstract
The past decades have witnessed great progress in cancer immunotherapy, which has profoundly revolutionized oncology, whereas low patient response rates and potential immune-related adverse events remain major clinical challenges. With the advantages of controlled delivery and modular flexibility, cancer nanomedicine has offered opportunities to strengthen antitumor immune responses and to sensitize tumor to immunotherapy. Furthermore, tumor-microenvironment (TME)-responsive nanomedicine has been demonstrated to achieve specific and localized amplification of the immune response in tumor tissue in a safe and effective manner, increasing patient response rates to immunotherapy and reducing the immune-related side effects simultaneously. Here, the recent progress of TME-responsive nanomedicine for cancer immunotherapy is summarized, which responds to the signals in the TME, such as weak acidity, reductive environment, high-level reactive oxygen species, hypoxia, overexpressed enzymes, and high-level adenosine triphosphate. Moreover, the potential to combine nanomedicine-based therapy and immunotherapeutic strategies to overcome each step of the cancer-immunity cycle and to enhance antitumor effects is discussed. Finally, existing challenges and further perspectives in this rising field with the hope for improved development of clinical applications are discussed.
Collapse
Affiliation(s)
- Shaojun Peng
- Zhuhai Institute of Translational MedicineZhuhai Precision Medical CenterZhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University)ZhuhaiGuangdong519000China
| | - Fengfeng Xiao
- Zhuhai Institute of Translational MedicineZhuhai Precision Medical CenterZhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University)ZhuhaiGuangdong519000China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauMacau999078China
| | - Huile Gao
- Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry and Sichuan ProvinceSichuan Engineering Laboratory for Plant‐Sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of PharmacySichuan UniversityChengdu610041China
| |
Collapse
|
77
|
Mehrabian A, Mashreghi M, Dadpour S, Badiee A, Arabi L, Hoda Alavizadeh S, Alia Moosavian S, Reza Jaafari M. Nanocarriers Call the Last Shot in the Treatment of Brain Cancers. Technol Cancer Res Treat 2022; 21:15330338221080974. [PMID: 35253549 PMCID: PMC8905056 DOI: 10.1177/15330338221080974] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Our brain is protected by physio-biological barriers. The blood–brain barrier (BBB) main mechanism of protection relates to the abundance of tight junctions (TJs) and efflux pumps. Although BBB is crucial for healthy brain protection against toxins, it also leads to failure in a devastating disease like brain cancer. Recently, nanocarriers have been shown to pass through the BBB and improve patients’ survival rates, thus becoming promising treatment strategies. Among nanocarriers, inorganic nanocarriers, solid lipid nanoparticles, liposomes, polymers, micelles, and dendrimers have reached clinical trials after delivering promising results in preclinical investigations. The size of these nanocarriers is between 10 and 1000 nm and is modified by surface attachment of proteins, peptides, antibodies, or surfactants. Multiple research groups have reported transcellular entrance as the main mechanism allowing for these nanocarriers to cross BBB. Transport proteins and transcellular lipophilic pathways exist in BBB for small and lipophilic molecules. Nanocarriers cannot enter via the paracellular route, which is limited to water-soluble agents due to the TJs and their small pore size. There are currently several nanocarriers in clinical trials for the treatment of brain cancer. This article reviews challenges as well as fitting attributes of nanocarriers for brain tumor treatment in preclinical and clinical studies.
Collapse
Affiliation(s)
- Amin Mehrabian
- School of Pharmacy, Biotechnology Research Center, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Pharmaceutical Technology Institute, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Warwick Medical School, University of Warwick, Coventry, UK
| | - Mohammad Mashreghi
- School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Pharmaceutical Technology Institute, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saba Dadpour
- School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Student Research Committee, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Pharmaceutical Technology Institute, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Pharmaceutical Technology Institute, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Hoda Alavizadeh
- School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Pharmaceutical Technology Institute, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Alia Moosavian
- School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Pharmaceutical Technology Institute, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- School of Pharmacy, Biotechnology Research Center, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, 37552Mashhad University of Medical Sciences, Mashhad, Iran.,Nanotechnology Research Center, Pharmaceutical Technology Institute, 37552Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
78
|
Shi Y, Lin G, Zheng H, Mu D, Chen H, Lu Z, He P, Zhang Y, Liu C, Lin Z, Liu G. Biomimetic nanoparticles blocking autophagy for enhanced chemotherapy and metastasis inhibition via reversing focal adhesion disassembly. J Nanobiotechnology 2021; 19:447. [PMID: 34952594 PMCID: PMC8710033 DOI: 10.1186/s12951-021-01189-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/07/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Autophagy is a conserved catabolic process, which plays an important role in regulating tumor cell motility and degrading protein aggregates. Chemotherapy-induced autophagy may lead to tumor distant metastasis and even chemo-insensitivity in the therapy of hepatocellular carcinoma (HCC). Therefore, a vast majority of HCC cases do not produce a significant response to monotherapy with autophagy inhibitors. RESULTS In this work, we developed a biomimetic nanoformulation (TH-NP) co-encapsulating Oxaliplatin (OXA)/hydroxychloroquine (HCQ, an autophagy inhibitor) to execute targeted autophagy inhibition, reduce tumor cell migration and invasion in vitro and attenuate metastasis in vivo. The tumor cell-specific ligand TRAIL was bioengineered to be stably expressed on HUVECs and the resultant membrane vesicles were wrapped on OXA/HCQ-loaded PLGA nanocores. Especially, TH-NPs could significantly improve OXA and HCQ effective concentration by approximately 21 and 13 times in tumor tissues compared to the free mixture of HCQ/OXA. Moreover, the tumor-targeting TH-NPs released HCQ alkalized the acidic lysosomes and inhibited the fusion of autophagosomes and lysosomes, leading to effective blockade of autophagic flux. In short, the system largely improved chemotherapeutic performance of OXA on subcutaneous and orthotopic HCC mice models. Importantly, TH-NPs also exhibited the most effective inhibition of tumor metastasis in orthotopic HCCLM3 models, and in the HepG2, Huh-7 or HCCLM3 metastatic mice models. Finally, we illustrated the enhanced metastasis inhibition was attributed to the blockade or reverse of the autophagy-mediated degradation of focal adhesions (FAs) including E-cadherin and paxillin. CONCLUSIONS TH-NPs can perform an enhanced chemotherapy and antimetastatic effect, and may represent a promising strategy for HCC therapy in clinics.
Collapse
Affiliation(s)
- Yesi Shi
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.,State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, 361004, China
| | - Gan Lin
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Huili Zheng
- Department of Anesthesiology, Zhongshan Hospital of Xiamen University, Xiamen, 361004, China
| | - Dan Mu
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.,Amoy Hopeful Biotechnology Co., Ltd., Xiamen, 361027, China
| | - Hu Chen
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.,Amoy Hopeful Biotechnology Co., Ltd., Xiamen, 361027, China
| | - Zhixiang Lu
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Pan He
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Yang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Chao Liu
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
| | - Zhongning Lin
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China. .,State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, 361004, China.
| |
Collapse
|
79
|
Ni S, Chen R, Hu K. Experimental murine models of brainstem gliomas. Drug Discov Today 2021; 27:1218-1235. [PMID: 34954326 DOI: 10.1016/j.drudis.2021.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/16/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022]
Abstract
As an intractable central nervous system (CNS) tumor, brainstem gliomas (BGs) are one of the leading causes of pediatric death by brain tumors. Owing to the risk of surgical resection and the little improvement in survival time after radiotherapy and chemotherapy, there is an urgent need to find reliable model systems to better understand the regional pathogenesis of the brainstem and improve treatment strategies. In this review, we outline the evolution of BG murine models, and discuss both their advantages and limitations in drug discovery.
Collapse
Affiliation(s)
- Shuting Ni
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rujing Chen
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kaili Hu
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
80
|
Xu Q, Zhang H, Liu H, Han Y, Qiu W, Li Z. Inhibiting autophagy flux and DNA repair of tumor cells to boost radiotherapy of orthotopic glioblastoma. Biomaterials 2021; 280:121287. [PMID: 34864449 DOI: 10.1016/j.biomaterials.2021.121287] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/18/2021] [Accepted: 11/25/2021] [Indexed: 12/27/2022]
Abstract
Radio-resistance of glioblastoma (GBM) remains a leading cause of radiotherapy failure because of the protective autophagy induced by X-Ray irradiation and tumor cells' strong capability of repairing damaged DNA. It is of great importance to overcome the radio-resistance for improving the efficacy of radiotherapy. Herein, we report the novel mechanism of core-shell copper selenide coated gold nanoparticles (Au@Cu2-xSe NPs) inhibiting the protective autophagy and DNA repair of tumor cells to drastically boost the radiotherapy efficacy of glioblastoma. We reveal that the core-shell Au@Cu2-xSe NPs can inhibit the autophagy flux by effectively alkalizing lysosomes. They can increase the SQSTM1/p62 protein levels of tumor cells without influencing their mRNA. We also reveal that Au@Cu2-xSe NPs can increase the ubiquitination of DNA repair protein Rad51, and promote the degradation of Rad51 by proteasomes to prevent the DNA repair. The simultaneous inhibition of protective autophagy and DNA repair significantly suppress the growth of orthotopic GBM by using radiotherapy and our novel Au@Cu2-xSe NPs. Our work provides a new insight and paradigm to significantly improve the efficacy of radiotherapy by rationally designing theranostic nano-agents to simultaneously inhibit protective autophagy and DNA repair of tumor cells.
Collapse
Affiliation(s)
- Qi Xu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P.R. China
| | - Hao Zhang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P.R. China; College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P.R. China.
| | - Hanghang Liu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P.R. China
| | - Yaobao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P.R. China
| | - Weibao Qiu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P.R. China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, P.R. China.
| |
Collapse
|
81
|
Sun X, Dong Y, Liu Y, Song N, Li F, Yang D. Self-assembly of artificial architectures in living cells — design and applications. Sci China Chem 2021. [DOI: 10.1007/s11426-021-1091-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
82
|
Zhang X, Chen Y, He X, Zhang Y, Zhou M, Peng C, He Z, Gui S, Li Z. Smart Nanogatekeepers for Tumor Theranostics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2103712. [PMID: 34677898 DOI: 10.1002/smll.202103712] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/05/2021] [Indexed: 06/13/2023]
Abstract
Nanoparticulate drug delivery systems (nano-DDSs) are required to reliably arrive and persistently reside at the tumor site with minimal off-target side effects for clinical theranostics. However, due to the complicated environment and high interstitial pressure in tumor tissue, they can return to the bloodstream and cause secondary side effects in normal organs. Recently, a number of nanogatekeepers have been engineered via structure-transformable/stable strategies to overcome this undesirable dilemma. The emerging structure-transformable nanogatekeepers for tumor imaging and therapy are first overviewed here, particularly for nanogatekeepers undergoing structural transformation in tumor microenvironments, cell membranes, and organelles. Thereafter, intelligent structure-stable nanogatekeepers through reversible activation and artificial individualization receptors are overviewed. Finally, the ongoing challenges and prospects of nanogatekeepers for clinical translation are briefly discussed.
Collapse
Affiliation(s)
- Xunfa Zhang
- College of Pharmacy, Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230012, China
| | - Yang Chen
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xian He
- College of Pharmacy, Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Education Office of Anhui Province, Hefei, 230012, China
| | - Yachao Zhang
- College of Pharmacy, Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Education Office of Anhui Province, Hefei, 230012, China
| | - Mei Zhou
- College of Pharmacy, Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230012, China
| | - Chengjun Peng
- College of Pharmacy, Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Education Office of Anhui Province, Hefei, 230012, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Shuangying Gui
- College of Pharmacy, Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Education Office of Anhui Province, Hefei, 230012, China
| | - Zhenbao Li
- College of Pharmacy, Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Education Office of Anhui Province, Hefei, 230012, China
| |
Collapse
|
83
|
Zhang X, Zhang JG, Mu W, Zhou HM, Liu GL, Li Q. The role of daurisoline treatment in hepatocellular carcinoma: Inhibiting vasculogenic mimicry formation and enhancing sensitivity to sorafenib. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 92:153740. [PMID: 34600176 DOI: 10.1016/j.phymed.2021.153740] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/27/2021] [Accepted: 09/05/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Vasculogenic mimicry (VM) is a newly described tumor vascular phenomenon that is independent of traditional angiogenesis and provides an adequate blood supply for tumor growth. VM has been consistently observed in different cancer types. Hence, inhibition of VM may be considered a new anticancer therapeutic strategy. PURPOSE This study aimed to elucidate the potential anticancer effect of daurisoline (DS) on hepatocellular carcinoma (HCC) and the potential molecular mechanism by which DS inhibits VM. We also verified whether combination treatment with sorafenib and DS constitutes a novel therapeutic approach to prevent HCC progression. METHODS The effects of DS on proliferation were evaluated by Cell Counting Kit-8 (CCK-8), colony formation, and 5-ethynyl-2'-deoxyuridine (EdU) incorporation assays. 4',6-Diamidino-2-phenylindole (DAPI) staining and flow cytometric analysis were employed to investigate its effects on apoptosis. Western blot analysis, Matrigel tube formation assays, pulldown assays and immunofluorescence staining were applied to validate the potential mechanism by which DS inhibits VM. Mouse xenograft models were used to evaluate anticancer activities. RESULTS DS inhibited HCC cell proliferation, induced HCC cell apoptosis and repressed VM formation by inactivating RhoA/ROCK2-mediated AKT and ERK-p38 MAPK signaling. Additionally, DS dramatically sensitized HCC cell lines to sorafenib, a curative anticancer drug for patients with advanced HCC. CONCLUSIONS Our study provides insights into the molecular mechanisms underlying DS-induced inhibition of VM, which may facilitate the development of a novel clinical anti-HCC drug. Moreover, our findings suggest that the combination of DS and sorafenib constitutes a potential therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai, 200080, China; Department of Pharmacy, Shanghai Eye Diseases Prevention & Treatment Center, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai General Hospital, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200040, China
| | - Ji-Gang Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai, 200080, China
| | - Wan Mu
- Department of Pharmacy, Shanghai Eye Diseases Prevention & Treatment Center, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai General Hospital, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200040, China
| | - He-Ming Zhou
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai, 200080, China
| | - Gao-Lin Liu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai, 200080, China; Department of Pharmacy, Shanghai Eye Diseases Prevention & Treatment Center, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai General Hospital, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200040, China.
| | - Qin Li
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai, 200080, China.
| |
Collapse
|
84
|
Constructing nanocomplexes by multicomponent self-assembly for curing orthotopic glioblastoma with synergistic chemo-photothermal therapy. Biomaterials 2021; 279:121193. [PMID: 34700227 DOI: 10.1016/j.biomaterials.2021.121193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 12/18/2022]
Abstract
The blood-brain barrier (BBB) is one of the major limitations of glioblastoma therapy in the clinic. Nanodrugs have shown great potential for glioblastoma therapy. Herein, we purposefully developed a multicomponent self-assembly nanocomplex with very high drug loading content for curing orthotopic glioblastoma with synergistic chemo-photothermal therapy. The nanocomplex consisted of self-assembled pH-responsive nanodrugs derived from amino acid-conjugated camptothecin (CPT) and canine dyes (IR783) coated with peptide Angiopep-2-conjugated copolymer of Ang-PEG-g-PLL. Specifically, the carrier-free nanocomplex exhibited a high drug loading content (up to 62%), good biocompatibility, and effective glioma accumulation ability. Moreover, the nanocomplex displayed good stability and pH-responsive behavior ex vivo. Both in vitro and in vivo results revealed that the nanocomplex could effectively cross the BBB and target glioma cells. Furthermore, the combination of chemotherapy and photothermal therapy of the nanocomplex achieved a better therapeutic effect, longer survival time, and minimized toxic side effects in orthotopic glioblastoma tumor-bearing nude mice. Overall, we modified the chemotherapeutic drug CPT so that it could self-assemble with other molecules into nanoparticles, which providing an alternative for the preparation of the carrier-free nanodrugs. The results highlighted the potential of self-assembly nanodrugs as a novel platform for effective glioblastoma therapy.
Collapse
|
85
|
Lin Z, Wen M, Yu E, Lin X, Wang H, Chen J, Yao C, Zhang H, Ru J, Wang K, Zhang Y, Huang L, Zhuge Q, Yang S. ANXA1 as a Prognostic and Immune Microenvironmental Marker for Gliomas Based on Transcriptomic Analysis and Experimental Validation. Front Cell Dev Biol 2021; 9:659080. [PMID: 34422796 PMCID: PMC8371204 DOI: 10.3389/fcell.2021.659080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 07/16/2021] [Indexed: 01/17/2023] Open
Abstract
The tumor microenvironment (TME) plays an important role in the growth and invasion of glioma. This study aimed to analyze the composition of the immune microenvironment in glioma samples and analyze the important differentially expressed genes to identify novel immune-targeted therapy for glioma. We downloaded transcriptomic data of 669 glioma samples from The Cancer Genome Atlas database. CIBERSORT and ESTIMATE methods were used to calculate the proportion of tumor-infiltrating immune cells and ratio of immune and stromal components in the TME. The differentially expressed genes (DEGs) were screened by comparing the genes expressed by both stromal and immune cells. Annexin A1 (ANXA1) was determined to be an important prognostic indicator through the common overlap of univariate Cox regression analysis and protein–protein interaction network analysis. The proportion of tumor-infiltrating immune cells, calculated by CIBERSORT algorithm, had a significant difference in distribution among the high and low ANXA1 expression groups, indicating that ANXA1 could be an important immune marker of TME. Furthermore, ANXA1 level was positively correlated with the histopathological factors and negatively related to the survival of glioma patients based on the analysis of multiple databases. Finally, in vitro experiments verified that antagonizing ANXA1 expression promoted cell apoptosis and inhibited the invasion and migration capacities of glioma cells. Therefore, ANXA1 due to its immune-related functions, can be an important prognostic indicator and immune microenvironmental marker for gliomas. Further studies are warranted to confirm ANXA1 as a potential immunotherapeutic target for gliomas.
Collapse
Affiliation(s)
- Zhongxiao Lin
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Min Wen
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Neurosurgery, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Enxing Yu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao Lin
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hua Wang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiayu Chen
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - ChaoJie Yao
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hengli Zhang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Junnan Ru
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kankai Wang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ying Zhang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lijie Huang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Su Yang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
86
|
Xu S, Zhong Y, Nie C, Pan Y, Adeli M, Haag R. Co-Delivery of Doxorubicin and Chloroquine by Polyglycerol Functionalized MoS2 Nanosheets for Efficient Multidrug-Resistant Cancer Therapy. Macromol Biosci 2021; 21:e2100233. [PMID: 34411417 DOI: 10.1002/mabi.202100233] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/04/2021] [Indexed: 01/11/2023]
Abstract
2D MoS2 has shown a great potential in biomedical applications, due to its superior loading capacity, photothermal property, and biodegradation. In this work, polyglycerol functionalized MoS2 nanosheets with photothermal and pH dual-stimuli responsive properties are used for the co-delivery of doxorubicin and chloroquine and treatment of multidrug-resistant HeLa (HeLa-R) cells. The polyglycerol functionalized MoS2 nanosheets with 80 nm average size show a high biocompatibility and loading efficiency (≈90%) for both drugs. The release of drugs from the nanosheets at pH 5.5 is significantly promoted by laser irradiation leading to efficient destruction of incubated HeLa-R cells. In vitro evaluation shows that the designed nanoplatform has a high ability to kill HeLa-R cells. Confocal experiments demonstrate that the synthesized drug delivery system enhances the cellular uptake of DOX via folic acid targeting ligand. Taking advantage of the combined properties including biocompatibility and targeting ability as well as high loading capacity and photothermal release, this multifunctional nanosystem is a promising candidate for anticancer therapy.
Collapse
Affiliation(s)
- Shaohui Xu
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Yinan Zhong
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Chuanxiong Nie
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Yuanwei Pan
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Mohsen Adeli
- Faculty of Science, Department of Chemistry, Lorestan University, Khorramabad, Iran
| | - Rainer Haag
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| |
Collapse
|
87
|
Chen Y, Guo M, Qu D, Liu Y, Guo J, Chen Y. Furin-responsive triterpenine-based liposomal complex enhances anticervical cancer therapy through size modulation. Drug Deliv 2021; 27:1608-1624. [PMID: 33179521 PMCID: PMC7676817 DOI: 10.1080/10717544.2020.1827086] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The accumulation and penetration of antitumor drugs in tumor tissues are directly related to their antitumor effects. The particle size of the nanodrug delivery system is one of the most important factors for the accumulation and penetration of antitumor drugs within tumor tissues. Generally, nanodelivery systems of intermediate size (100–120 nm) are capable of efficient accumulation owing to prolonged circulation and enhanced permeability and retention (EPR) effect; however, smaller ones (20–40 nm) are effective for deep penetration within tumor tissue. Currently a conventional drug delivery system cannot possess two types of optimal sizes, simultaneously. To solve this and to enhance cervical cancer treatment, a furin-responsive triterpenine-based liposomal complex (PEGcleavable Tf-CTM/L), with Tf-CTM (transferrin-modified tripterine-loaded coix seed oil microemulsion) in core, coated with a thermo-sensitive lipid and a kind of PEG shell modified with a furin-cleavable peptide was developed to improve tumor-specific accumulation and penetration. Herein, PEGcleavable Tf-CTM/L was capable of efficient accumulation because of EPR effect. The PEG shells could timely detach under stimulation of overexpressed furin protein to solve the problem of the steric hindrance dilemma. The small-sized Tf-CTM released under stimulation of tumor microthermal environment in cervical cancer, which was efficient with regards to deep penetration at tumor sites. Notably, compared to the use of triterpenine alone, PEGcleavable Tf-CTM/L promoted anticervical efficacy and displayed diminished systemic toxicity by efficient accumulation and deep penetration of antitumor drugs within tumor tissues. Our study provides a new strategy, and holds promising potential for anticervical cancer treatment.
Collapse
Affiliation(s)
- Yunyan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China.,School of Pharmacy,Wannan Medical College, Wuhu, China
| | - Mengfei Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China
| | - Ding Qu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China
| | - Yuping Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China
| | - Jian Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, China
| |
Collapse
|
88
|
Li Y, Gao S, Du X, Ji J, Xi Y, Zhai G. Advances in autophagy as a target in the treatment of tumours. J Drug Target 2021; 30:166-187. [PMID: 34319838 DOI: 10.1080/1061186x.2021.1961792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Autophagy is a multi-step lysosomal degradation process, which regulates energy and material metabolism and has been used to maintain homeostasis. Autophagy has been shown to be involved in the regulation of health and disease. But at present, there is no consensus on the relationship between autophagy and tumour, and we consider that it plays a dual role in the occurrence and development of tumour. That is to say, under certain conditions, it can inhibit the occurrence of tumour, but it can also promote the process of tumour. Therefore, autophagy could be used as a target for tumour treatment. The regulation of autophagy plays a synergistic role in the radiotherapy, chemotherapy, phototherapy and immunotherapy of tumour, and nano drug delivery system provides a promising strategy for improving the efficacy of autophagy regulation. This review summarised the progress in the regulatory pathways and factors of autophagy as well as nanoformulations as carriers for the delivery of autophagy modulators.
Collapse
Affiliation(s)
- Yingying Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Shan Gao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Xiyou Du
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Yanwei Xi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| |
Collapse
|
89
|
Protease-triggered bioresponsive drug delivery for the targeted theranostics of malignancy. Acta Pharm Sin B 2021; 11:2220-2242. [PMID: 34522585 PMCID: PMC8424222 DOI: 10.1016/j.apsb.2021.01.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/17/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Proteases have a fundamental role in maintaining physiological homeostasis, but their dysregulation results in severe activity imbalance and pathological conditions, including cancer onset, progression, invasion, and metastasis. This striking importance plus superior biological recognition and catalytic performance of proteases, combining with the excellent physicochemical characteristics of nanomaterials, results in enzyme-activated nano-drug delivery systems (nanoDDS) that perform theranostic functions in highly specific response to the tumor phenotype stimulus. In the tutorial review, the key advances of protease-responsive nanoDDS in the specific diagnosis and targeted treatment for malignancies are emphatically classified according to the effector biomolecule types, on the premise of summarizing the structure and function of each protease. Subsequently, the incomplete matching and recognition between enzyme and substrate, structural design complexity, volume production, and toxicological issues related to the nanocomposites are highlighted to clarify the direction of efforts in nanotheranostics. This will facilitate the promotion of nanotechnology in the management of malignant tumors.
Collapse
|
90
|
Wang J, Chen P, Dong Y, Xie H, Wang Y, Soto F, Ma P, Feng X, Du W, Liu BF. Designer exosomes enabling tumor targeted efficient chemo/gene/photothermal therapy. Biomaterials 2021; 276:121056. [PMID: 34364178 DOI: 10.1016/j.biomaterials.2021.121056] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/22/2021] [Accepted: 07/30/2021] [Indexed: 12/31/2022]
Abstract
Exosomes, endogenous nanosized particles (50-150 nm) secreted and absorbed by cells, have been recently used as diagnostic and therapeutic platforms in cancer treatment. The integration of exosome-based delivery with multiple therapeutic modalities could result in better clinical outcomes and reduced-sided effects. Here, we combined the targeting and biocompatibility of designer exosomes with chemo/gene/photothermal therapy. Our platform consists of exosomes loaded with internalized doxorubicin (DOX, a model cancer drug) and coated with magnetic nanoparticles conjugated with molecular beacons capable of targeting miR-21 for responsive molecular imaging. The coated magnetic nanoparticle enables enrichment of the exosomes at the tumor site by external magnetic field guidance. After the exosomes are gathered at the tumor site, the application of near-infrared radiation (NIR) induces localized hyperthermia and triggers the release of cargoes loaded inside the exosome. The released molecular beacon can target the miR-21 for both imaging and gene silencing. Meanwhile, the released doxorubicin serves to kill the cancer cells. About 91.04 % of cancer cells are killed after treatment with Exo-DOX-Fe3O4@PDA-MB under NIR. The ability of the exosome-based method for cancer therapy has been demonstrated by animal models, in which the tumor size is reduced dramatically by 97.57 % with a magnetic field-guided tumor-targeted chemo/gene/photothermal approach. Thus, we expected this designer exosome-mediated multi-mode therapy to be a promising platform for the next-generation precision cancer nanomedicines.
Collapse
Affiliation(s)
- Jie Wang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China; Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA, 94304, United States
| | - Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yue Dong
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China; Department of Mechanical and Automation Engineering, Chinese University of Hong Kong, Shatin, New Territories, 999077, Hong Kong, China
| | - Han Xie
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yachao Wang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Fernando Soto
- Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA, 94304, United States
| | - Peng Ma
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiaojun Feng
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Wei Du
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
91
|
Wang XL, Han X, Tang XY, Chen XJ, Li HJ. A Review of Off-On Fluorescent Nanoprobes: Mechanisms, Properties, and Applications. J Biomed Nanotechnol 2021; 17:1249-1272. [PMID: 34446130 DOI: 10.1166/jbn.2021.3117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
With the development of nanomaterials, fluorescent nanoprobes have attracted enormous attention in the fields of chemical sensing, optical materials, and biological detection. In this paper, the advantages of "off-on" fluorescent nanoprobes in disease detection, such as high sensitivity and short response time, are attentively highlighted. The characteristics, sensing mechanisms, and classifications of disease-related target substances, along with applications of these nanoprobes in cancer diagnosis and therapy are summarized systematically. In addition, the prospects of "off-on" fluorescent nanoprobe in disease detection are predicted. In this review, we presented information from all the papers published in the last 5 years discussing "off-on" fluorescent nanoprobes. This review was written in the hopes of being useful to researchers who are interested in further developing fluorescent nanoprobes. The characteristics of these nanoprobes are explained systematically, and data references and supports for biological analysis, clinical drug improvement, and disease detection have been provided appropriately.
Collapse
Affiliation(s)
- Xiao-Lin Wang
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China
| | - Xiao Han
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China
| | - Xiao-Ying Tang
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China
| | - Xiao-Jun Chen
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China
| | - Han-Jun Li
- School of Life Science, Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
92
|
Huang Z, Chen H, Ye H, Chen Z, Jaffrezic-Renault N, Guo Z. An ultrasensitive aptamer-antibody sandwich cortisol sensor for the noninvasive monitoring of stress state. Biosens Bioelectron 2021; 190:113451. [PMID: 34171819 DOI: 10.1016/j.bios.2021.113451] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 11/19/2022]
Abstract
Cortisol is a major glucocorticoid that can affect physiological activities in the human body. Besides, it is also a biomarker that can reflect the stress state of the body. Therefore, in order to monitor stress states in a sensitive and non-invasive manner, an ultra-sensitive aptamer-antibody sandwich sensor modified with multi-walled carbon nanotubes, ordered mesoporous carbon CMK-3, and silver nanoparticles (MWCNTs/CMK-3/AgNPs) was proposed for non-invasive detection of cortisol in human saliva. The MWCNTs/CMK-3/AgNPs nanocomposite was fixed on the surface of the glassy carbon electrodes (GCEs) as the material for the first round of signal amplification, and secondary signal amplification was realized by conjugating cortisol antibodies with gold nanoparticles (AuNPs). Finally, the aptamer-antibody sandwich pattern was used to specifically recognize and bind cortisol. The concentration response range for this aptamer-antibody sandwich sensor is 0.1 pg/mL-10 ng/mL, and the limit of detection (LOD) is 0.09 pg/mL. So far, the LOD of this sensor has been relatively low, showing its good sensitivity, selectivity, stability, and reproducibility. Furthermore, it has been successfully applied to detect cortisol in saliva samples to compare the stress states of postgraduates and undergraduates.
Collapse
Affiliation(s)
- Ziyu Huang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, 430065, PR China; School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, PR China
| | - Hao Chen
- Department of Anaesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Huarong Ye
- China Resources and Wisco General Hospital, Wuhan, 430080, PR China
| | - Zixuan Chen
- Department of Clinical Medicine, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, PR China
| | - Nicole Jaffrezic-Renault
- University of Lyon, Institute of Analytical Sciences, UMR-CNRS 5280, 5, La Doua Street, Villeurbanne, 69100, France.
| | - Zhenzhong Guo
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, 430065, PR China.
| |
Collapse
|
93
|
He JS, Liu SJ, Zhang YR, Chu XD, Lin ZB, Zhao Z, Qiu SH, Guo YG, Ding H, Pan YL, Pan JH. The Application of and Strategy for Gold Nanoparticles in Cancer Immunotherapy. Front Pharmacol 2021; 12:687399. [PMID: 34163367 PMCID: PMC8215714 DOI: 10.3389/fphar.2021.687399] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/24/2021] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy of malignant tumor is a verified and crucial anti-tumor strategy to help patients with cancer for prolonging prognostic survival. It is a novel anticancer tactics that activates the immune system to discern and damage cancer cells, thereby prevent them from proliferating. However, immunotherapy still faces many challenges in view of clinical efficacy and safety issues. Various nanomaterials, especially gold nanoparticles (AuNPs), have been developed not only for anticancer treatment but also for delivering antitumor drugs or combining other treatment strategies. Recently, some studies have focused on AuNPs for enhancing cancer immunotherapy. In this review, we summarized how AuNPs applicated as immune agents, drug carriers or combinations with other immunotherapies for anticancer treatment. AuNPs can not only act as immune regulators but also deliver immune drugs for cancer. Therefore, AuNPs are candidates for enhancing the efficiency and safety of cancer immunotherapy.
Collapse
Affiliation(s)
- Jia-Shuai He
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shi-Jin Liu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yi-Ran Zhang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiao-Dong Chu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zheng-Bin Lin
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhan Zhao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Sheng-Hui Qiu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yan-Guan Guo
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hui Ding
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yun-Long Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jing-Hua Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
94
|
Xie R, Ruan S, Liu J, Qin L, Yang C, Tong F, Lei T, Shevtsov M, Gao H, Qin Y. Furin-instructed aggregated gold nanoparticles for re-educating tumor associated macrophages and overcoming breast cancer chemoresistance. Biomaterials 2021; 275:120891. [PMID: 34051669 DOI: 10.1016/j.biomaterials.2021.120891] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 12/26/2022]
Abstract
Insufficient drug accumulation and chemoresistance remain two major challenges in cancer chemotherapy. Herein, we designed a furin-responsive aggregated nanoplatform loaded with doxorubicin (DOX) and hydroxychloroquine (HCQ) (AuNPs-D&H-R&C) to combine chemotherapy, autophagy inhibition and macrophage polarization. AuNPs-D&H-R&C could passively target breast tumor via enhanced permeability and retention (EPR) effect after systemic administration and further aggregate together triggered by furin overexpressed in breast cancer. The in situ aggregations hindered the back-flow of NPs to the bloodstream and exocytosis of tumor cells, leading to enhanced drug accumulation within tumors. Moreover, upon exposure to acidic pH in the endosomes/lysosomes, HCQ was efficiently released and it inhibited autophagy and thus restored the sensitivity of tumor cell to DOX. Meanwhile, autophagy inhibition could reprogram tumor-promoting M2-like TAMs to anti-tumor M1 phenotype, exerting a synergistic effect in overcoming chemoresistance. In vitro studies demonstrated the superiority of furin-triggered aggregated AuNPs delivery system in enhancing drug accumulation in breast tumor, compared with PEGlyated AuNPs. The co-delivery of DOX and HCQ showed much improved chemotherapeutic efficiency to chemoresistant MCF-7/ADR breast tumor, in large part due to macrophage polarization. In conclusion, we developed a stimulus-responsive delivery system and proposed a potential combination strategy to overcome chemoresistance in cancer chemotherapy.
Collapse
Affiliation(s)
- Rou Xie
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Shaobo Ruan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Jiaqi Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Lin Qin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Chuanyao Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Fan Tong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Ting Lei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Maxim Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, 194064, Russia
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.
| | - Yi Qin
- Department of Orthopedics, Zhuhai Hospital, Jinan University, Zhuhai People's Hospital, 79 Kangning Road, Zhuhai, 519000, China.
| |
Collapse
|
95
|
Xiao W, Wang Y, Zhang H, Liu Y, Xie R, He X, Zhou Y, Liang L, Gao H. The protein corona hampers the transcytosis of transferrin-modified nanoparticles through blood-brain barrier and attenuates their targeting ability to brain tumor. Biomaterials 2021; 274:120888. [PMID: 34029915 DOI: 10.1016/j.biomaterials.2021.120888] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 04/20/2021] [Accepted: 05/04/2021] [Indexed: 02/07/2023]
Abstract
The modification of targeting ligands on nanoparticles (NPs) is anticipated to enhance the delivery of therapeutics to diseased tissues. However, once exposed to the blood stream, NPs can immediately adsorb proteins to form the "protein corona," which may greatly hinder the targeting ligand from binding to its receptor. For brain-targeting delivery, nanotherapeutics must traverse the blood-brain barrier (BBB) to enter the brain parenchyma and then target the diseased cells. However, it remains elusive whether, apart from receptor recognition, the protein corona can affect other processes involved in BBB transcytosis, such as endocytosis, intracellular trafficking, and exocytosis. Furthermore, the targeting ability of NPs toward diseased cells after transcytosis remains unclear. Herein, transferrin (Tf), a brain-targeting ligand, was coupled to NPs to evaluate BBB transcytosis and brain tumor targeting ability. Different impacts of the in vitro and in vivo protein corona on receptor targeting, lysosomal escape, and BBB transcytosis were found. The in vitro protein corona abolished the Tf-mediated effects of the abovementioned processes, whereas the in vivo protein corona attenuated these effects. After crossing the BBB, Tf retained its targeting specificity towards brain tumor cells. Together, these results revealed that several bound apolipoproteins, especially apolipoprotein A-I, may help NPs traverse the BBB, thereby providing novel insights into the development of brain-targeted delivery.
Collapse
Affiliation(s)
- Wei Xiao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, PR China
| | - Yazhen Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, PR China
| | - Huilin Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, PR China
| | - Yuwei Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, PR China
| | - Rou Xie
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, PR China
| | - Xueqin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, PR China
| | - Yang Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, PR China
| | - Luqing Liang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, PR China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
96
|
Zhang W, Lin Y. The Mechanism of Asparagine Endopeptidase in the Progression of Malignant Tumors: A Review. Cells 2021; 10:cells10051153. [PMID: 34068767 PMCID: PMC8151911 DOI: 10.3390/cells10051153] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/24/2021] [Accepted: 05/07/2021] [Indexed: 12/20/2022] Open
Abstract
Asparagine endopeptidase (AEP), also called legumain, is currently the only known cysteine protease that specifically cleaves peptide bonds in asparaginyl residue in the mammalian genome. Since 2003, AEP has been reported to be widely expressed in a variety of carcinomas and is considered a potential therapeutic target. In the following years, researchers intensively investigated the substrates of AEP and the mechanism of AEP in partial tumors. With the identification of substrate proteins such as P53, integrin αvβ3, MMP-2, and MMP-9, the biochemical mechanism of AEP in carcinomas is also more precise. This review will clarify the probable mechanisms of AEP in the progression of breast carcinoma, glioblastoma, gastric carcinoma, and epithelial ovarian carcinoma. This review will also discuss the feasibility of targeted therapy with AEP inhibitor (AEPI) in these carcinomas.
Collapse
|
97
|
Fan Y, Cui Y, Hao W, Chen M, Liu Q, Wang Y, Yang M, Li Z, Gong W, Song S, Yang Y, Gao C. Carrier-free highly drug-loaded biomimetic nanosuspensions encapsulated by cancer cell membrane based on homology and active targeting for the treatment of glioma. Bioact Mater 2021; 6:4402-4414. [PMID: 33997516 PMCID: PMC8111096 DOI: 10.1016/j.bioactmat.2021.04.027] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 12/15/2022] Open
Abstract
Nanosuspensions, as a new drug delivery system for insoluble drugs, are only composed of a drug and a small amount of stabilizer, which is dispersed in an aqueous solution with high drug-loading, small particle size, high dispersion, and large specific surface area. It can significantly improve the dissolution, bioavailability, and efficacy of insoluble drugs. In this study, paclitaxel nanosuspensions ((PTX)NS) were prepared by an ultrasonic precipitation method, with the characteristics of simple preparation and easy repetition. With the help of a homologous targeting mechanism, a kind of glioma C6 cancer cell membrane (CCM)-coated (PTX)NS was developed and modified with DWSW peptide to obtain DWSW-CCM-(PTX)NS with the functions of BBB penetration and tumor targeting. The results showed that the cancer cell membrane could effectively camouflage the nanosuspensions so that it was not cleared by the immune system and could cross the blood-brain-barrier (BBB) and selectively target tumor tissues. Cell uptake experiments and in vivo imaging confirmed that the uptake of DWSW-CCM-(PTX)NS by tumor cells and the distribution in intracranial gliomas increased. Cytotoxicity test and in vivo anti-glioma studies showed that DWSW-CCM-(PTX)NS could significantly inhibit the growth of glioma cells and significantly prolong the survival time of glioma-bearing mice. Finally, the cancer cell membrane coating endowed the nanosuspensions with the biological properties of homologous adhesion and immune escape. This study provides an integrated solution for improving the targeting of nanosuspensions and demonstrates the encouraging potential of biomimetic nanosuspensions applicable to tumor therapy. Paclitaxel nanosuspensions with high drug-loading and without carrier. Biomimetic nanosuspensions wrapped by peptide-modified cancer cell membranes. Penetrate BBB and BBTB to transport drugs to glioma. Dual effects of active and homology targeting improve therapeutic efficiency.
Collapse
Affiliation(s)
- Yueyue Fan
- College of Pharmacy, Henan University, Kaifeng, 475000, PR China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Yuexin Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Wenyan Hao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Mengyu Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Qianqian Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Yuli Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Meiyan Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Zhiping Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Wei Gong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Shiyong Song
- College of Pharmacy, Henan University, Kaifeng, 475000, PR China
| | - Yang Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| | - Chunsheng Gao
- College of Pharmacy, Henan University, Kaifeng, 475000, PR China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, PR China
| |
Collapse
|
98
|
Khan NU, Ni J, Ju X, Miao T, Chen H, Han L. Escape from abluminal LRP1-mediated clearance for boosted nanoparticle brain delivery and brain metastasis treatment. Acta Pharm Sin B 2021; 11:1341-1354. [PMID: 34094838 PMCID: PMC8148067 DOI: 10.1016/j.apsb.2020.10.015] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/14/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
Breast cancer brain metastases (BCBMs) are one of the most difficult malignancies to treat due to the intracranial location and multifocal growth. Chemotherapy and molecular targeted therapy are extremely ineffective for BCBMs due to the inept brain accumulation because of the formidable blood‒brain barrier (BBB). Accumulation studies prove that low density lipoprotein receptor-related protein 1 (LRP1) is promising target for BBB transcytosis. However, as the primary clearance receptor for amyloid beta and tissue plasminogen activator, LRP1 at abluminal side of BBB can clear LRP1-targeting therapeutics. Matrix metalloproteinase-1 (MMP1) is highly enriched in metastatic niche to promote growth of BCBMs. Herein, it is reported that nanoparticles (NPs-K-s-A) tethered with MMP1-sensitive fusion peptide containing HER2-targeting K and LRP1-targeting angiopep-2 (A), can surmount the BBB and escape LRP1-mediated clearance in metastatic niche. NPs-K-s-A revealed infinitely superior brain accumulation to angiopep-2-decorated NPs-A in BCBMs bearing mice, while comparable brain accumulation in normal mice. The delivered doxorubicin and lapatinib synergistically inhibit BCBMs growth and prolongs survival of mice bearing BCBMs. Due to the efficient BBB penetration, special and remarkable clearance escape, and facilitated therapeutic outcome, the fusion peptide-based drug delivery strategy may serve as a potential approach for clinical management of BCBMs.
Collapse
Key Words
- 231Br, MDA-MB-231Br-HER2
- A, angiopep-2
- AUC0‒t, area under the curve from zero to time t
- Abluminal LRP1
- Amyloid beta
- Aβ, amyloid beta
- BBB, blood‒brain barrier
- BCBMs, breast cancer brain metastases
- BMECs, brain microvascular endothelial cells
- Blood‒brain barrier
- Brain clearance
- Breast cancer brain metastases
- CI, combination index
- CL, clearance
- DMEM, Dulbecco's modified eagle medium
- DMSO, dimethyl sulfoxide
- DOX, doxorubicin
- FBS, fetal bovine serum
- Fa, the fraction of tumor cells affected
- Fusion peptide
- K, KAAYSL
- LAP, lapatinib
- LRP1, low density lipoprotein receptor-related protein 1
- MAL-PEG-SCM, maleimide polyethylene glycol succinimidyl carboxymethyl ester
- MCM, MDA-MB-231Br-HER2 conditioned medium
- MMP
- MMP1, matrix metalloproteinase-1
- MRT0‒t, mean residence time from zero to time t
- NPs, nanoparticles
- Nanoparticles
- PLGA, poly(lactic-co-glycolic acid)
- PLGA-PLL, poly(lactic-co-glycolic acid)-poly(ε-carbobenzoxy-l-lysine)
- PLL, poly(ε-carbobenzoxy-l-lysine)
- PVA, polyvinyl alcohol
- SDS, sodium dodecyl sulfate
- i, insensitive GDQGIAGF
- s, sensitive VPMS-MRGG
- t1/2, half time
- tPA, tissue plasminogen activator
Collapse
|
99
|
Xiong Y, Li M, Bai J, Sheng Y, Zhang Y. High Level of METTL7B Indicates Poor Prognosis of Patients and Is Related to Immunity in Glioma. Front Oncol 2021; 11:650534. [PMID: 33996568 PMCID: PMC8117938 DOI: 10.3389/fonc.2021.650534] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/06/2021] [Indexed: 01/22/2023] Open
Abstract
Glioma is the most common primary intracranial malignant tumor in adults. Although there have been many efforts on potential targeted therapy of glioma, the patient’s prognosis remains dismal. Methyltransferase Like 7B (METTL7B) has been found to affect the development of a variety of tumors. In this study, we collected RNA-seq data of glioma in CGGA and TCGA, analyzed them separately. Then, Kaplan-Meier survival analysis, univariate and multivariate Cox analysis, and receiver operating characteristic curve (ROC curve) analysis were used to evaluate the effect of METTL7B on prognosis. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Set Enrichment Analysis (GSEA) enrichment analyses were used to identify the function or pathway associated with METTL7B. Moreover, the ESTIMATE algorithm, Cibersort algorithm, Spearman correlation analysis, and TIMER database were used to explore the relationship between METTL7B and immunity. Finally, the role of METTL7B was explored in glioma cells. We found that METTL7B is highly expressed in glioma, and high expression of METTL7B in glioma is associated with poor prognosis. In addition, there were significant differences in immune scores and immune cell infiltration between the two groups with different expression levels of METTL7B. Moreover, METTL7B was also correlated with immune checkpoints. Knockdown of METTL7B revealed that METTL7B promoted the progression of glioma cells. The above results indicate that METTL7B affects the prognosis of patients and is related to tumor immunity, speculating that METTL7B may be a new immune-related target for the treatment of glioma.
Collapse
Affiliation(s)
- Yujia Xiong
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Mingxuan Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jiwei Bai
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yutao Sheng
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Institute for Brain Disorders Brain Tumor Center, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
100
|
Zhou L, Huo M, Qian X, Ding L, Yu L, Feng W, Cui X, Chen Y. Autophagy blockade synergistically enhances nanosonosensitizer-enabled sonodynamic cancer nanotherapeutics. J Nanobiotechnology 2021; 19:112. [PMID: 33879173 PMCID: PMC8056542 DOI: 10.1186/s12951-021-00855-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/07/2021] [Indexed: 01/07/2023] Open
Abstract
Ultrasound-triggered sonodynamic therapy (SDT) represents an emerging therapeutic modality for cancer treatment based on its specific feature of noninvasiveness, high tissue-penetrating depth and desirable therapeutic efficacy, but the SDT-induced pro-survival cancer-cell autophagy would significantly lower the SDT efficacy for cancer treatment. Here we propose an "all-in-one" combined tumor-therapeutic strategy by integrating nanosonosensitizers-augmented noninvasive SDT with autophagy inhibition based on the rationally constructed nanoliposomes that co-encapsulates clinically approved sonosensitizers protoporphyrin IX (PpIX) and early-phase autophagy-blocking agent 3-methyladenine (3-MA). It has been systematically demonstrated that nanosonosensitizers-augmented SDT induced cytoprotective pro-survival autophagy through activation of MAPK signaling pathway and inhibition of AMPK signaling pathway, and this could be efficaciously inhibited by 3-MA in early-phase autophagy, which significantly decreased the cell resistance to intracellular oxidative stress and complied a remarkable synergistic effect on SDT medicated cancer-cell apoptosis both in vitro at cellular level and in vivo on tumor-bearing animal model. Therefore, our results provide a proof-of-concept combinatorial tumor therapeutics based on nanosonosensitizers for the treatment of ROS-resistant cancer by autophagy inhibition-augmented SDT.
Collapse
Affiliation(s)
- Liqiang Zhou
- Sino-German Tongji-Caritas Research Center of Ultrasound in Medicine, Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Minfeng Huo
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China
| | - Xiaoqin Qian
- Department of Ultrasound, Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, People's Republic of China
| | - Li Ding
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China.
| | - Luodan Yu
- School of Life Sciences, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Wei Feng
- School of Life Sciences, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Xinwu Cui
- Sino-German Tongji-Caritas Research Center of Ultrasound in Medicine, Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Yu Chen
- School of Life Sciences, Shanghai University, Shanghai, 200444, People's Republic of China.
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, People's Republic of China.
| |
Collapse
|