1
|
Cui G, Deng S, Zhang B, Wang M, Lin Z, Lan X, Li Z, Yao G, Yu M, Yan J. Overcoming the Tumor Collagen Barriers: A Multistage Drug Delivery Strategy for DDR1-Mediated Resistant Colorectal Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402107. [PMID: 38953306 PMCID: PMC11434232 DOI: 10.1002/advs.202402107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 06/20/2024] [Indexed: 07/04/2024]
Abstract
The extracellular matrix (ECM) is critical for drug resistance in colorectal cancer (CRC). The abundant collagen within the ECM significantly influences tumor progression and matrix-mediated drug resistance (MMDR) by binding to discoidin domain receptor 1 (DDR1), but the specific mechanisms by which tumor cells modulate ECM via DDR1 and ultimately regulate TME remain poorly understand. Furthermore, overcoming drug resistance by modulating the tumor ECM remains a challenge in CRC treatment. In this study, a novel mechanism is elucidated by which DDR1 mediates the interactions between tumor cells and collagen, enhances collagen barriers, inhibits immune infiltration, promotes drug efflux, and leads to MMDR in CRC. To address this issue, a multistage drug delivery system carrying DDR1-siRNA and chemotherapeutic agents is employed to disrupt collagen barriers by silencing DDR1 in tumor, enhancing chemotherapy drugs diffusion and facilitating immune infiltration. These findings not only revealed a novel role for collagen-rich matrix mediated by DDR1 in tumor resistance, but also introduced a promising CRC treatment strategy.
Collapse
Affiliation(s)
- Guangman Cui
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal TumorNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Shaohui Deng
- The Tenth Affiliated Hospital of Southern Medical UniversityDongguanGuangdong523059China
| | - Biao Zhang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal TumorNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Manchun Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong‐Hongkong‐Macao Joint Laboratory for New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Zhousheng Lin
- Breast CenterDepartment of General SurgeryNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Xinyue Lan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong‐Hongkong‐Macao Joint Laboratory for New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Zelong Li
- Breast CenterDepartment of General SurgeryNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Guangyu Yao
- Breast CenterDepartment of General SurgeryNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Meng Yu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong‐Hongkong‐Macao Joint Laboratory for New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
- Zhujiang Hospital, Southern Medical UniversityGuangzhou510282China
| | - Jun Yan
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal TumorNanfang HospitalSouthern Medical UniversityGuangzhou510515China
- Department of Gastrointestinal SurgeryShenzhen People's HospitalSecond Clinical Medical College of Jinan UniversityFirst Affiliated Hospital of Southern University of Science and TechnologyShenzhenGuangdong518020China
| |
Collapse
|
2
|
Huang A, Li Q, Shi X, Gao J, Ma Y, Ding J, Hua S, Zhou W. An iron-containing nanomedicine for inducing deep tumor penetration and synergistic ferroptosis in enhanced pancreatic cancer therapy. Mater Today Bio 2024; 27:101132. [PMID: 38994471 PMCID: PMC11237974 DOI: 10.1016/j.mtbio.2024.101132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/05/2024] [Accepted: 06/16/2024] [Indexed: 07/13/2024] Open
Abstract
Pancreatic cancer is an aggressive and challenging malignancy with limited treatment options, largely attributed to the dense tumor stroma and intrinsic drug resistance. Here, we introduce a novel iron-containing nanoparticle formulation termed PTFE, loaded with the ferroptosis inducer Erastin, to overcome these obstacles and enhance pancreatic cancer therapy. The PTFE nanoparticles were prepared through a one-step assembly process, consisting of an Erastin-loaded PLGA core stabilized by a MOF shell formed by coordination between Fe3+ and tannic acid. PTFE demonstrated a unique capability to repolarize tumor-associated macrophages (TAMs) into the M1 phenotype, leading to the regulation of dense tumor stroma by modulating the activation of tumor-associated fibroblasts (TAFs) and reducing collagen deposition. This resulted in enhanced nanoparticle accumulation and deep penetration, as confirmed by in vitro multicellular tumor spheroids and in vivo mesenchymal-rich subcutaneous pancreatic tumor models. Moreover, PTFE effectively combated tumor resistance by synergistically employing the Fe3+-induced Fenton reaction and Erastin-induced ferroptosis, thereby disrupting the redox balance. As a result, significant tumor growth inhibition was achieved in mice-bearing tumor model. Comprehensive safety evaluations demonstrated PTFE's favorable biocompatibility, highlighting its potential as a promising therapeutic platform to effectively address the formidable challenges in pancreatic cancer treatment.
Collapse
Affiliation(s)
- Aiping Huang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Qingnian Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Xinyi Shi
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
- Department of Pharmacy, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine (Haikou People's Hospital), Haikou, Hainan, 570208, China
| | - Junyi Gao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yiran Ma
- Hunan Prize Life Science Research Institute Co., LTD, China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Surong Hua
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| |
Collapse
|
3
|
Lv D, Fei Y, Chen H, Wang J, Han W, Cui B, Feng Y, Zhang P, Chen J. Crosstalk between T lymphocyte and extracellular matrix in tumor microenvironment. Front Immunol 2024; 15:1340702. [PMID: 38690275 PMCID: PMC11058664 DOI: 10.3389/fimmu.2024.1340702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/26/2024] [Indexed: 05/02/2024] Open
Abstract
The extracellular matrix (ECM) is a complex three-dimensional structure composed of proteins, glycans, and proteoglycans, constituting a critical component of the tumor microenvironment. Complex interactions among immune cells, extracellular matrix, and tumor cells promote tumor development and metastasis, consequently influencing therapeutic efficacy. Hence, elucidating these interaction mechanisms is pivotal for precision cancer therapy. T lymphocytes are an important component of the immune system, exerting direct anti-tumor effects by attacking tumor cells or releasing lymphokines to enhance immune effects. The ECM significantly influences T cells function and infiltration within the tumor microenvironment, thereby impacting the behavior and biological characteristics of tumor cells. T cells are involved in regulating the synthesis, degradation, and remodeling of the extracellular matrix through the secretion of cytokines and enzymes. As a result, it affects the proliferation and invasive ability of tumor cells as well as the efficacy of immunotherapy. This review discusses the mechanisms underlying T lymphocyte-ECM interactions in the tumor immune microenvironment and their potential application in immunotherapy. It provides novel insights for the development of innovative tumor therapeutic strategies and drug.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jiao Chen
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Tian Y, Cheng T, Sun F, Zhou Y, Yuan C, Guo Z, Wang Z. Effect of biophysical properties of tumor extracellular matrix on intratumoral fate of nanoparticles: Implications on the design of nanomedicine. Adv Colloid Interface Sci 2024; 326:103124. [PMID: 38461766 DOI: 10.1016/j.cis.2024.103124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/11/2024] [Accepted: 03/04/2024] [Indexed: 03/12/2024]
Abstract
Nanomedicine has a profound impact on various research domains including drug delivery, diagnostics, theranostics, and regenerative medicine. Nevertheless, the clinical translation of nanomedicines for solid cancer remains limited due to the abundant physiological and pathological barriers in tumor that hinder the intratumoral penetration and distribution of these nanomedicines. In this article, we review the dynamic remodeling of tumor extracellular matrix during the tumor progression, discuss the impact of biophysical obstacles within tumors on the penetration and distribution of nanomedicines within the solid tumor and collect innovative approaches to surmount these obstacles for improving the penetration and accumulation of nanomedicines in tumor. Furthermore, we dissect the challenges and opportunities of the respective approaches, and propose potential avenues for future investigations. The purpose of this review is to provide a perspective guideline on how to effectively enhance the penetration of nanomedicines within tumors using promising methods.
Collapse
Affiliation(s)
- Yachao Tian
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China; Heilongjiang Guoru Biotechnology Co., Ltd., Xiangfang District, Harbin City 150030, China; School of Food Science and Engineering, Qilu University of Technology, Jinan, Shandong 250353, China
| | - Tianfu Cheng
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Fuwei Sun
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Yaxin Zhou
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Chao Yuan
- School of Food Science and Engineering, Qilu University of Technology, Jinan, Shandong 250353, China
| | - Zengwang Guo
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
| | - Zhongjiang Wang
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
| |
Collapse
|
5
|
Cao R, Tian H, Tian Y, Fu X. A Hierarchical Mechanotransduction System: From Macro to Micro. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302327. [PMID: 38145330 PMCID: PMC10953595 DOI: 10.1002/advs.202302327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 10/27/2023] [Indexed: 12/26/2023]
Abstract
Mechanotransduction is a strictly regulated process whereby mechanical stimuli, including mechanical forces and properties, are sensed and translated into biochemical signals. Increasing data demonstrate that mechanotransduction is crucial for regulating macroscopic and microscopic dynamics and functionalities. However, the actions and mechanisms of mechanotransduction across multiple hierarchies, from molecules, subcellular structures, cells, tissues/organs, to the whole-body level, have not been yet comprehensively documented. Herein, the biological roles and operational mechanisms of mechanotransduction from macro to micro are revisited, with a focus on the orchestrations across diverse hierarchies. The implications, applications, and challenges of mechanotransduction in human diseases are also summarized and discussed. Together, this knowledge from a hierarchical perspective has the potential to refresh insights into mechanotransduction regulation and disease pathogenesis and therapy, and ultimately revolutionize the prevention, diagnosis, and treatment of human diseases.
Collapse
Affiliation(s)
- Rong Cao
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Huimin Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Yan Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Xianghui Fu
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| |
Collapse
|
6
|
Luo Z, He Y, Li M, Ge Y, Huang Y, Liu X, Hou J, Zhou S. Tumor Microenvironment-Inspired Glutathione-Responsive Three-Dimensional Fibrous Network for Efficient Trapping and Gentle Release of Circulating Tumor Cells. ACS APPLIED MATERIALS & INTERFACES 2023; 15:24013-24022. [PMID: 37178127 DOI: 10.1021/acsami.3c00307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Detection of circulating tumor cells (CTCs) is important for early cancer diagnosis, prediction of postoperative recurrence, and individualized treatment. However, it is still challenging to achieve efficient capture and gentle release of CTCs from the complex peripheral blood due to their rarity and fragility. Herein, inspired by the three-dimensional (3D) network structure and high glutathione (GSH) level of the tumor microenvironment (TME), a 3D stereo (3D-G@FTP) fibrous network is developed by combining the liquid-assisted electrospinning method, gas foaming technique, and metal-polyphenol coordination interactions to achieve efficient trapping and gentle release of CTCs. Compared with the traditional 2D@FTP fibrous scaffold, the 3D-G@FTP fibrous network could achieve higher capture efficiency (90.4% vs 78.5%) toward cancer cells in a shorter time (30 min vs 90 min). This platform showed superior capture performance toward heterogeneous cancer cells (HepG2, HCT116, HeLa, and A549) in an epithelial cell adhesion molecule (EpCAM)-independent manner. In addition, the captured cells with high cell viability (>90.0%) could be gently released under biologically friendly GSH stimulus. More importantly, the 3D-G@FTP fibrous network could sensitively detect 4-19 CTCs from six kinds of cancer patients' blood samples. We expect this TME-inspired 3D stereo fibrous network integrating efficient trapping, broad-spectrum recognition, and gentle release will promote the development of biomimetic devices for rare cell analysis.
Collapse
Affiliation(s)
- Zhouying Luo
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Yang He
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Ming Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Yumeng Ge
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Yisha Huang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Xia Liu
- School of Chemistry, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Jianwen Hou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P. R. China
| |
Collapse
|
7
|
Chen W, Li C, Jiang X. Advanced Biomaterials with Intrinsic Immunomodulation Effects for Cancer Immunotherapy. SMALL METHODS 2023; 7:e2201404. [PMID: 36811240 DOI: 10.1002/smtd.202201404] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/17/2023] [Indexed: 05/17/2023]
Abstract
In recent years, tumor immunotherapy has achieved significant success in tumor treatment based on immune checkpoint blockers and chimeric antigen receptor T-cell therapy. However, about 70-80% of patients with solid tumors do not respond to immunotherapy due to immune evasion. Recent studies found that some biomaterials have intrinsic immunoregulatory effects, except serve as carriers for immunoregulatory drugs. Moreover, these biomaterials have additional advantages such as easy functionalization, modification, and customization. In this review, the recent advances of these immunoregulatory biomaterials in cancer immunotherapy and their interaction with cancer cells, immune cells, and the immunosuppressive tumor microenvironment are summarized. Finally, the opportunities and challenges of immunoregulatory biomaterials used in the clinic and the prospect of their future in cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Weizhi Chen
- MOE Key Laboratory of High Performance Polymer Materials and Technology and Department of Polymer Science and Engineering, College of Chemistry and Chemical Engineering, Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210023, P. R. China
| | - Cheng Li
- MOE Key Laboratory of High Performance Polymer Materials and Technology and Department of Polymer Science and Engineering, College of Chemistry and Chemical Engineering, Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210023, P. R. China
| | - Xiqun Jiang
- MOE Key Laboratory of High Performance Polymer Materials and Technology and Department of Polymer Science and Engineering, College of Chemistry and Chemical Engineering, Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
8
|
Dzobo K, Dandara C. The Extracellular Matrix: Its Composition, Function, Remodeling, and Role in Tumorigenesis. Biomimetics (Basel) 2023; 8:146. [PMID: 37092398 PMCID: PMC10123695 DOI: 10.3390/biomimetics8020146] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/25/2023] Open
Abstract
The extracellular matrix (ECM) is a ubiquitous member of the body and is key to the maintenance of tissue and organ integrity. Initially thought to be a bystander in many cellular processes, the extracellular matrix has been shown to have diverse components that regulate and activate many cellular processes and ultimately influence cell phenotype. Importantly, the ECM's composition, architecture, and stiffness/elasticity influence cellular phenotypes. Under normal conditions and during development, the synthesized ECM constantly undergoes degradation and remodeling processes via the action of matrix proteases that maintain tissue homeostasis. In many pathological conditions including fibrosis and cancer, ECM synthesis, remodeling, and degradation is dysregulated, causing its integrity to be altered. Both physical and chemical cues from the ECM are sensed via receptors including integrins and play key roles in driving cellular proliferation and differentiation and in the progression of various diseases such as cancers. Advances in 'omics' technologies have seen an increase in studies focusing on bidirectional cell-matrix interactions, and here, we highlight the emerging knowledge on the role played by the ECM during normal development and in pathological conditions. This review summarizes current ECM-targeted therapies that can modify ECM tumors to overcome drug resistance and better cancer treatment.
Collapse
Affiliation(s)
- Kevin Dzobo
- Medical Research Council, SA Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
| | - Collet Dandara
- Division of Human Genetics and Institute of Infectious Disease and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
- The South African Medical Research Council-UCT Platform for Pharmacogenomics Research and Translation, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
| |
Collapse
|
9
|
Zhou S, Li C, Yuan Y, Jiang L, Chen W, Jiang X. Dendritic lipopeptide liposomes decorated with dual-targeted proteins. Biomater Sci 2022; 10:7032-7041. [PMID: 36318065 DOI: 10.1039/d2bm00952h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Due to their homing effects, cell and cell membrane-derived nanocarriers have been widely used to enhance drug target delivery. Inspired by the protein-anchored cell membrane architecture, we here report a tumor-targeted liposome, dtDLP, which was constructed through the electrostatic interaction between dendritic lipopeptide liposomes and a dual-targeted recombinant protein, achieving superior tumor homing, cellular endocytotic and penetration abilities. The dual-targeted recombinant protein consists of an anti-epidermal growth factor receptor single domain antibody and a peptide ligand for the integrin αvβ3. dtDLPs substantially reduced macrophage phagocytosis and increased drug internalization in both 4T1 cells and HeLa cells by providing more endocytic pathways. In addition, the dtDLPs showed great penetration ability in both multicellular spheroids and tumor tissues. Due to the improved cancer cellular uptake and tumor penetration, the dtDLPs exhibited a superior anticancer effect in both HeLa and 4T1 tumor-bearing mice. This work will be helpful for the design of cell-specific liposomes with admirable tumor targeting, endocytotic and penetration abilities.
Collapse
Affiliation(s)
- Sensen Zhou
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Cheng Li
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Yang Yuan
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Lei Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China. .,State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Weizhi Chen
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| | - Xiqun Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, and Jiangsu Key Laboratory for Nanotechnology, Nanjing University, Nanjing, 210093, P.R. China.
| |
Collapse
|
10
|
Yuan Y, Zhou S, Li C, Zhang X, Mao H, Chen W, Jiang X. Cascade Downregulation of the HER Family by a Dual-Targeted Recombinant Protein-Drug Conjugate to Inhibit Tumor Growth and Metastasis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2201558. [PMID: 35365900 DOI: 10.1002/adma.202201558] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/30/2022] [Indexed: 06/14/2023]
Abstract
Human epidermal growth factor receptor type 2 (HER2)-targeted therapy can significantly improve the outcome of patients with HER2 positive cancer. However, relapse after this treatment remains a great challenge in the clinic due to tumor resistance, in which the HER network induces constitutive signal transduction. In addition, integrin receptors in the tumor extracellular matrix can mitigate the therapeutic effect of inhibitors to the growth factors receptors and tyrosine kinases. Here, the development of a recombinant protein (RP-HI) and its drug conjugates (RPDC-HI) to target both HER2 and integrin is reported. When simultaneously blocking HER2 and integrin by RP-HI, functions of the HER family and their interactions with the integrin are disrupted by downregulated expressions of HER family members, leading to inhibition of several downstream signal pathways. In combination with targeted delivery of the anticancer agent, doxorubicin (DOX), RPDC-HI significantly improves the tumor inhibition efficacy to 97.5% in treating HER2-positive breast cancer, comparing to 34.3% for free DOX. RPDC-HI shows even better antitumor efficiency than a monoclonal antibody, trastuzumab, when treating larger tumors. The developed dual-targeted RPDC platform offers a new and promising strategy for treating HER2-positive patients with synergistic therapeutic effects against tumor resistance to the conventional HER2-targeted treatment.
Collapse
Affiliation(s)
- Yang Yuan
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science & Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, China
| | - Sensen Zhou
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science & Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, China
| | - Cheng Li
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science & Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, China
| | - Xiaoke Zhang
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science & Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, China
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA
| | - Weizhi Chen
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science & Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, China
| | - Xiqun Jiang
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science & Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
11
|
Barone A, d’Avanzo N, Cristiano MC, Paolino D, Fresta M. Macrophage-Derived Extracellular Vesicles: A Promising Tool for Personalized Cancer Therapy. Biomedicines 2022; 10:1252. [PMID: 35740274 PMCID: PMC9220135 DOI: 10.3390/biomedicines10061252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022] Open
Abstract
The incidence of cancer is increasing dramatically, affecting all ages of the population and reaching an ever higher worldwide mortality rate. The lack of therapies' efficacy is due to several factors such as a delay in diagnosis, tumor regrowth after surgical resection and the occurrence of multidrug resistance (MDR). Tumor-associated immune cells and the tumor microenvironment (TME) deeply affect the tumor's progression, leading to several physicochemical changes compared to physiological conditions. In this scenario, macrophages play a crucial role, participating both in tumor suppression or progression based on the polarization of onco-suppressive M1 or pro-oncogenic M2 phenotypes. Moreover, much evidence supports the pivotal role of macrophage-derived extracellular vesicles (EVs) as mediators in TME, because of their ability to shuttle the cell-cell and organ-cell communications, by delivering nucleic acids and proteins. EVs are lipid-based nanosystems with a broad size range distribution, which reflect a similar composition of native parent cells, thus providing a natural selectivity towards target sites. In this review, we discuss the impact of macrophage-derived EVs in the cancer's fate as well as their potential implications for the development of personalized anticancer nanomedicine.
Collapse
Affiliation(s)
- Antonella Barone
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro Campus Universitario-Germaneto, Viale Europa, 88100 Catanzaro, Italy; (A.B.); (M.C.C.)
| | - Nicola d’Avanzo
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini n.31, 66100 Chieti, Italy;
| | - Maria Chiara Cristiano
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro Campus Universitario-Germaneto, Viale Europa, 88100 Catanzaro, Italy; (A.B.); (M.C.C.)
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro Campus Universitario-Germaneto, Viale Europa, 88100 Catanzaro, Italy; (A.B.); (M.C.C.)
| | - Massimo Fresta
- Department of Health Science, University “Magna Græcia” of Catanzaro Campus Universitario-Germaneto, Viale Europa, 88100 Catanzaro, Italy;
| |
Collapse
|