201
|
Zeng Y, Li S, Zhang S, Wang L, Yuan H, Hu F. Cell membrane coated-nanoparticles for cancer immunotherapy. Acta Pharm Sin B 2022; 12:3233-3254. [PMID: 35967284 PMCID: PMC9366230 DOI: 10.1016/j.apsb.2022.02.023] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/14/2021] [Accepted: 02/19/2022] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy can effectively inhibit cancer progression by activating the autoimmune system, with low toxicity and high effectiveness. Some of cancer immunotherapy had positive effects on clinical cancer treatment. However, cancer immunotherapy is still restricted by cancer heterogeneity, immune cell disability, tumor immunosuppressive microenvironment and systemic immune toxicity. Cell membrane-coated nanoparticles (CMCNs) inherit abundant source cell-relevant functions, including “self” markers, cross-talking with the immune system, biological targeting, and homing to specific regions. These enable them to possess preferred characteristics, including better biological compatibility, weak immunogenicity, immune escaping, a prolonged circulation, and tumor targeting. Therefore, they are applied to precisely deliver drugs and promote the effect of cancer immunotherapy. In the review, we summarize the latest researches of biomimetic CMCNs for cancer immunotherapy, outline the existing specific cancer immune therapies, explore the unique functions and molecular mechanisms of various cell membrane-coated nanoparticles, and analyze the challenges which CMCNs face in clinical translation.
Collapse
|
202
|
Guo Y, Wang Z, Shi X, Shen M. Engineered cancer cell membranes: An emerging agent for efficient cancer theranostics. EXPLORATION (BEIJING, CHINA) 2022; 2:20210171. [PMID: 37324583 PMCID: PMC10190949 DOI: 10.1002/exp.20210171] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/24/2021] [Indexed: 06/15/2023]
Abstract
For efficient cancer theranostics, surface modification of nanomaterials plays an important role in improving targeting ability and reducing the non-specific interactions with normal tissues. Recently, the biomimetic technology represented by coating of cancer cell membranes (CCMs) has been regarded as a promising method to strengthen the biocompatibility and targeting specificity of nanomaterials. Furthermore, the engineered CCMs (ECCMs) integrated with the natural biological properties of CCMs and specific functions from other cells or proteins have offered more possibilities in the field of cancer theranostics. Herein, the recent progresses in the design and preparation of ECCMs are summarized, and the applications of ECCMs in targeting delivery, activation of immunity, and detection of circulating tumor cells are reviewed. Finally, the current challenges and future perspectives with regard to the development of ECCMs are briefly discussed.
Collapse
Affiliation(s)
- Yunqi Guo
- Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and BiotechnologyDonghua UniversityShanghaiP. R. China
| | - Zhiqiang Wang
- Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and BiotechnologyDonghua UniversityShanghaiP. R. China
| | - Xiangyang Shi
- Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and BiotechnologyDonghua UniversityShanghaiP. R. China
| | - Mingwu Shen
- Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and BiotechnologyDonghua UniversityShanghaiP. R. China
| |
Collapse
|
203
|
Homotypic Cancer Cell Membranes Camouflaged Nanoparticles for Targeting Drug Delivery and Enhanced Chemo-Photothermal Therapy of Glioma. Pharmaceuticals (Basel) 2022; 15:ph15020157. [PMID: 35215270 PMCID: PMC8879672 DOI: 10.3390/ph15020157] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/13/2022] Open
Abstract
Glioma is among the deadliest types of brain cancer, for which there currently is no effective treatment. Chemotherapy is mainstay in the treatment of glioma. However, drug tolerance, non-targeting, and poor blood–brain barrier penetrance severely inhibits the efficacy of chemotherapeutics. An improved treatment method is thus urgently needed. Herein, a multifunctional biomimetic nanoplatform was developed by encapsulating graphene quantum dots (GQDs) and doxorubicin (DOX) inside a homotypic cancer cell membrane (CCM) for targeted chemo-photothermal therapy of glioma. The GQDs with stable fluorescence and a superior light-to-heat conversion property were synthesized as photothermal therapeutic agents and co-encapsulated with DOX in CCM. The as-prepared nanoplatform exhibited a high DOX loading efficiency. The cell membrane coating protected drugs from leakage. Upon an external laser stimuli, the membrane could be destroyed, resulting in rapid DOX release. By taking advantage of the homologous targeting of the cancer cell membrane, the GQDs/DOX@CCM were found to actively target tumor cells, resulting in significantly enhanced cellular uptake. Moreover, a superior suppression efficiency of GQDs/DOX@CCM to cancer cells through chemo-photothermal treatment was also observed. The results suggest that this biomimetic nanoplatform holds potential for efficient targeting of drug delivery and synergistic chemo-photothermal therapy of glioma.
Collapse
|
204
|
Wei Y, Wang Z, Yang J, Xu R, Deng H, Ma S, Fang T, Zhang J, Shen Q. Reactive oxygen species / photothermal therapy dual-triggered biomimetic gold nanocages nanoplatform for combination cancer therapy via ferroptosis and tumor-associated macrophage repolarization mechanism. J Colloid Interface Sci 2022; 606:1950-1965. [PMID: 34695762 DOI: 10.1016/j.jcis.2021.09.160] [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: 06/17/2021] [Revised: 09/06/2021] [Accepted: 09/26/2021] [Indexed: 12/12/2022]
Abstract
With the continuous development of cancer nanotechnology, an important trend in the research is to combine the broad application prospects of functional nanomaterials with recent biological discoveries and technological advances. Herein, a cancer cell membrane-camouflaged gold nanocage loading doxorubicin (DOX) and l-buthionine sulfoximine (BSO) (denoted as m@Au-D/B NCs) was constructed as an innovative nanoplatform to confer promising cancer combination therapy by evoking effective ferroptosis and immune responses. Briefly, the loading of BSO and DOX could induce ferroptosis through simultaneous effective glutathione (GSH) consumption and reactive oxygen species (ROS) accumulation. Gold nanocages (AuNCs) with distinct anti-tumor application performance was utilized as ideal nanocarrier for drug loading, evoking photothermal effects and photochemical catalysis to generate more ROS under near-infrared (NIR) irradiation. Moreover, m@Au-D/B NCs-mediated photothermal therapy (PTT) combined with ROS production could repolarize the tumor-associated macrophages (TAMs) from pro-tumor (M2) phenotype to anti-tumor (M1) phenotype, thus improving tumor-suppressive immune environment and then promoting the activation of effector cells and release of pro-inflammatory cytokines, in which the antitumor responses were evoked robustly in a methodical approach. The anti-tumor effects in vivo implied that m@Au-D/B NCs could significantly inhibit tumor growth without severe toxicity. Hence, this homotypic targeting nanosystem could offer an auspicious anticancer access by triggering combination cancer therapy via ferroptosis and tumor-associated macrophage repolarization mechanism.
Collapse
Affiliation(s)
- Yawen Wei
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Zhihua Wang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Jie Yang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Rui Xu
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Huizi Deng
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Siyu Ma
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Tianxu Fang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Jun Zhang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Qi Shen
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| |
Collapse
|
205
|
Xiong J, Wu M, Chen J, Liu Y, Chen Y, Fan G, Liu Y, Cheng J, Wang Z, Wang S, Liu Y, Zhang W. Cancer-Erythrocyte Hybrid Membrane-Camouflaged Magnetic Nanoparticles with Enhanced Photothermal-Immunotherapy for Ovarian Cancer. ACS NANO 2021; 15:19756-19770. [PMID: 34860006 DOI: 10.1021/acsnano.1c07180] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Cell-membrane-coated nanoparticles are widely studied due to their inherent cellular properties, such as immune escape and homologous homing. A cell membrane coating can also maintain the relative stability of nanoparticles during circulation in a complex blood environment through cell membrane encapsulation technology. In this study, we fused a murine-derived ID8 ovarian cancer cell membrane with a red blood cell (RBC) membrane to create a hybrid biomimetic coating (IRM), and hybrid IRM camouflaged indocyanine green (ICG)-loaded magnetic nanoparticles (Fe3O4-ICG@IRM) were fabricated for combination therapy of ovarian cancer. Fe3O4-ICG@IRM retained both ID8 and RBC cell membrane proteins and exhibited highly specific self-recognition of ID8 cells in vitro and in vivo as well as a prolonged circulation lifetime in blood. Interestingly, in the bilateral flank tumor model, the IRM-coated nanoparticles also activated specific immunity, which killed homologous ID8 tumor cells but had no effect on B16-F10 tumor cells. Furthermore, Fe3O4-ICG@IRM showed synergistic photothermal therapy, resulting in the release of whole-cell tumor antigens by photothermal-induced tumor necrosis, which further enhanced antitumor immunotherapy for primary tumor and metastatic tumor by activating CD8+ cytotoxic T cells and reducing regulatory Foxp3+ T cells. Together, the biomimetic Fe3O4-ICG@IRM nanoparticles showed synergistic photothermal-immunotherapy for ovarian cancer.
Collapse
Affiliation(s)
- Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, China
| | - Jilei Chen
- Department of Chemistry, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Yaofa Liu
- Department of Chemistry, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Yurou Chen
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Guanlan Fan
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yanyan Liu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Jing Cheng
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zhenhua Wang
- Institute of Flexible Electronics, Institute of Biomedical Materials and Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, China
| | - Yi Liu
- Department of Chemistry, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
- State Key Laboratory of Separation Membranes and Membrane Process, School of Chemistry, Tiangong University, Tianjin 300387, China
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
206
|
Qu Y, Chu B, Wei X, Chen Y, Yang Y, Hu D, Huang J, Wang F, Chen M, Zheng Y, Qian Z. Cancer-Cell-Biomimetic Nanoparticles for Targeted Therapy of Multiple Myeloma Based on Bone Marrow Homing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 34:e2107883. [PMID: 34877715 DOI: 10.1002/adma.202107883] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/05/2021] [Indexed: 02/05/2023]
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy. It is characterized by abnormal transformation and uncontrolled clonal proliferation of malignant plasma cells in the bone marrow (BM), which can destroy bone structure and inhibit hematopoiesis. Although there are new therapeutic methods, they are not curative, mainly because it is difficult to deliver an effective amount of drug to BM, leading to a failure to eradicate MM cells inside the BM. BM homing is an important and unique characteristic of MM cells and it is mainly affected by surface molecules on the tumor cell membrane. Inspired by this mechanism, an MM-mimicking nanocarrier is developed by coating bortezomib (BTZ)-loaded poly(ε-caprolactone)-poly(ethylene glycol)-poly(ε-caprolactone) (PCEC) nanoparticles with the MM cell membrane. The MM-mimicking nanoparticles can enter the BM based on BM homing as a "Trojan horse" and target the tumor cells through homologous targeting. In this way, drug availability at the myeloma site is enhanced so as to inhibit MM growth. In addition, these MM-mimicking nanoparticles can escape phagocytosis by the MPS and have a long circulation effect. The in vivo therapeutic results demonstrate an excellent treatment efficacy for MM. Accordingly, this strategy may be a promising platform for the treatment of MM.
Collapse
Affiliation(s)
- Ying Qu
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan 610041 P. R. China
- Department of Hematology and Institute of Hematology West China Hospital Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Bingyang Chu
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Xue Wei
- Department of Hematology and Institute of Hematology West China Hospital Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Yingying Chen
- Department of Hematology and Institute of Hematology West China Hospital Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Yun Yang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Danrong Hu
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Jingcao Huang
- Department of Hematology and Institute of Hematology West China Hospital Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Fangfang Wang
- Department of Hematology and Institute of Hematology West China Hospital Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Mengran Chen
- Department of Hematology and Institute of Hematology West China Hospital Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Yuhuan Zheng
- Department of Hematology and Institute of Hematology West China Hospital Sichuan University Chengdu Sichuan 610041 P. R. China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan 610041 P. R. China
| |
Collapse
|
207
|
Zhu R, Lang T, Yin Q, Li Y. Nano drug delivery systems improve metastatic breast cancer therapy. MEDICAL REVIEW (BERLIN, GERMANY) 2021; 1:244-274. [PMID: 37724299 PMCID: PMC10388745 DOI: 10.1515/mr-2021-0011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/03/2021] [Indexed: 09/20/2023]
Abstract
Despite continual progress in the technologies and regimens for cancer therapy, the treatment outcome of fatal metastatic breast cancer is far from satisfactory. Encouragingly, nanotechnology has emerged as a valuable tool to optimize drug delivery process in cancer therapy via preventing the cargos from degradation, improving the tumor-targeting efficiency, enhancing therapeutic agents' retention in specific sites, and controlling drug release. In the last decade, several mechanisms of suppressing tumor metastasis by functional nano drug delivery systems (NDDSs) have been revealed and a guidance for the rational design of anti-metastasis NDDSs is summarized, which consist of three aspects: optimization of physiochemical properties, tumor microenvironment remodeling, and biomimetic strategies. A series of medicinal functional biomaterials and anti-metastatic breast cancer NDDSs constructed by our team are introduced in this review. It is hoped that better anti-metastasis strategies can be inspired and applied in clinic.
Collapse
Affiliation(s)
- Runqi Zhu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tianqun Lang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong Province, China
| | - Qi Yin
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong Province, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Bohai rim Advanced Research Institute for Drug Discovery, Yantai, Shandong Province, China
- School of Pharmacy, Yantai University, Yantai, Shandong Province, China
| |
Collapse
|
208
|
Liang L, Wen L, Weng Y, Song J, Li H, Zhang Y, He X, Zhao W, Zhan M, Li Y, Lu L, Xin Y, Lu C. Homologous-targeted and tumor microenvironment-activated hydroxyl radical nanogenerator for enhanced chemoimmunotherapy of non-small cell lung cancer. CHEMICAL ENGINEERING JOURNAL 2021; 425:131451. [DOI: 10.1016/j.cej.2021.131451] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/11/2024]
|
209
|
Kumar N, Fazal S, Miyako E, Matsumura K, Rajan R. Avengers against cancer: A new era of nano-biomaterial-based therapeutics. MATERIALS TODAY 2021; 51:317-349. [DOI: 10.1016/j.mattod.2021.09.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
210
|
Yang Y, Wang K, Pan Y, Rao L, Luo G. Engineered Cell Membrane-Derived Nanoparticles in Immune Modulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102330. [PMID: 34693653 PMCID: PMC8693058 DOI: 10.1002/advs.202102330] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/19/2021] [Indexed: 05/26/2023]
Abstract
Immune modulation is one of the most effective approaches in the therapy of complex diseases, including public health emergency. However, most immune therapeutics such as drugs, vaccines, and cellular therapy suffer from the limitations of poor efficacy and adverse side effects. Fortunately, cell membrane-derived nanoparticles (CMDNs) have superior compatibility with other therapeutics and offer new opportunities to push the limits of current treatments in immune modulation. As the interface between cells and outer surroundings, cell membrane contains components which instruct intercellular communication and the plasticity of cytomembrane has significantly potentiated CMDNs to leverage our immune system. Therefore, cell membranes employed in immunomodulatory CMDNs have gradually shifted from natural to engineered. In this review, unique properties of immunomodulatory CMDNs and engineering strategies of emerging CMDNs for immune modulation, with an emphasis on the design logic are summarized. Further, this review points out some pressing problems to be solved during clinical translation and put forward some suggestions on the prospect of immunoregulatory CMDNs. It is anticipated that this review can provide new insights on the design of immunoregulatory CMDNs and expand their potentiation in the precise control of the dysregulated immune system.
Collapse
Affiliation(s)
- Yixiao Yang
- Institute of Burn ResearchThe First Affiliated HospitalState Key Lab of TraumaBurn and Combined InjuryChongqing Key Laboratory for Disease ProteomicsThird Military Medical University (Army Medical University)Chongqing400038China
| | - Kai Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS)School of Basic Medical Sciences and Shanghai Public Health Clinical CenterShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Yuanwei Pan
- Institute of Biomedical Health Technology and EngineeringShenzhen Bay LaboratoryShenzhen518132China
| | - Lang Rao
- Institute of Biomedical Health Technology and EngineeringShenzhen Bay LaboratoryShenzhen518132China
| | - Gaoxing Luo
- Institute of Burn ResearchThe First Affiliated HospitalState Key Lab of TraumaBurn and Combined InjuryChongqing Key Laboratory for Disease ProteomicsThird Military Medical University (Army Medical University)Chongqing400038China
| |
Collapse
|
211
|
Guo K, Xiao N, Liu Y, Wang Z, Tóth J, Gyenis J, Thakur VK, Oyane A, Shubhra QT. Engineering polymer nanoparticles using cell membrane coating technology and their application in cancer treatments: Opportunities and challenges. NANO MATERIALS SCIENCE 2021. [DOI: 10.1016/j.nanoms.2021.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
212
|
Huang X, Chen L, Lin Y, Tou KIP, Cai H, Jin H, Lin W, Zhang J, Cai J, Zhou H, Pi J. Tumor targeting and penetrating biomimetic mesoporous polydopamine nanoparticles facilitate photothermal killing and autophagy blocking for synergistic tumor ablation. Acta Biomater 2021; 136:456-472. [PMID: 34562660 DOI: 10.1016/j.actbio.2021.09.030] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/23/2021] [Accepted: 09/16/2021] [Indexed: 12/12/2022]
Abstract
The synergistic manipulation of autophagy blocking with tumor targeting and penetration effects to enhance cancer cell killing during photothermal therapy (PTT) remains a substantial challenge. Herein, we fabricated a biomimetic nanoplatform by precisely coating homologous prostate cancer cell membranes (CMs) onto the surface of mesoporous polydopamine nanoparticles (mPDA NPs) encapsulating the autophagy inhibitor chloroquine (CQ) for synergistically manipulating PTT and autophagy for anticancer treatment. The resulting biomimetic mPDA@CMs NPs-CQ system could escape macrophage phagocytosis, overcome the vascular barrier, and home in the homologous prostate tumor xenograft with high tumor targeting and penetrating efficiency. The mPDA NPs core endowed the mPDA@CMs NPs-CQ with good photothermal capability to mediate PTT killing of prostate cancer cells, while NIR-triggered CQ release from the nanosystem further arrested PTT-induced protective autophagy of cancer cells, thus weakening the resistance of prostate cancer cells to PTT. This combined PTT killing and autophagy blocking anticancer strategy could induce significant autophagosome accumulation, ROS generation, mitochondrial damage, endoplasmic reticulum stress, and apoptotic signal transduction, which finally results in synergistic prostate tumor ablation in vivo. This prostate cancer biomimetic nanosystem with synergistically enhanced anticancer efficiency achieved by manipulating PTT killing and autophagy blocking is expected to serve as a more effective anticancer strategy against prostate cancer. STATEMENT OF SIGNIFICANCE: Autophagy is considered as one of the most efficient rescuer and reinforcement mechanisms of cancer cells against photothermal therapy (PTT)-induced cancer cell eradication. How to synergistically manipulate autophagy blocking with significant tumor targeting and penetration to enhance PTT-mediated cancer cell killing remains a substantial challenge. Herein, we fabricated a biomimetic nanoplatform by precisely coating homologous cancer cell membranes onto the surface of mesoporous polydopamine nanoparticles with encapsulation of the autophagy inhibitor chloroquine for synergistic antitumor treatment with high tumor targeting and penetrating efficiency both in vitro and in vivo. This biomimetic nanosystem with synergistically enhanced anticancer efficiency by manipulating PTT killing and autophagy blocking is expected to serve as a more effective anticancer strategy.
Collapse
|
213
|
Niu D, He J, Qin X, Liu Y, Liu H, Hu P, Li Y, Shi J. Superstable and Large-Scalable Organosilica-Micellar Hybrid Nanosystem via a Confined Gelation Strategy for Ultrahigh-Dosage Chemotherapy. NANO LETTERS 2021; 21:9388-9397. [PMID: 34747626 DOI: 10.1021/acs.nanolett.1c02342] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Although various drug nanocarriers have been developed for treating solid tumors, their clinical transformation is greatly limited by the difficulties in quantity production and unpredictable in vivo toxic effects. Herein, a facile "confined-gelation" strategy is developed to quantity-produce intelligent pluronic organosilica micelles (designated as IPOMs) with an undetectable critical micelle concentration (CMC), which features the self-assembly induced core confinement by block copolymers, the inner hydrolysis-condensation of silane to the oligomer skeleton, and oxidative cross-linking of disulfide skeleton to core gelation. The docetaxel-loaded IPOMs (DTX@IPOMs) with precise glutathione (GSH) responsiveness not only display an ultrahigh tolerated dose (360 mg/kg) in healthy Kunming mice model but also exhibit a remarkable tumor inhibition efficacy in both subcutaneous and orthotopic mice tumor models upon an extraordinarily large dosage (50 mg/kg). The present confined-gelation strategy provides a novel pathway to design and quantity-produce low-toxic and high-efficacy organic-inorganic hybrid nanodrugs in future clinical transformations.
Collapse
Affiliation(s)
- Dechao Niu
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontier Science Center of the Materials Biology and Dynamic Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jianping He
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontier Science Center of the Materials Biology and Dynamic Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Xing Qin
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontier Science Center of the Materials Biology and Dynamic Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yu Liu
- State Key Laboratory of Chemical Engineering and School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Honglai Liu
- State Key Laboratory of Chemical Engineering and School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Ping Hu
- State Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Yongsheng Li
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontier Science Center of the Materials Biology and Dynamic Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
- Key Laboratory for Green Processing of Chemical Engineering of Xinjiang Bingtuan, School of Chemistry and Chemical Engineering, Shihezi University, Shihezi 832003, China
| | - Jianlin Shi
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontier Science Center of the Materials Biology and Dynamic Chemistry, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
- State Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| |
Collapse
|
214
|
Chugh V, Vijaya Krishna K, Pandit A. Cell Membrane-Coated Mimics: A Methodological Approach for Fabrication, Characterization for Therapeutic Applications, and Challenges for Clinical Translation. ACS NANO 2021; 15:17080-17123. [PMID: 34699181 PMCID: PMC8613911 DOI: 10.1021/acsnano.1c03800] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 10/13/2021] [Indexed: 05/04/2023]
Abstract
Cell membrane-coated (CMC) mimics are micro/nanosystems that combine an isolated cell membrane and a template of choice to mimic the functions of a cell. The design exploits its physicochemical and biological properties for therapeutic applications. The mimics demonstrate excellent biological compatibility, enhanced biointerfacing capabilities, physical, chemical, and biological tunability, ability to retain cellular properties, immune escape, prolonged circulation time, and protect the encapsulated drug from degradation and active targeting. These properties and the ease of adapting them for personalized clinical medicine have generated a significant research interest over the past decade. This review presents a detailed overview of the recent advances in the development of cell membrane-coated (CMC) mimics. The primary focus is to collate and discuss components, fabrication methodologies, and the significance of physiochemical and biological characterization techniques for validating a CMC mimic. We present a critical analysis of the two main components of CMC mimics: the template and the cell membrane and mapped their use in therapeutic scenarios. In addition, we have emphasized on the challenges associated with CMC mimics in their clinical translation. Overall, this review is an up to date toolbox that researchers can benefit from while designing and characterizing CMC mimics.
Collapse
Affiliation(s)
| | | | - Abhay Pandit
- CÚRAM, SFI Research
Centre for Medical Devices, National University
of Ireland Galway, Galway H91 W2TY, Ireland
| |
Collapse
|
215
|
Hao W, Cui Y, Fan Y, Chen M, Yang G, Wang Y, Yang M, Li Z, Gong W, Yang Y, Gao C. Hybrid membrane-coated nanosuspensions for multi-modal anti-glioma therapy via drug and antigen delivery. J Nanobiotechnology 2021; 19:378. [PMID: 34801032 PMCID: PMC8606100 DOI: 10.1186/s12951-021-01110-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/02/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Glioma is one of the deadliest human cancers. Although many therapeutic strategies for glioma have been explored, these strategies are seldom used in the clinic. The challenges facing the treatment of glioma not only involve the development of chemotherapeutic drugs and immunotherapeutic agents, but also the lack of a powerful platform that could deliver these two moieties to the targeted sites. Herein, we developed chemoimmunotherapy delivery vehicles based on C6 cell membranes and DC membranes to create hybrid membrane-coated DTX nanosuspensions (DNS-[C6&DC]m). RESULTS Results demonstrated successful hybrid membrane fusion and nanosuspension functionalization, and DNS-[C6&DC]m could be used for different modes of anti-glioma therapy. For drug delivery, membrane coating could be applied to target the source cancer cells via a homotypic-targeting mechanism of the C6 cell membrane. For cancer immunotherapy, biomimetic nanosuspension enabled an immune response based on the professional antigen-presenting characteristic of the dendritic cell membrane (DCm), which carry the full array of cancer cell membrane antigens and facilitate the uptake of membrane-bound tumor antigens for efficient presentation and downstream immune n. CONCLUSION DNS-[C6&DC]m is a multifunctional biomimetic nano-drug delivery system with the potential to treat gliomas through tumor-targeted drug delivery combined with immunotherapy, thereby presenting a promising approach that may be utilized for multiple modes of cancer therapy.
Collapse
Affiliation(s)
- Wenyan Hao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Yuexin Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Yueyue Fan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Mengyu Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Guobao Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Yuli Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China.
| | - Meiyan Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Zhiping Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Wei Gong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China
| | - Yang Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China.
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China.
| |
Collapse
|
216
|
Li H, Li S, Lin Y, Chen S, Yang L, Huang X, Wang H, Yu X, Zhang L. Artificial exosomes mediated spatiotemporal-resolved and targeted delivery of epigenetic inhibitors. J Nanobiotechnology 2021; 19:364. [PMID: 34789273 PMCID: PMC8597284 DOI: 10.1186/s12951-021-01107-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/31/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Malignant tumor is usually associated with epigenetic dysregulation, such as overexpression of histone deacetylase (HDAC), thus HDAC has emerged as a therapeutic target for cancer. Histone deacetylase inhibitor has been approved for clinical use to treat hematological cancers. However, the low solubility, short circulation lifetime, and high cytotoxicity partially limited their applications in solid tumor. METHODS The upconversion nanoparticles (UC) modified with mesoporous silica (SUC) was used to load an HDACI, suberoylanilide hydroxamic acid (SAHA), and further camouflaged with M1 macrophage-derived exosome membranes (EMS). EMS was characterized in size and compositions. We also analyzed the epigenetic regulation induced by EMS. Furthermore, we evaluate the biodistribution and in vivo tumor inhibition after the systemic administration of EMS. RESULTS This novel style spatiotemporal-resolved drug delivery system, EMS showed a high loading efficiency of SAHA. EMS could be taken up by lung cancer cells and lead to efficient epigenetic inhibition. We found that the integrin α4β1 on M1-EM, was crucial for the homing of EMS to tumor tissues for the first time. In tumor-bearing mice, EMS showed spatiotemporal-resolved properties and facilitated the drug accumulation in the tumors, which induced superior anti-tumor effects. CONCLUSION This novel style of spatiotemporal-resolved nanoparticles can be used as a theranostic platform for lung cancer therapy.
Collapse
Affiliation(s)
- Huan Li
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Songpei Li
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Yinshan Lin
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Sheng Chen
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Langyu Yang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Xin Huang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Hao Wang
- Department of Oncology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China.
| | - Xiyong Yu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China.
| | - Lingmin Zhang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China.
| |
Collapse
|
217
|
Zhang K, Fu H, Xing C, Luo Y, Cheng F, Fu Q, Huang Y, Qiu L. "Don't eat me/eat me"-combined apoptotic body analogues for efficient targeted therapy of triple-negative breast cancer. J Mater Chem B 2021; 9:8472-8479. [PMID: 34550154 DOI: 10.1039/d1tb01116b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
For the purpose of efficient targeted therapies, suppressing phagocytosis by a mononuclear phagocyte system (MPS), enhancing the "active" targeted delivery, and meeting clinical production criteria are extremely critical for engineering strategies of novel drug delivery systems. Herein, we used a chemically-induced membrane blebbing and extrusion combined method to induce triple-negative breast cancer (TNBC) cell apoptosis to secrete apoptotic body analogue (ABA) vesicles on a large scale for therapeutic drug delivery. After optimization, the ABAs have a desirable size, good biocompatibility, and long-term colloidal stability. Furthermore, ABAs present anti-phagocytosis ("don't eat me") and specific homologous targeting ("eat me") capacities because of their inheritance of membrane proteins such as CD47 and cellular adhesion molecules from parent cells. After loading with toxic protein saporin and anti-twist siRNA, ABAs can significantly inhibit the growth and lung metastasis of TNBC in an orthotopic metastasis model due to their reduced clearance of immune organs, long circulation time, and enhanced targeted accumulation at the tumor sites. These results suggest the great potential of ABAs for targeted drug delivery therapy, in particular efficient TNBC treatment.
Collapse
Affiliation(s)
- Kailong Zhang
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Province Universities Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), School of Life Sciences, Longyan University, Longyan 364012, P. R. China.
| | - Huanru Fu
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Province Universities Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), School of Life Sciences, Longyan University, Longyan 364012, P. R. China.
| | - Chao Xing
- Fujian Key Laboratory of Functional Marine Sensing Materials, Center for Advanced Marine Materials and Smart Sensors, Minjiang University, Fuzhou 350108, P. R. China.
| | - Yi Luo
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Province Universities Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), School of Life Sciences, Longyan University, Longyan 364012, P. R. China.
| | - Fangfang Cheng
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Province Universities Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), School of Life Sciences, Longyan University, Longyan 364012, P. R. China.
| | - Qiang Fu
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Province Universities Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), School of Life Sciences, Longyan University, Longyan 364012, P. R. China.
| | - Yujie Huang
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Province Universities Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), School of Life Sciences, Longyan University, Longyan 364012, P. R. China.
| | - Longxin Qiu
- Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Fujian Province Universities Key Laboratory of Preventive Veterinary Medicine and Biotechnology (Longyan University), School of Life Sciences, Longyan University, Longyan 364012, P. R. China.
| |
Collapse
|
218
|
Tang Y, Jia C, Wang Y, Wan W, Li H, Huang G, Zhang X. Lactate Consumption via Cascaded Enzymes Combined VEGF siRNA for Synergistic Anti-Proliferation and Anti-Angiogenesis Therapy of Tumors. Adv Healthc Mater 2021; 10:e2100799. [PMID: 34310079 DOI: 10.1002/adhm.202100799] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/08/2021] [Indexed: 02/07/2023]
Abstract
Lactate, as the most abundant component with concentrations of 4-40 mm in tumors, contributes to the regulation of metabolic pathways, angiogenesis, and immunosuppression, exhibiting remarkable potential in cancer treatment. Therefore, a codelivery strategy that combined the cascaded enzymes Lactate oxidase/Catalase (LOx/CAT) and vascular endothelial growth factor (VEGF) siRNA (siVEGF) to suppress tumor proliferation and angiogenesis synergistically is creatively proposed. In brief, the cationic liposomes (LIP) encapsulated with LOx/CAT and siVEGF via hydrophilic interaction and electrostatic adsorption followed by coating with PEGylated phenylboronic acid (PP) is established (PPL@[LOX+CAT]). Moreover, a simple 3-aminophenylboronic acid (PBA)-shielded strategy via fructose (Fru) is applied to further enhance the targeting efficiency in the tumor site. The obtained co-encapsulated nanoparticles (NPs) can simultaneous intracellular release of LOx/CAT and siVEGF, and the collaborative use of LOx and CAT can promote lactate consumption even under a hypoxic tumor microenvironment (TME) without producing systemic toxicity. The combined application of lactate depletion and VEGF silencing demonstrated the efficient migration suppression of 4T1 cells in vitro and superior antitumor and antimetastatic properties in vivo. This work offers a promising tumor treatment strategy via integrating cascaded enzymes and gene therapy, and explores a promising therapy regimen for 4T1 triple-negative breast cancer.
Collapse
Affiliation(s)
- Yan Tang
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Changhao Jia
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Yu Wang
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Wenjun Wan
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Hui Li
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Gui Huang
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Xuenong Zhang
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| |
Collapse
|
219
|
Li D, Wang Y, Li C, Wang Q, Sun B, Zhang H, He Z, Sun J. Cancer-specific calcium nanoregulator suppressing the generation and circulation of circulating tumor cell clusters for enhanced anti-metastasis combinational chemotherapy. Acta Pharm Sin B 2021; 11:3262-3271. [PMID: 34729314 PMCID: PMC8546850 DOI: 10.1016/j.apsb.2021.04.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/01/2021] [Accepted: 04/12/2021] [Indexed: 12/18/2022] Open
Abstract
Tumor metastasis is responsible for chemotherapeutic failure and cancer-related death. Moreover, circulating tumor cell (CTC) clusters play a pivotal role in tumor metastasis. Herein, we develop cancer-specific calcium nanoregulators to suppress the generation and circulation of CTC clusters by cancer membrane-coated digoxin (DIG) and doxorubicin (DOX) co-encapsulated PLGA nanoparticles (CPDDs). CPDDs could precisely target the homologous primary tumor cells and CTC clusters in blood and lymphatic circulation. Intriguingly, CPDDs induce the accumulation of intracellular Ca2+ by inhibiting Na+/K+-ATPase, which help restrain cell–cell junctions to disaggregate CTC clusters. Meanwhile, CPDDs suppress the epithelial–mesenchymal transition (EMT) process, resulting in inhibiting tumor cells escape from the primary site. Moreover, the combination of DOX and DIG at a mass ratio of 5:1 synergistically induces the apoptosis of tumor cells. In vitro and in vivo results demonstrate that CPDDs not only effectively inhibit the generation and circulation of CTC clusters, but also precisely target and eliminate primary tumors. Our findings present a novel approach for anti-metastasis combinational chemotherapy.
Collapse
Key Words
- Breast cancer
- CI, combination index
- CLSM, confocal laser scanning microscopy
- CTC, circulating tumor cell
- Cell–cell junctions
- Circulating tumor cell clusters
- DAPI, 4ʹ,6-diamidino-2-phenylindole
- DIG, digoxin
- DLS, dynamic light scattering
- DOX, doxorubicin
- DiR, 1,1-dioctadecyl-3,3,3,3-tetramethylindotricarbocyaineiodide
- Digoxin
- Doxorubicin
- EMT, epithelial–mesenchymal transition
- Epithelial–mesenchymal transition
- H&E, hematoxylin and eosin
- Homologous targeting
- Lung metastasis
- MMP-9, matrix metalloproteinase-9
- MTT, 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazoliumbromide
- TEM, transmission electron microscopy
Collapse
|
220
|
Cheng X, Hao Z, Chu S, Zhang T, Cong C, Liu L, Zhang W, Gu J, Ni S, Wang D, Gao D. Plasmonic enhanced enzyme activity by catalytic cascade induced mutual benefit tumor starvation/immune/photothermal therapy. Biomater Sci 2021; 9:6116-6125. [PMID: 34519735 DOI: 10.1039/d1bm00551k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Single tumor starvation therapy can activate other signaling pathways in tumor cells and easily induce tumor cell metastasis. This research proposes an intelligent nanoparticle, which is effectively combined with plasmonic and immunotherapy to realize a new strategy of "upstream consumption and downstream blocking" of nutrients in tumor sites. The intelligent nanoparticle (Ag-G/C@M) was composed of Ag NCs loaded with glucose oxidase (GOx), catalase (CAT) and coated with the tumor cytomembrane (M). Homologous targeting of tumor cytomembrane facilitated more delivery of Ag-G/C@M to tumor sites and then the plasmonic excited from Ag-G/C@M can increase the catalytic efficiency of the enzymatic reaction. Hydrogen peroxide (H2O2) produced by Ag-G/C@M through the consumption of glucose is further catalyzed by CAT to produce oxygen (O2). This self-reinforcing cascade reaction not only consumes the nutrients of tumor cells, but also the plasmonic-induced photothermal therapy can further stimulate the immune system to produce interferon-γ (IFN-γ), blocking angiogenesis and restricting the nutrient supply of tumor cells. This strategy takes the nutrition necessary for cell survival as the entry point, through endogenous continuous consumption of intracellular nutrients and containment of exogenous supplementation, combined with plasmonic thermal effect and immunotherapy to kill tumor cells, which provides a new way of treating cancer safely and effectively.
Collapse
Affiliation(s)
- Xin Cheng
- State Key Laboratory of Metastable Materials Science and Technology, Key Laboratory of Nanobiotechnology of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao 066004, China.
| | - Zining Hao
- State Key Laboratory of Metastable Materials Science and Technology, Key Laboratory of Nanobiotechnology of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao 066004, China.
| | - Shuzhen Chu
- State Key Laboratory of Metastable Materials Science and Technology, Key Laboratory of Nanobiotechnology of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao 066004, China.
| | - Tiantian Zhang
- State Key Laboratory of Metastable Materials Science and Technology, Key Laboratory of Nanobiotechnology of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao 066004, China.
| | - Cong Cong
- State Key Laboratory of Metastable Materials Science and Technology, Key Laboratory of Nanobiotechnology of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao 066004, China.
| | - Lanxiang Liu
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, P. R. China
| | - Weidong Zhang
- State Key Laboratory of Metastable Materials Science and Technology, Key Laboratory of Nanobiotechnology of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao 066004, China. .,Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, P. R. China
| | - Jianmin Gu
- State Key Laboratory of Metastable Materials Science and Technology, Key Laboratory of Nanobiotechnology of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao 066004, China.
| | - Song Ni
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Desong Wang
- State Key Laboratory of Metastable Materials Science and Technology, Key Laboratory of Nanobiotechnology of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao 066004, China.
| | - Dawei Gao
- State Key Laboratory of Metastable Materials Science and Technology, Key Laboratory of Nanobiotechnology of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao 066004, China.
| |
Collapse
|
221
|
Yan C, Jin Y, Zhao C. Environment Responsive Metal-Organic Frameworks as Drug Delivery System for Tumor Therapy. NANOSCALE RESEARCH LETTERS 2021; 16:140. [PMID: 34480260 PMCID: PMC8417173 DOI: 10.1186/s11671-021-03597-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/27/2021] [Indexed: 05/10/2023]
Abstract
Nanoparticles as drug delivery systems can alter the drugs' hydrophilicity to affect drug uptake and efflux in tissues. They prevent drugs from non-specifically binding with bio-macromolecules and enhance drug accumulation at the lesion sites, improving therapy effects and reducing unnecessary side effects. Metal-organic frameworks (MOFs), the typical nanoparticles, a class of crystalline porous materials via self-assembled organic linkers and metal ions, exhibit excellent biodegradability, pore shape and sizes, and finely tunable chemical composition. MOFs have a rigid molecular structure, and tunable pore size can improve the encapsulation drug's stability under harsh conditions. Besides, the surface of MOFs can be modified with small-molecule ligands and biomolecule, and binding with the biomarkers which is overexpressed on the surface of cancer cells. MOFs formulations for therapeutic have been developed to effectively respond to the unique tumor microenvironment (TEM), such as high H2O2 levels, hypoxia, and high concentration glutathione (GSH). Thus, MOFs as a drug delivery system should avoid drugs leaking during blood circulation and releasing at the lesion sites via a controlling manner. In this article, we will summary environment responsive MOFs as drug delivery systems for tumor therapy under different stimuli.
Collapse
Affiliation(s)
- Chao Yan
- The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, No. 62, Huaihai Road (S.), Huai'an, 223002, China
| | - Yue Jin
- The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, No. 62, Huaihai Road (S.), Huai'an, 223002, China
| | - Chuanxiang Zhao
- School of Medical Technology, Jiangsu College of Nursing, Huai'an City, Jiangsu Province, China.
| |
Collapse
|
222
|
Ran L, Lu B, Qiu H, Zhou G, Jiang J, Hu E, Dai F, Lan G. Erythrocyte membrane-camouflaged nanoworms with on-demand antibiotic release for eradicating biofilms using near-infrared irradiation. Bioact Mater 2021; 6:2956-2968. [PMID: 33732966 PMCID: PMC7930507 DOI: 10.1016/j.bioactmat.2021.01.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/07/2021] [Accepted: 01/28/2021] [Indexed: 12/20/2022] Open
Abstract
The increase in the number of resistant bacteria caused by the abuse of antibiotics and the emergence of biofilms significantly reduce the effectiveness of antibiotics. Bacterial infections are detrimental to our life and health. To reduce the abuse of antibiotics and treat biofilm-related bacterial infections, a biomimetic nano-antibacterial system, RBCM-NW-G namely, that controls the release of antibiotics through near infrared was prepared. The hollow porous structure and the high surface activity of nanoworms are used to realize antibiotic loading, and then, biomimetics are applied with red blood cell membranes (RBCM). RBCM-NW-G, which retains the performance of RBCM, shows enhanced permeability and retention effects. Fluorescence imaging in mice showed the effective accumulation of RBCM-NW-G at the site of infection. In addition, the biomimetic nanoparticles showed a longer blood circulation time and good biocompatibility. Anti-biofilm test results showed damage to biofilms due to a photothermal effect and a highly efficient antibacterial performance under the synergy of the photothermal effect, silver iron, and antibiotics. Finally, by constructing a mouse infection model, the great potential of RBCM-NW-G in the treatment of in vivo infections was confirmed.
Collapse
Affiliation(s)
- Luoxiao Ran
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Bitao Lu
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Haoyu Qiu
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Guofang Zhou
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Jing Jiang
- Chongqing General Hospital, University of Chinese Academy of Sciences No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, China
| | - Enling Hu
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing, 400715, China
| | - Fangyin Dai
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing, 400715, China
| | - Guangqian Lan
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing, 400715, China
| |
Collapse
|
223
|
Yougbaré S, Mutalik C, Okoro G, Lin IH, Krisnawati DI, Jazidie A, Nuh M, Chang CC, Kuo TR. Emerging Trends in Nanomaterials for Antibacterial Applications. Int J Nanomedicine 2021; 16:5831-5867. [PMID: 34475754 PMCID: PMC8405884 DOI: 10.2147/ijn.s328767] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/04/2021] [Indexed: 01/11/2023] Open
Abstract
Around the globe, surges of bacterial diseases are causing serious health threats and related concerns. Recently, the metal ion release and photodynamic and photothermal effects of nanomaterials were demonstrated to have substantial efficiency in eliminating resistance and surges of bacteria. Nanomaterials with characteristics such as surface plasmonic resonance, photocatalysis, structural complexities, and optical features have been utilized to control metal ion release, generate reactive oxygen species, and produce heat for antibacterial applications. The superior characteristics of nanomaterials present an opportunity to explore and enhance their antibacterial activities leading to clinical applications. In this review, we comprehensively list three different antibacterial mechanisms of metal ion release, photodynamic therapy, and photothermal therapy based on nanomaterials. These three different antibacterial mechanisms are divided into their respective subgroups in accordance with recent achievements, showcasing prospective challenges and opportunities in clinical, environmental, and related fields.
Collapse
Affiliation(s)
- Sibidou Yougbaré
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- Institut de Recherche en Sciences de la Santé (IRSS-DRCO)/Nanoro, Ouagadougou, Burkina Faso
| | - Chinmaya Mutalik
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| | - Goodluck Okoro
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| | - I-Hsin Lin
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| | | | - Achmad Jazidie
- Department of Electrical Engineering, Institut Teknologi Sepuluh Nopember, Surabaya, 60111, Indonesia
- Universitas Nahdlatul Ulama Surabaya, Surabaya, 60237, Indonesia
| | - Mohammad Nuh
- Universitas Nahdlatul Ulama Surabaya, Surabaya, 60237, Indonesia
- Department of Biomedical Engineering, Institut Teknologi Sepuluh Nopember, Surabaya, 60111, Indonesia
| | - Che-Chang Chang
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Tsung-Rong Kuo
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| |
Collapse
|
224
|
Xia W, Tao Z, Zhu B, Zhang W, Liu C, Chen S, Song M. Targeted Delivery of Drugs and Genes Using Polymer Nanocarriers for Cancer Therapy. Int J Mol Sci 2021; 22:9118. [PMID: 34502028 PMCID: PMC8431379 DOI: 10.3390/ijms22179118] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/16/2021] [Accepted: 08/21/2021] [Indexed: 12/15/2022] Open
Abstract
Cancer is one of the primary causes of worldwide human deaths. Most cancer patients receive chemotherapy and radiotherapy, but these treatments are usually only partially efficacious and lead to a variety of serious side effects. Therefore, it is necessary to develop new therapeutic strategies. The emergence of nanotechnology has had a profound impact on general clinical treatment. The application of nanotechnology has facilitated the development of nano-drug delivery systems (NDDSs) that are highly tumor selective and allow for the slow release of active anticancer drugs. In recent years, vehicles such as liposomes, dendrimers and polymer nanomaterials have been considered promising carriers for tumor-specific drug delivery, reducing toxicity and improving biocompatibility. Among them, polymer nanoparticles (NPs) are one of the most innovative methods of non-invasive drug delivery. Here, we review the application of polymer NPs in drug delivery, gene therapy, and early diagnostics for cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Siyu Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; (W.X.); (Z.T.); (B.Z.); (W.Z.); (C.L.)
| | - Mingming Song
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; (W.X.); (Z.T.); (B.Z.); (W.Z.); (C.L.)
| |
Collapse
|
225
|
Hu J, Guan Z, Chen J. Multifunctional biomaterials that modulate oxygen levels in the tumor microenvironment. Cancer Lett 2021; 521:39-49. [PMID: 34419500 DOI: 10.1016/j.canlet.2021.08.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 12/27/2022]
Abstract
A characteristic feature of solid tumors is their low oxygen tension, which confers resistance to radiotherapy, photodynamic therapy, and chemotherapy. Therefore, to improve treatment outcomes, it is critical to develop biomaterials capable of targeted modulation of oxygen levels in tumors. In this review, we summarize four types of oxygen-modulating biomaterials, namely, oxygen-carrying biomaterials to deliver oxygen into tumors (e.g., perfluorocarbon and hemoglobin), oxygen-generating biomaterials to promote in situ oxygen generation (e.g., MnO2, catalase, and CuO), oxygen-consuming biomaterials to starve tumors (e.g., photosensitizer, glucose oxidase, and magnesium silicide), and oxygen-circulating biomaterials capable of both providing and consuming oxygen (e.g., ENBS-B). The current literature suggests that these biomaterials are useful for anticancer therapeutics. We present the key molecular mechanisms involved in modulating oxygen levels and the potential applications of these biomaterials in the context of hypoxic tumor treatment.
Collapse
Affiliation(s)
- Jinghui Hu
- School of Rehabilitation, Institute of Rehabilitation Engineering, Binzhou Medical University, Yantai, 264003, PR China
| | - Zhenxin Guan
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, PR China
| | - Jing Chen
- School of Pharmacy, The Key Laboratory of Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine of China, Binzhou Medical University, Yantai, 264003, PR China.
| |
Collapse
|
226
|
Han S, Lee Y, Lee M. Biomimetic cell membrane-coated DNA nanoparticles for gene delivery to glioblastoma. J Control Release 2021; 338:22-32. [PMID: 34391836 DOI: 10.1016/j.jconrel.2021.08.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 01/29/2023]
Abstract
Gene therapy has been introduced as an alternative to radiation and chemical therapy for glioblastoma. Biomimetic nanoparticles coated with cell membranes (CM) have advantages such as high biocompatibility and prolong half-life. To apply CM coated nanoparticles to gene delivery, the polyethylenimine (PEI25k)/plasmid DNA (pDNA) complexes were coated with CM from C6 rat glioblastoma cells. With the CM covering, the PEI25k/pDNA complexes formed stable nanoparticles with negative surface charge. The PEI25k/pDNA/CM nanoparticles had high colloidal stability and could be stored for approximately 20 days without aggregation. The transfection efficiency of the PEI25k/pDNA/CM nanoparticles was higher than that of the PEI25k/pDNA complex in serum-containing medium. This suggests that serum does not interfere with transfection efficiency of the nanoparticles. Moreover, the PEI25k/pDNA/CM nanoparticles had lower toxicity than the PEI25k/DNA complex in vitro and in vivo. The PEI25k/pDNA/CM nanoparticles prepared with CMs of different types of cells were transfected into cells. The results showed that the PEI25k/pDNA/CM nanoparticles with the C6 CM had the highest transfection efficiency to C6 cells, suggesting the homotypic targeting effect. The therapeutic effects of the nanoparticles were evaluated in intracranial C6 transplanted glioblastoma animal models. The PEI25k/pDNA/CM nanoparticles were prepared with herpes simplex virus thymidine kinase plasmid (pHSVtk) and injected into the tumor locally. The results showed that the PEI25k/pHSVtk/CM nanoparticles induced higher HSVtk expression compared with the PEI25k/pHSVtk complex. Furthermore, tumor size was reduced more efficiently by the PEI25k/pHSVtk/CM nanoparticles than by the PEI25k/pHSVtk complex. Overall results indicate that PEI25k/pDNA/CM nanoparticles are suitable for pDNA delivery to glioblastoma.
Collapse
Affiliation(s)
- Sangrok Han
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Youngki Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Minhyung Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea.
| |
Collapse
|
227
|
Zhu YX, Jia HR, Guo Y, Liu X, Zhou N, Liu P, Wu FG. Repurposing Erythrocytes as a "Photoactivatable Bomb": A General Strategy for Site-Specific Drug Release in Blood Vessels. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2100753. [PMID: 34259382 DOI: 10.1002/smll.202100753] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 05/20/2021] [Indexed: 06/13/2023]
Abstract
Tumor vasculature has long been considered as an extremely valuable therapeutic target for cancer therapy, but how to realize controlled and site-specific drug release in tumor blood vessels remains a huge challenge. Despite the widespread use of nanomaterials in constructing drug delivery systems, they are suboptimal in principle for meeting this demand due to their easy blood cell adsorption/internalization and short lifetime in the systemic circulation. Here, natural red blood cells (RBCs) are repurposed as a remote-controllable drug vehicle, which retains RBC's morphology and vessel-specific biodistribution pattern, by installing photoactivatable molecular triggers on the RBC membrane via covalent conjugation with a finely tuned modification density. The molecular triggers can burst the RBC vehicle under short and mild laser irradiation, leading to a complete and site-specific release of its payloads. This cell-based vehicle is generalized by loading different therapeutic agents including macromolecular thrombin, a blood clotting-inducing enzyme, and a small-molecule hypoxia-activatable chemodrug, tirapazamine. In vivo results demonstrate that the repurposed "anticancer RBCs" exhibit long-term stability in systemic circulation but, when tumors receive laser irradiation, precisely releases their cargoes in tumor vessels for thrombosis-induced starvation therapy and local deoxygenation-enhanced chemotherapy. This study proposes a general strategy for blood vessel-specific drug delivery.
Collapse
Affiliation(s)
- Ya-Xuan Zhu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Hao-Ran Jia
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Yuxin Guo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Xiaoyang Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Ningxuan Zhou
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Peidang Liu
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| |
Collapse
|
228
|
Zhang M, Gao S, Yang D, Fang Y, Lin X, Jin X, Liu Y, Liu X, Su K, Shi K. Influencing factors and strategies of enhancing nanoparticles into tumors in vivo. Acta Pharm Sin B 2021; 11:2265-2285. [PMID: 34522587 PMCID: PMC8424218 DOI: 10.1016/j.apsb.2021.03.033] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/05/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023] Open
Abstract
The administration of nanoparticles (NPs) first faces the challenges of evading renal filtration and clearance of reticuloendothelial system (RES). After that, NPs infiltrate through the expanded endothelial space and penetrated the dense stroma of tumor microenvironment to tumor cells. As long as possible to prolong the time of NPs remaining in tumor tissue, NPs release active agent and induce pharmacological action. This review provides a comprehensive summary of the physical and chemical properties of NPs and the influence of various biological factors in tumor microenvironment, and discusses how to improve the final efficacy through adjusting the characteristics and structure of NPs. Perspectives and future directions are also provided.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Kai Shi
- Corresponding author. Tel./fax: +86 24 43520557.
| |
Collapse
|
229
|
Mainini F, De Santis F, Fucà G, Di Nicola M, Rivoltini L, Eccles M. Nanobiotechnology and Immunotherapy: Two Powerful and Cooperative Allies against Cancer. Cancers (Basel) 2021; 13:3765. [PMID: 34359665 PMCID: PMC8345046 DOI: 10.3390/cancers13153765] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/19/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
A number of novel cancer therapies have recently emerged that have rapidly moved from the bench to the clinic. Onco-immunotherapies, such as immune checkpoint blockade inhibitors and adoptive cell therapies, have revolutionized the field, since they provide a way to induce strong anti-tumor immune responses, which are able to fight cancer effectively. However, despite showing great efficacy in hematological and some solid tumors, unresponsiveness, development of therapy resistance and the development of serious adverse effects, limit their capacity to impact the vast majority of tumors. Nanoparticle-based delivery systems are versatile vehicles for a wide variety of molecular cargoes and provide an innovative strategy to improve conventional onco-immunotherapies. They can be finely tuned to release their contents in the tumor microenvironment, or to deliver combinations of adjuvants and antigens in the case of nanovaccines. In this review, we summarize the recent advancements in the field of nanobiotechnology, to remodel the tumor microenvironment and to enhance immunotherapies.
Collapse
Affiliation(s)
- Francesco Mainini
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.M.); (F.D.S.); (G.F.); (M.D.N.)
| | - Francesca De Santis
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.M.); (F.D.S.); (G.F.); (M.D.N.)
| | - Giovanni Fucà
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.M.); (F.D.S.); (G.F.); (M.D.N.)
| | - Massimo Di Nicola
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.M.); (F.D.S.); (G.F.); (M.D.N.)
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Michael Eccles
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
230
|
Cell membrane-camouflaged liposomes for tumor cell-selective glycans engineering and imaging in vivo. Proc Natl Acad Sci U S A 2021; 118:2022769118. [PMID: 34301864 DOI: 10.1073/pnas.2022769118] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The dynamic change of cell-surface glycans is involved in diverse biological and pathological events such as oncogenesis and metastasis. Despite tremendous efforts, it remains a great challenge to selectively distinguish and label glycans of different cancer cells or cancer subtypes. Inspired by biomimetic cell membrane-coating technology, herein, we construct pH-responsive azidosugar liposomes camouflaged with natural cancer-cell membrane for tumor cell-selective glycan engineering. With cancer cell-membrane camouflage, the biomimetic liposomes can prevent protein corona formation and evade phagocytosis of macrophages, facilitating metabolic glycans labeling in vivo. More importantly, due to multiple membrane receptors, the biomimetic liposomes have prominent cell selectivity to homotypic cancer cells, showing higher glycan-labeling efficacy than a single-ligand targeting strategy. Further in vitro and in vivo experiments indicate that cancer cell membrane-camouflaged azidosugar liposomes not only realize cell-selective glycan imaging of different cancer cells and triple-negative breast cancer subtypes but also do well in labeling metastatic tumors. Meanwhile, the strategy is also applicable to the use of tumor tissue-derived cell membranes, which shows the prospect for individual diagnosis and treatment. This work may pave a way for efficient cancer cell-selective engineering and visualization of glycans in vivo.
Collapse
|
231
|
Chen X, Sui X, Lu S, Qu Y, Liu T, Wang T. Preparation of carbon dots-based nanoparticles and their research of bioimaging and targeted antitumor therapy. J Biomed Mater Res B Appl Biomater 2021; 110:220-228. [PMID: 34231969 DOI: 10.1002/jbm.b.34905] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/23/2021] [Accepted: 06/27/2021] [Indexed: 11/08/2022]
Abstract
Carbon dots (CDs) are nanomaterials with excellent photoluminescence property, usually used in the field of bioimaging tumor cells. However, its practical applicability in cancer therapeutics is limited by CDs' insensitive surface properties to complicated tumor microenvironment in vivo. Herein, a new type of innovative biomimetic nanoparticles has been formed with HeLa cell membranes (CM) and multifunctional CDs containing antitumor and bioimaging activities. The CDs are prepared by a facile one-step microwave-assisted procedure. Gallic acid is used as carbon resource and antitumor active molecule. Gelatin is treated as the nitrogen resource. Citric acid monohydrate is used as the auxiliary carbon source and the Hela CM is used for tumor targeting. A series of fluorescence analyses has proved its homotypic targeting and ability of diagnosis. Besides, in vitro and in vivo antitumor experiments further indicate their better antitumor efficiency. The findings show the totally new nanoparticles' feasibilities of dealing with the clinical therapy problems as well as applying for the integration of diagnosis and targeting therapy.
Collapse
Affiliation(s)
- Xuan Chen
- College of Chemistry, Chemical Engineering and Resources Utilization, Northeast Forestry University, Harbin, Heilongjiang, China
| | - Xiaoyu Sui
- College of Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Shuting Lu
- College of Chemistry, Chemical Engineering and Resources Utilization, Northeast Forestry University, Harbin, Heilongjiang, China
| | - Yanmei Qu
- College of Chemistry, Chemical Engineering and Resources Utilization, Northeast Forestry University, Harbin, Heilongjiang, China
| | - Tingting Liu
- College of Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Ting Wang
- College of Chemistry, Chemical Engineering and Resources Utilization, Northeast Forestry University, Harbin, Heilongjiang, China
| |
Collapse
|
232
|
Zhang J, Wang N, Li Q, Zhou Y, Luan Y. A two-pronged photodynamic nanodrug to prevent metastasis of basal-like breast cancer. Chem Commun (Camb) 2021; 57:2305-2308. [PMID: 33533351 DOI: 10.1039/d0cc08162k] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A two-pronged concept combining photodynamic therapy (PDT) and epithelial-mesenchymal transition (EMT) blockade in a minimalist nanoplatform was proposed to combat basal-like breast cancer (BLBC) metastasis. Based on PDT-mediated tumor killing and epalrestat (Epa)-mediated EMT blockade, as-prepared Ce6/Epa nanoparticles prevented BLBC metastasis effectively in vivo, providing a very promising two-pronged strategy against BLBC metastasis.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Ningning Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Qian Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Yaxin Zhou
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Yuxia Luan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
233
|
Li M, Xu Z, Zhang L, Cui M, Zhu M, Guo Y, Sun R, Han J, Song E, He Y, Su Y. Targeted Noninvasive Treatment of Choroidal Neovascularization by Hybrid Cell-Membrane-Cloaked Biomimetic Nanoparticles. ACS NANO 2021; 15:9808-9819. [PMID: 34037377 DOI: 10.1021/acsnano.1c00680] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Choroidal neovascularization (CNV) is the leading cause of vision loss in many blinding diseases, but current antiangiogenic therapies with invasively intravitreal injection suffer from poor patient compliance and a rate of devastating ocular complications. Here, we develop an alternative antiangiogenic agent based on hybrid cell-membrane-cloaked nanoparticles for noninvasively targeted treatment of CNV. The retinal endotheliocyte membrane coating provides as-fabricated nanoagents with homotypic targeting capability and binding ability to the vascular endothelial growth factor. The fusion of red blood cell membranes protects the hybrid membrane-coated nanoparticles from phagocytosis by macrophages. In a laser-induced wet age-related macular degeneration mouse model, a significantly enhanced accumulation is observed in CNV regions after intravenous delivery of the hybrid membrane-coated nanoparticles. Moreover, an excellent therapeutic efficacy is achieved in reducing the leakage and area of CNV. Overall, the biomimetic antiangiogenic nanoagents provide an effective approach for noninvasive treatment of CNV.
Collapse
Affiliation(s)
- Manjing Li
- Laboratory of Nanoscale Biochemical Analysis, Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhaojian Xu
- Laboratory of Nanoscale Biochemical Analysis, Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Lu Zhang
- Laboratory of Nanoscale Biochemical Analysis, Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Mingyue Cui
- Laboratory of Nanoscale Biochemical Analysis, Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Manhui Zhu
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou 215123, China
| | - Yang Guo
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou 215123, China
| | - Rong Sun
- Laboratory of Nanoscale Biochemical Analysis, Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Junfei Han
- Laboratory of Nanoscale Biochemical Analysis, Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - E Song
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou 215123, China
| | - Yao He
- Laboratory of Nanoscale Biochemical Analysis, Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yuanyuan Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangdong 510120, China
| |
Collapse
|
234
|
Shen J, Karges J, Xiong K, Chen Y, Ji L, Chao H. Cancer cell membrane camouflaged iridium complexes functionalized black-titanium nanoparticles for hierarchical-targeted synergistic NIR-II photothermal and sonodynamic therapy. Biomaterials 2021; 275:120979. [PMID: 34166910 DOI: 10.1016/j.biomaterials.2021.120979] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 12/15/2022]
Abstract
The diagnosis and treatment of cancer is one of the biggest medical challenges of the century. Despite significant improvements, there remains an urgent need for novel anticancer procedures. Among the most promising approaches, increasing attention has been devoted towards photothermal and sonodynamic therapy in which sensitizers are activated upon light/ultrasound radiation to generate a cytotoxic effect. While these methods have undoubtedly shown a high therapeutic success, these techniques are intrinsically limited. Herein, the functionalization of black-titanium nanoparticle with iridium complexes and cancer cell membranes in a nanoplatform for hierarchical targeted synergistic photothermal and sonodynamic cancer imaging and therapy is proposed. The particles showed to generate efficiently heat upon irradiation and catalytically form reactive oxygen species upon ultrasound radiation. The nanoparticle formulation demonstrated to selectively localize in the mitochondria as well as to preferentially accumulate in cancerous over non-cancerous cells as well as in the tumor inside a mouse model, presenting a hierarchical targeting strategy. Upon synergistic irradiation at 1064 nm and ultrasound radiation, the nanoparticles were able to act as an imaging agent and identify the tumor site with high spatial resolution as well as act as a therapeutic agent and completely eradicate a tumor inside a mouse model.
Collapse
Affiliation(s)
- Jinchao Shen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Johannes Karges
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, United States
| | - Kai Xiong
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Yu Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510006, PR China; MOE Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, 400201, PR China.
| |
Collapse
|
235
|
Russo E, Spallarossa A, Tasso B, Villa C, Brullo C. Nanotechnology of Tyrosine Kinase Inhibitors in Cancer Therapy: A Perspective. Int J Mol Sci 2021; 22:6538. [PMID: 34207175 PMCID: PMC8235113 DOI: 10.3390/ijms22126538] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/23/2022] Open
Abstract
Nanotechnology is an important application in modern cancer therapy. In comparison with conventional drug formulations, nanoparticles ensure better penetration into the tumor mass by exploiting the enhanced permeability and retention effect, longer blood circulation times by a reduced renal excretion and a decrease in side effects and drug accumulation in healthy tissues. The most significant classes of nanoparticles (i.e., liposomes, inorganic and organic nanoparticles) are here discussed with a particular focus on their use as delivery systems for small molecule tyrosine kinase inhibitors (TKIs). A number of these new compounds (e.g., Imatinib, Dasatinib, Ponatinib) have been approved as first-line therapy in different cancer types but their clinical use is limited by poor solubility and oral bioavailability. Consequently, new nanoparticle systems are necessary to ameliorate formulations and reduce toxicity. In this review, some of the most important TKIs are reported, focusing on ongoing clinical studies, and the recent drug delivery systems for these molecules are investigated.
Collapse
Affiliation(s)
- Eleonora Russo
- Section of Medicinal and Cosmetic Chemistry, Department of Pharmacy, University of Genova, Viale Benedetto XV, 3-16132 Genova, Italy; (A.S.); (B.T.); (C.V.)
| | | | | | | | - Chiara Brullo
- Section of Medicinal and Cosmetic Chemistry, Department of Pharmacy, University of Genova, Viale Benedetto XV, 3-16132 Genova, Italy; (A.S.); (B.T.); (C.V.)
| |
Collapse
|
236
|
Rawal S, Patel M. Bio-Nanocarriers for Lung Cancer Management: Befriending the Barriers. NANO-MICRO LETTERS 2021; 13:142. [PMID: 34138386 PMCID: PMC8196938 DOI: 10.1007/s40820-021-00630-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/23/2021] [Indexed: 05/03/2023]
Abstract
Lung cancer is a complex thoracic malignancy developing consequential to aberrations in a myriad of molecular and biomolecular signaling pathways. It is one of the most lethal forms of cancers accounting to almost 1.8 million new annual incidences, bearing overall mortality to incidence ratio of 0.87. The dismal prognostic scenario at advanced stages of the disease and metastatic/resistant tumor cell populations stresses the requisite of advanced translational interdisciplinary interventions such as bionanotechnology. This review article deliberates insights and apprehensions on the recent prologue of nanobioengineering and bionanotechnology as an approach for the clinical management of lung cancer. The role of nanobioengineered (bio-nano) tools like bio-nanocarriers and nanobiodevices in secondary prophylaxis, diagnosis, therapeutics, and theranostics for lung cancer management has been discussed. Bioengineered, bioinspired, and biomimetic bio-nanotools of considerate translational value have been reviewed. Perspectives on existent oncostrategies, their critical comparison with bio-nanocarriers, and issues hampering their clinical bench side to bed transformation have also been summarized.
Collapse
Affiliation(s)
- Shruti Rawal
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad, Gujarat, 382 481, India
| | - Mayur Patel
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad, Gujarat, 382 481, India.
| |
Collapse
|
237
|
Liu Z, Zhang L, Cui T, Ma M, Ren J, Qu X. A Nature-Inspired Metal-Organic Framework Discriminator for Differential Diagnosis of Cancer Cell Subtypes. Angew Chem Int Ed Engl 2021; 60:15436-15444. [PMID: 33960090 DOI: 10.1002/anie.202102286] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Indexed: 12/13/2022]
Abstract
Metabolic glycan labeling (MGL) followed by bioorthogonal chemistry provides a powerful tool for tumor imaging and therapy. However, selectively metabolic labeling of cells or tissues of interest remains a challenge. Particularly, owing to tumor heterogeneity including tumor subtypes and interpatient heterogeneity, it is far more difficult to realize tumor-cell-selective metabolic labeling for precise diagnosis. Inspired by nature, we designed azidosugar-functionalized metal-organic frameworks camouflaged with cancer cell membranes to accomplish cancer-cell-selective MGL in vivo. With abundant receptors, this biomimetic platform not only selectively targets homotypic cells but also realizes different breast cancer subtype-selective MGL. Moreover, the endo/lysosomal-escaped ZIF-8 can make azidosugar escape from lysosomes and accelerate its metabolic incorporation. This strategy also takes advantage of cancer-tissue-derived cell membranes, which may have huge potential for personalized diagnosis and therapy.
Collapse
Affiliation(s)
- Zhengwei Liu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100039, P. R. China
| | - Lu Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100039, P. R. China
| | - Tingting Cui
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.,University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Mengmeng Ma
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.,University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.,University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China.,University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| |
Collapse
|
238
|
Liu Z, Zhang L, Cui T, Ma M, Ren J, Qu X. A Nature‐Inspired Metal–Organic Framework Discriminator for Differential Diagnosis of Cancer Cell Subtypes. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202102286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Zhengwei Liu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Chinese Academy of Sciences Beijing 100039 P. R. China
| | - Lu Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Chinese Academy of Sciences Beijing 100039 P. R. China
| | - Tingting Cui
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Mengmeng Ma
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| |
Collapse
|
239
|
Liu L, Chen Y, Liu C, Yan Y, Yang Z, Chen X, Liu G. Effect of Extracellular Matrix Coating on Cancer Cell Membrane-Encapsulated Polyethyleneimine/DNA Complexes for Efficient and Targeted DNA Delivery In Vitro. Mol Pharm 2021; 18:2803-2822. [PMID: 34086466 DOI: 10.1021/acs.molpharmaceut.1c00359] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Polyethyleneimine (PEI) has a good spongy proton effect and is an excellent nonviral gene vector, but its high charge density leads to the instability and toxicity of PEI/DNA complexes. Cell membrane (CM) capsules provide a universal and natural solution for this problem. Here, CM-coated PEI/DNA capsules (CPDcs) were prepared through extrusion, and the extracellular matrix was coated on CPDcs (ECM-CPDcs) for improved targeting. The results showed that compared with PEI/DNA complexes, CPDcs had core-shell structures (PEI/DNA complexes were coated by a 6-10 nm layer), lower cytotoxicity, and obvious homologous targeting. The internalization and transfection efficiency of 293T-CM-coated PEI70k/DNA capsules (293T-CP70Dcs) were 91.8 and 74.5%, respectively, which were higher than those of PEI70k/DNA complexes. Then, the internalization and transfection efficiency of 293T-CP70Dcs were further improved by ECM coating, which were 94.7 and 78.9%, respectively. Then, the internalization and transfection efficiency of 293T-CP70Dcs were further improved by ECM coating, which were 94.7 and 78.9%, respectively. Moreover, the homologous targeting of various CPDcs was improved by ECM coating, and other CPDcs also showed similar effects as 293T-CP70Dcs after ECM coating. These findings suggest that tumor-targeted CPDcs may have considerable advantages in gene delivery.
Collapse
Affiliation(s)
- Liang Liu
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yiran Chen
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Chaobing Liu
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yujian Yan
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Zhaojun Yang
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Xin Chen
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Gang Liu
- School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| |
Collapse
|
240
|
AlHumaidi RB, Fayed B, Sharif SI, Noreddin A, Soliman SSM. Role of Exosomes in Breast Cancer Management: Evidence-Based Review. Curr Cancer Drug Targets 2021; 21:666-675. [PMID: 34077346 DOI: 10.2174/1568009621666210601115707] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 12/09/2022]
Abstract
The field of cancer research has massively grown in recent decades, leading to a better understanding of the underlying causes and greatly improved the therapeutic approaches. Breast cancer (BC) is the third leading cause of mortality among all cancers and the most common malignant disease in women worldwide, representing one in four cancers in women. The crosstalk between cancer cells and the surrounding microenvironment is crucial for tumor progression and metastatic process. Tumor cells communicate not only through classical paracrine signaling mechanisms, including cytokines, chemokines, growth factors, but also through"exosomes". Exosomes are nano-vesicles that are released by various types of cells. Over the last decade, researchers have been attracted to the role of exosomes in breast cancer. It has been proven that exosomes influence major tumor-related pathways, including invasion, migration, epithelial-to-mesenchymal transition (EMT), metastasis, and drug resistance. Additionally, exosomes play important roles in clinical applications. Several studies have demonstrated the potential applications of exosomes in cancer therapy and diagnosis. Furthermore, exosomes have been engineered to function as nano-delivery systems of chemotherapeutic drugs. They can also be designed as vaccines to trigger the patient's immune system. This review discusses the recent progress regarding the use of exosomes as drug delivery systems, therapeutic agents, biomarkers, and vaccines against breast cancer.
Collapse
Affiliation(s)
- Razan B AlHumaidi
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Bahgat Fayed
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Suleiman I Sharif
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Ayman Noreddin
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Sameh S M Soliman
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| |
Collapse
|
241
|
Biomimetic Nanotechnology: A Natural Path Forward for Tumor-Selective and Tumor-Specific NIR Activable Photonanomedicines. Pharmaceutics 2021; 13:pharmaceutics13060786. [PMID: 34070233 PMCID: PMC8225032 DOI: 10.3390/pharmaceutics13060786] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/08/2021] [Accepted: 05/14/2021] [Indexed: 12/18/2022] Open
Abstract
The emergence of biomimetic nanotechnology has seen an exponential rise over the past decade with applications in regenerative medicine, immunotherapy and drug delivery. In the context of nanomedicines activated by near infrared (NIR) photodynamic processes (photonanomedicines; PNMs), biomimetic nanotechnology is pushing the boundaries of activatable tumor targeted nanoscale drug delivery systems. This review discusses how, by harnessing a unique collective of biological processes critical to targeting of solid tumors, biomimetic PNMs (bPNMs) can impart tumor cell specific and tumor selective photodynamic therapy-based combination regimens. Through molecular immune evasion and self-recognition, bPNMs can confer both tumor selectivity (preferential bulk tumor accumulation) and tumor specificity (discrete molecular affinity for cancer cells), respectively. They do so in a manner that is akin, yet arguably superior, to synthetic molecular-targeted PNMs. A particular emphasis is made on how bPNMs can be engineered to circumvent tumor cell heterogeneity, which is considered the Achilles’ heel of molecular targeted therapeutics. Forward-looking propositions are also presented on how patient tumor heterogeneity can ultimately be recapitulated to fabricate patient-specific, heterogeneity-targeting bPNMs.
Collapse
|
242
|
Wang R, Zhang Z, Liu B, Xue J, Liu F, Tang T, Liu W, Feng F, Qu W. Strategies for the design of nanoparticles: starting with long-circulating nanoparticles, from lab to clinic. Biomater Sci 2021; 9:3621-3637. [PMID: 34008587 DOI: 10.1039/d0bm02221g] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Short half-life is one of the main causes of drug attrition in clinical development, which also leads to the failure of many leading compounds and hits to become drug candidates. Nowadays, nanomaterials have been applied to drug development to address this problem. In fact, the clinical application of nanoparticles (NPs) is severely limited due to their rapid elimination by the reticuloendothelial system (RES) in vivo. In this paper, we aim to summarize representative strategies on prolonging the circulation time for bridging the gap between excellent pharmaceutics and proper half-life and encourage clinical translation.
Collapse
Affiliation(s)
- Ruyi Wang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Zhongtao Zhang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Bowen Liu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Jingwei Xue
- The Joint Laboratory of China Pharmaceutical University and Taian City Central Hospital, Taian City Central Hospital, Taian, 271000, China and Taian City institute of Digestive Disease, Taian City Central Hospital, Taian, 271000, China
| | - Fulei Liu
- The Joint Laboratory of China Pharmaceutical University and Taian City Central Hospital, Taian City Central Hospital, Taian, 271000, China and Pharmaceutical Department, Taian City Central Hospital, Taian, 271000, China
| | - Tongzhong Tang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China and Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China. and Jiangsu Food and Pharmaceutical Science College, Huaian, 223003, China.
| | - Wei Qu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| |
Collapse
|
243
|
Asrorov AM, Gu Z, Li F, Liu L, Huang Y. Biomimetic camouflage delivery strategies for cancer therapy. NANOSCALE 2021; 13:8693-8706. [PMID: 33949576 DOI: 10.1039/d1nr01127h] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cancer remains a significant challenge despite the progress in developing different therapeutic approaches. Nanomedicine has been explored as a promising novel cancer therapy. Recently, biomimetic camouflage strategies have been investigated to change the bio-fate of therapeutics and target cancer cells while reducing the unwanted exposure on normal tissues. Endogenous components (e.g., proteins, polysaccharides, and cell membranes) have been used to develop anticancer drug delivery systems. These biomimetic systems can overcome biological barriers and enhance tumor cell-specific uptake. The tumor-targeting mechanisms include ligand-receptor interactions and stimuli-responsive (e.g., pH-sensitive and light-sensitive) delivery. Drug delivery carriers composed of endogenous components represent a promising approach for improving cancer treatment efficacy. In this paper, different biomimetic drug delivery strategies for cancer treatment are reviewed with a focus on the discussion of their advantages and potential applications.
Collapse
Affiliation(s)
- Akmal M Asrorov
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China. and Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, 83, M. Ulughbek Street, Tashkent 100125, Uzbekistan
| | - Zeyun Gu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China.
| | - Feng Li
- Harrison School of Pharmacy, Auburn University, Auburn, AL 36849, USA.
| | - Lingyun Liu
- First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou 510450, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China. and Zhongshan Institute for Drug Discovery, Institutes of Drug Discovery and Development, Chinese Academy of Sciences, Zhongshan 528437, China and NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China
| |
Collapse
|
244
|
Cell membrane cloaked nanomedicines for bio-imaging and immunotherapy of cancer: Improved pharmacokinetics, cell internalization and anticancer efficacy. J Control Release 2021; 335:130-157. [PMID: 34015400 DOI: 10.1016/j.jconrel.2021.05.018] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 01/13/2023]
Abstract
Despite enormous advancements in the field of oncology, the innocuous and effectual treatment of various types of malignancies remained a colossal challenge. The conventional modalities such as chemotherapy, radiotherapy, and surgery have been remained the most viable options for cancer treatment, but lacking of target-specificity, optimum safety and efficacy, and pharmacokinetic disparities are their impliable shortcomings. Though, in recent decades, numerous encroachments in the field of onco-targeted drug delivery have been adapted but several limitations (i.e., short plasma half-life, early clearance by reticuloendothelial system, immunogenicity, inadequate internalization and localization into the onco-tissues, chemoresistance, and deficient therapeutic efficacy) associated with these onco-targeted delivery systems limits their clinical viability. To abolish the aforementioned inadequacies, a promising approach has been emerged in which stealthing of synthetic nanocarriers has been attained by cloaking them into the natural cell membranes. These biomimetic nanomedicines not only retain characteristics features of the synthetic nanocarriers but also inherit the cell-membrane intrinsic functionalities. In this review, we have summarized preparation methods, mechanism of cloaking, and pharmaceutical and therapeutic superiority of cell-membrane camouflaged nanomedicines in improving the bio-imaging and immunotherapy against various types of malignancies. These pliable adaptations have revolutionized the current drug delivery strategies by optimizing the plasma circulation time, improving the permeation into the cancerous microenvironment, escaping the immune evasion and rapid clearance from the systemic circulation, minimizing the immunogenicity, and enabling the cell-cell communication via cell membrane markers of biomimetic nanomedicines. Moreover, the preeminence of cell-membrane cloaked nanomedicines in improving the bio-imaging and theranostic applications, alone or in combination with phototherapy or radiotherapy, have also been pondered.
Collapse
|
245
|
Pei X, Pan X, Xu X, Xu X, Huang H, Wu Z, Qi X. 4T1 cell membrane fragment reunited PAMAM polymer units disguised as tumor cell clusters for tumor homotypic targeting and anti-metastasis treatment. Biomater Sci 2021; 9:1325-1333. [PMID: 33355563 DOI: 10.1039/d0bm01731k] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cell membrane-based nanoparticles have garnered increasing attention owing to their inherent biomimetic properties, such as homotypic targeting, prolong circulation, and immune escaping mechanisms. However, most of these biomimetic nanoparticles appear as an orientated core-shell unit because of the lack of the full utilization and direction control of membranes. Different from those single-unit delivery systems, we reported a multiple-unit nanocluster by randomly reuniting multiple PAMAM polymeric core units into a single nanocluster via simple electrostatic interactions between 4T1 cell membrane fragments and PAMAM. Similar to tumor cell clusters, the doxorubicin (DOX)-loaded nanoclusters (CCNCs) could actively metastasis towards cancer cells after getting access to the systemic circulation due to their specific homotypic targeting ability. In this study, CCNCs showed significantly higher tumor inhibition efficacy in 4T1 tumor-bearing mice compared with that of free DOX and PAMAM@DOX-treated groups. Furthermore, the quantitative analysis showed that the number of pulmonary metastatic nodules remarkably reduced, indicating the potential anti-metastasis effect of CCNCs. Overall, these tumor cell membrane fragment reunited PAMAM polymer units could disguise as tumor cell clusters for encouraging tumor homotypic targeting and anti-metastasis treatment.
Collapse
Affiliation(s)
- Xiaochen Pei
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China.
| | - Xiuhua Pan
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China.
| | - Xiaoyi Xu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China.
| | - Xiang Xu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China.
| | - Haiqin Huang
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Zhenghong Wu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China.
| | - Xiaole Qi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China.
| |
Collapse
|
246
|
Chen X, Liu B, Tong R, Zhan L, Yin X, Luo X, Huang Y, Zhang J, He W, Wang Y. Orchestration of biomimetic membrane coating and nanotherapeutics in personalized anticancer therapy. Biomater Sci 2021; 9:590-625. [PMID: 33305765 DOI: 10.1039/d0bm01617a] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Nanoparticle-based therapeutic and detectable modalities can augment anticancer efficiency, holding potential in capable target and suppressive metastases post administration. However, the individual discrepancies of the current "one-size-fits-all" strategies for anticancer nanotherapeutics have heralded the need for "personalized therapy". Benefiting from the special inherency of various cells, diverse cell membrane-coated nanoparticles (CMCNs) were established on a patient-by-patient basis, which would facilitate the personalized treatment of individual cancer patients. CMCNs in a complex microenvironment can evade the immune system and target homologous tumors with a suppressed immune response, as well as a prolonged circulation time, consequently increasing the drug accumulation at the tumor site and anticancer therapeutic efficacy. This review focuses on the emerging strategies and advances of CMCNs to synergistically integrate the merit of source cells with nanoparticulate delivery systems for the orchestration of personalized anticancer nanotherapeutics, thus discussing their rationalities in facilitating chemotherapy, imaging, immunotherapy, phototherapy, radiotherapy, sonodynamic, magnetocaloric, chemodynamic and gene therapy. Furthermore, the mechanism, challenges and opportunities of CMCNs in personalized anticancer therapy were highlighted to further boost cooperation from different fields, including materials science, chemistry, medicine, pharmacy and biology for the lab-to-clinic translation of CMCNs combined with the individual advantages of source cells and nanotherapeutics.
Collapse
Affiliation(s)
- Xuerui Chen
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Bingbing Liu
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| | - Rongliang Tong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Lin Zhan
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Xuelian Yin
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Xin Luo
- Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yanan Huang
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Junfeng Zhang
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Wen He
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yanli Wang
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| |
Collapse
|
247
|
Chen H, Zheng D, Pan W, Li X, Lv B, Gu W, Machuki JO, Chen J, Liang W, Qin K, Greven J, Hildebrand F, Yu Z, Zhang X, Guo K. Biomimetic Nanotheranostics Camouflaged with Cancer Cell Membranes Integrating Persistent Oxygen Supply and Homotypic Targeting for Hypoxic Tumor Elimination. ACS APPLIED MATERIALS & INTERFACES 2021; 13:19710-19725. [PMID: 33890760 DOI: 10.1021/acsami.1c03010] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Treatment resistance of the tumors to photodynamic therapy (PDT) owing to O2 deficiency largely compromised the therapeutic efficacy, which could be addressed via modulating oxygen levels by using O2 self-enriched nanosystems. Here, we report on augmenting the O2-evolving strategy based on a biomimetic, catalytic nanovehicle (named as N/P@MCC), constructed by the catalase-immobilized hollow mesoporous nanospheres by enveloping a cancer cell membrane (CCM), which acts as an efficient nanocontainer to accommodate nitrogen-doped graphene quantum dots (N-GQDs) and protoporphyrin IX (PpIX). Inheriting the virtues of biomimetic CCM cloaking, the CCM-derived shell conferred N/P@MCC nanovehicles with highly specific self-recognition and homotypic targeting toward cancerous cells, ensuring tumor-specific accumulation and superior circulation durations. N-GQDs, for the first time, have been evidenced as a new dual-functional nanoagents with PTT and PDT capacities, enabling the generation of 1O2 for PDT and inducing local low-temperature hyperthermia for thermally ablating cancer cells and infrared thermal imaging (IRT). Leveraging the intrinsic catalytic features of catalase, such N/P@MCC nanovehicles effectively scavenged the excessive H2O2 to sustainably evolve oxygen for a synchronous O2 self-supply and hypoxia alleviation, with an additional benefit because the resulting O2 bubbles could function as an echo amplifier, leading to the sufficient echogenic reflectivity for ultrasound imaging. Concurrently, the elevated O2 reacted with N-GQDs and PpIX to elicit a maximally increased 1O2 output for augmented PDT. Significantly, the ultrasound imaging coupled with fluorescence imaging, IRT, performs a tumor-modulated trimodal bioimaging effect. Overall, this offers a paradigm to rationally explore O2 self-supply strategies focused on versatile nanotheranostics for hypoxic tumor elimination.
Collapse
Affiliation(s)
- Hongliang Chen
- Institute of Orthopedics, Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P. R. China
| | - Donghui Zheng
- Department of Nephrology, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, P. R. China
| | - Wenzhen Pan
- Institute of Orthopedics, Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P. R. China
| | - Xiang Li
- Department of Nephrology, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, P. R. China
- Department of Clinical Laboratory, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, P. R. China
| | - Bin Lv
- Department of Orthopedics, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu Province 212002, P. R. China
| | - Wenxiang Gu
- Institute of Orthopedics, Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P. R. China
| | - Jeremiah Ong'achwa Machuki
- Institute of Orthopedics, Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P. R. China
| | - Jiahui Chen
- Key Laboratory of Animal Products Processing, Ministry of Agriculture, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Weiqiang Liang
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Kang Qin
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Johannes Greven
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Frank Hildebrand
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Zhiqiang Yu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, P. R. China
| | - Xing Zhang
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Kaijin Guo
- Institute of Orthopedics, Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P. R. China
| |
Collapse
|
248
|
Ruan S, Zhou Y, Jiang X, Gao H. Rethinking CRITID Procedure of Brain Targeting Drug Delivery: Circulation, Blood Brain Barrier Recognition, Intracellular Transport, Diseased Cell Targeting, Internalization, and Drug Release. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2004025. [PMID: 33977060 PMCID: PMC8097396 DOI: 10.1002/advs.202004025] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/03/2020] [Indexed: 05/06/2023]
Abstract
The past decades have witnessed great progress in nanoparticle (NP)-based brain-targeting drug delivery systems, while their therapeutic potentials are yet to be fully exploited given that the majority of them are lost during the delivery process. Rational design of brain-targeting drug delivery systems requires a deep understanding of the entire delivery process along with the issues that they may encounter. Herein, this review first analyzes the typical delivery process of a systemically administrated NPs-based brain-targeting drug delivery system and proposes a six-step CRITID delivery cascade: circulation in systemic blood, recognizing receptor on blood-brain barrier (BBB), intracellular transport, diseased cell targeting after entering into parenchyma, internalization by diseased cells, and finally intracellular drug release. By dissecting the entire delivery process into six steps, this review seeks to provide a deep understanding of the issues that may restrict the delivery efficiency of brain-targeting drug delivery systems as well as the specific requirements that may guarantee minimal loss at each step. Currently developed strategies used for troubleshooting these issues are reviewed and some state-of-the-art design features meeting these requirements are highlighted. The CRITID delivery cascade can serve as a guideline for designing more efficient and specific brain-targeting drug delivery systems.
Collapse
Affiliation(s)
- Shaobo Ruan
- Key laboratory of Drug Targeting and Drug Delivery Systems of the Education MinistrySichuan Engineering Laboratory for Plant‐sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of PharmacySichuan UniversityChengdu610041China
- Department of PharmaceuticsCollege of PharmacyUniversity of FloridaGainesvilleFlorida32610USA
| | - Yang Zhou
- Key laboratory of Drug Targeting and Drug Delivery Systems of the Education MinistrySichuan Engineering Laboratory for Plant‐sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of PharmacySichuan UniversityChengdu610041China
| | - Xinguo Jiang
- Key laboratory of Smart Drug DeliveryMinistry of EducationSchool of PharmacyFudan UniversityShanghai201203China
| | - Huile Gao
- Key laboratory of Drug Targeting and Drug Delivery Systems of the Education MinistrySichuan Engineering Laboratory for Plant‐sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of PharmacySichuan UniversityChengdu610041China
| |
Collapse
|
249
|
Gong C, Zhang X, Shi M, Li F, Wang S, Wang Y, Wang Y, Wei W, Ma G. Tumor Exosomes Reprogrammed by Low pH Are Efficient Targeting Vehicles for Smart Drug Delivery and Personalized Therapy against their Homologous Tumor. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002787. [PMID: 34026432 PMCID: PMC8132050 DOI: 10.1002/advs.202002787] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 02/08/2021] [Indexed: 05/17/2023]
Abstract
As membrane-bound extracellular vesicles, exosomes have targeting ability for specific cell types, and the cellular environment strongly impacts their content and uptake efficiency. Inspired by these natural properties, the impacts of various cellular stress conditions on the uptake efficiency of tumor iterated exosomes are evaluated, and low-pH treatment caused increased uptake efficiency and retained cell-type specificity is found. Lipidomics analyses and molecular dynamics simulations reveal a glycerolipid self-aggregation-based mechanism for the enhanced homologous uptake. Furthermore, these low-pH reprogrammed exosomes are developed into a smart drug delivery platform, which is capable of specifically targeting tumor cells and selectively releasing diverse chemodrugs in response to the exosome rupture by the near-infrared irradiance-triggered burst of reactive oxygen species. This platform exerts safe and enhanced antitumor effects demonstrated by multiple model mice experiments. These results open a new avenue to reprogram exosomes for smart drug delivery and potentially personalized therapy against their homologous tumor.
Collapse
Affiliation(s)
- Changguo Gong
- Department of GastroenterologyTongren HospitalShanghai Jiao Tong University School of MedicineNo. 1111 Xianxia Road, Changning DistrictShanghai200336P. R. China
| | - Xiao Zhang
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of SciencesNo. 1 Bei‐Er‐Tiao, Zhong‐Guan‐Cun, Haidian DistrictBeijing100190P. R. China
| | - Min Shi
- Department of GastroenterologyTongren HospitalShanghai Jiao Tong University School of MedicineNo. 1111 Xianxia Road, Changning DistrictShanghai200336P. R. China
| | - Feng Li
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of SciencesNo. 1 Bei‐Er‐Tiao, Zhong‐Guan‐Cun, Haidian DistrictBeijing100190P. R. China
- School of Chemical EngineeringUniversity of Chinese Academy of SciencesNo. 19A Yuquan RoadBeijing100049P. R. China
| | - Shuang Wang
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of SciencesNo. 1 Bei‐Er‐Tiao, Zhong‐Guan‐Cun, Haidian DistrictBeijing100190P. R. China
| | - Yan Wang
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of SciencesNo. 1 Bei‐Er‐Tiao, Zhong‐Guan‐Cun, Haidian DistrictBeijing100190P. R. China
- School of Chemical EngineeringUniversity of Chinese Academy of SciencesNo. 19A Yuquan RoadBeijing100049P. R. China
| | - Yugang Wang
- Department of GastroenterologyTongren HospitalShanghai Jiao Tong University School of MedicineNo. 1111 Xianxia Road, Changning DistrictShanghai200336P. R. China
| | - Wei Wei
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of SciencesNo. 1 Bei‐Er‐Tiao, Zhong‐Guan‐Cun, Haidian DistrictBeijing100190P. R. China
- School of Chemical EngineeringUniversity of Chinese Academy of SciencesNo. 19A Yuquan RoadBeijing100049P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of SciencesNo. 1 Bei‐Er‐Tiao, Zhong‐Guan‐Cun, Haidian DistrictBeijing100190P. R. China
- School of Chemical EngineeringUniversity of Chinese Academy of SciencesNo. 19A Yuquan RoadBeijing100049P. R. China
| |
Collapse
|
250
|
Obaid G, Samkoe K, Tichauer K, Bano S, Park Y, Silber Z, Hodge S, Callaghan S, Guirguis M, Mallidi S, Pogue B, Hasan T. Is Tumor Cell Specificity Distinct from Tumor Selectivity In Vivo?: A Quantitative NIR Molecular Imaging Analysis of Nanoliposome Targeting. NANO RESEARCH 2021; 14:1344-1354. [PMID: 33717420 PMCID: PMC7951968 DOI: 10.1007/s12274-020-3178-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The significance and ability for receptor targeted nanoliposomes (tNLs) to bind to their molecular targets in solid tumors in vivo has been questioned, particularly as the efficiency of their tumor accumulation and selectivity is not always predictive of their efficacy or molecular specificity. This study presents, for the first time, in situ NIR molecular imaging-based quantitation of the in vivo specificity of tNLs for their target receptors, as opposed to tumor selectivity, which includes influences of enhanced tumor permeability and retention. Results show that neither tumor delivery nor selectivity (tumor-to-normal ratio) of cetuximab and IRDye conjugated tNLs correlate with EGFR expression in U251, U87 and 9L tumors, and in fact underrepresent their imaging-derived molecular specificity by up to 94.2%. Conversely, their in vivo specificity, which we quantify as the concentration of tNL-reported tumor EGFR provided by NIR molecular imaging, correlates positively with EGFR expression levels in vitro and ex vivo (Pearson's r= 0.92 and 0.96, respectively). This study provides a unique opportunity to address the problematic disconnect between tNL synthesis and in vivo specificity. The findings encourage their continued adoption as platforms for precision medicine, and facilitates intelligent synthesis and patient customization in order to improve safety profiles and therapeutic outcomes.
Collapse
Affiliation(s)
- Girgis Obaid
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, U.S
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, U.S
| | - Kimberley Samkoe
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 037551, U.S
| | - Kenneth Tichauer
- Armour College of Engineering, Illinois Institute of Technology, Chicago, Illinois 60616, U.S
| | - Shazia Bano
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, U.S
| | - Yeonjae Park
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 037551, U.S
| | - Zachary Silber
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, U.S
| | - Sassan Hodge
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 037551, U.S
| | - Susan Callaghan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, U.S
| | - Mina Guirguis
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, U.S
| | - Srivalleesha Mallidi
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, U.S
| | - Brian Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 037551, U.S
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, U.S
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, U.S
| |
Collapse
|