1
|
Du L, Shen H, Xu C, Zhu X, Wang B, Zhou Q, Liu C, Sung HHY, Kwok RTK, Lam JWY, Zhou Q, Liu T, Tang BZ. From Molecule to Aggregate: Designing AIE Nanocrystals for Low-Power Backward Third-Harmonic Generation Angiography. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414419. [PMID: 40165774 PMCID: PMC12087731 DOI: 10.1002/adma.202414419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/17/2025] [Indexed: 04/02/2025]
Abstract
Organic materials featuring third harmonic generation (THG) hold great promise for deep-tissue bioimaging due to their good biocompatibility and second near-infrared excitation. However, minimizing photodamage from the incident light necessitates significant improvements in the third-order nonlinear susceptibility. Herein, an organic luminogen called OTBP is developed as a backward THG (BTHG) contrast agent for second near-infrared (NIR-II) angiography. OTBP's intense absorption at 433 nm resonantly enhances its BTHG efficiency when excited by a 1300 nm femtosecond laser. In the aggregate state, the robust intermolecular interactions among OTBP molecules realize excellent crystallinity and the facile preparation of nanocrystals (NCs) with a high refractive index of 1.78. By leveraging Mie scattering theory, the best size of OTBP NCs for BTHG collection is attained. These integrated properties result in a high BTHG efficiency of OTBP NCs. Encapsulating the NCs with F-127 enables ultralow-power but high-contrast 3D vasculature imaging with negligible photodamage and background interference. Further elevating the laser power to 60 mW enables the visualization of microvessels at 500 µm with a high SNR of 143. This study offers insights into material design strategies toward efficient organic BTHG contrast agents and paves the way for the materials-oriented non-linear optics.
Collapse
Affiliation(s)
- Lidong Du
- MOE Frontiers Science Center for Precision OncologyUniversity of MacauMacau999078China
- Faculty of Health SciencesUniversity of MacauMacau999078China
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular NeuroscienceGuangdong‐Hong Kong‐Macau Joint Laboratory of Optoelectronic and Magnetic Functional MaterialsThe Hong Kong University of Science and TechnologyClear Water Bay, KowloonHong Kong999077China
| | - Hanchen Shen
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular NeuroscienceGuangdong‐Hong Kong‐Macau Joint Laboratory of Optoelectronic and Magnetic Functional MaterialsThe Hong Kong University of Science and TechnologyClear Water Bay, KowloonHong Kong999077China
| | - Changhuo Xu
- MOE Frontiers Science Center for Precision OncologyUniversity of MacauMacau999078China
- Faculty of Health SciencesUniversity of MacauMacau999078China
| | - Xinyan Zhu
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular NeuroscienceGuangdong‐Hong Kong‐Macau Joint Laboratory of Optoelectronic and Magnetic Functional MaterialsThe Hong Kong University of Science and TechnologyClear Water Bay, KowloonHong Kong999077China
| | - Bingnan Wang
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular NeuroscienceGuangdong‐Hong Kong‐Macau Joint Laboratory of Optoelectronic and Magnetic Functional MaterialsThe Hong Kong University of Science and TechnologyClear Water Bay, KowloonHong Kong999077China
| | - Qingqing Zhou
- MOE Frontiers Science Center for Precision OncologyUniversity of MacauMacau999078China
- Faculty of Health SciencesUniversity of MacauMacau999078China
| | - Chunxi Liu
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular NeuroscienceGuangdong‐Hong Kong‐Macau Joint Laboratory of Optoelectronic and Magnetic Functional MaterialsThe Hong Kong University of Science and TechnologyClear Water Bay, KowloonHong Kong999077China
| | - Herman H. Y. Sung
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular NeuroscienceGuangdong‐Hong Kong‐Macau Joint Laboratory of Optoelectronic and Magnetic Functional MaterialsThe Hong Kong University of Science and TechnologyClear Water Bay, KowloonHong Kong999077China
| | - Ryan T. K. Kwok
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular NeuroscienceGuangdong‐Hong Kong‐Macau Joint Laboratory of Optoelectronic and Magnetic Functional MaterialsThe Hong Kong University of Science and TechnologyClear Water Bay, KowloonHong Kong999077China
| | - Jacky W. Y. Lam
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular NeuroscienceGuangdong‐Hong Kong‐Macau Joint Laboratory of Optoelectronic and Magnetic Functional MaterialsThe Hong Kong University of Science and TechnologyClear Water Bay, KowloonHong Kong999077China
| | - Quan Zhou
- Department of Cell BiologyDepartment of Gastroenterology of the Fourth Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310058China
| | - Tzu‐Ming Liu
- MOE Frontiers Science Center for Precision OncologyUniversity of MacauMacau999078China
- Faculty of Health SciencesUniversity of MacauMacau999078China
| | - Ben Zhong Tang
- Department of ChemistryHong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and ReconstructionDivision of Life ScienceState Key Laboratory of Molecular NeuroscienceGuangdong‐Hong Kong‐Macau Joint Laboratory of Optoelectronic and Magnetic Functional MaterialsThe Hong Kong University of Science and TechnologyClear Water Bay, KowloonHong Kong999077China
- School of Science and EngineeringShenzhen Institute of Aggregate Science and TechnologyThe Chinese University of Hong Kong (CUHK‐Shenzhen)ShenzhenGuangdong518172China
| |
Collapse
|
2
|
Mossburg KJ, Barragan D, O NH, Kian AC, Maidment ADA, Cormode DP. Emerging nanoparticle-based x-ray imaging contrast agents for breast cancer screening. Nanomedicine (Lond) 2025; 20:1149-1166. [PMID: 40261216 PMCID: PMC12068350 DOI: 10.1080/17435889.2025.2496129] [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: 02/05/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025] Open
Abstract
Breast cancer is one of the most common types of cancer, however, preventive screening has contributed to a significant reduction in mortality over the past four decades. The first-line screening methods for breast cancer, such as mammography and tomosynthesis, are x-ray-based modalities. Unfortunately, their cancer detection rates are low in patients with dense breasts. These, and other high-risk women, are now encouraged to receive supplemental screening. The supplemental imaging methods are diverse, including ultrasound, MRI, nuclear imaging, and X-ray-based modalities such as breast CT and contrast-enhanced mammography/tomosynthesis. Due to their low cost and wide availability, x-ray-based modalities see significant clinical use worldwide. These techniques benefit from the use of contrast agents, which are currently iodinated small molecules designed for other purposes. Consequently, developing new contrast agents that are specifically for breast cancer screening is of interest. This review describes these modalities and the nanoparticle-based contrast agents being researched for their enhanced performance. The relevant parameters for nanoparticle-based contrast agent design are evaluated, including contrast generation and potential biointeractions. Iodinated agents are discussed for comparison. Nanoparticles covered include silver sulfide, silver telluride, gold, and bismuth sulfide-based agents, among others. Finally, perspectives on future developments in this field are offered.
Collapse
Affiliation(s)
- Katherine J. Mossburg
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Diego Barragan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathaniel H. O
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pharmaceutical Sciences, St. Joseph’s University, Philadelphia, PA, USA
- Department of Physics, St. Joseph’s University, Philadelphia, PA, USA
| | - Andrea C. Kian
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew D. A. Maidment
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - David P. Cormode
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
3
|
Panja P, Manne U, Awasthi V, Bhattacharya R, Mukherjee P. Interrogation of the tumor microenvironment by nanoparticles. Cancer Lett 2025; 612:217454. [PMID: 39805387 DOI: 10.1016/j.canlet.2025.217454] [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: 10/25/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
The tumor microenvironment (TME) plays a pivotal role in cancer progression by fostering intricate multicellular crosstalk among cancer cells, stromal cells, and immune cells. This review explores the emerging paradigm of utilizing nanoparticles to disrupt this crosstalk within the TME as a therapeutic strategy. Nanoparticles are engineered with precise physicochemical properties to target specific cell types and deliver therapeutic payloads, thereby inhibiting critical signaling pathways involved in tumor growth, invasion, and metastasis. The mechanisms involved include modulation of the immune response, interference with growth factor signaling, and induction of programmed cell death in cancer cells. Challenges such as biocompatibility, efficient delivery, and potential development of resistance are discussed alongside promising advancements in nanoparticle design. Moving forward, integration of nanoparticle-based therapies with existing treatment modalities holds great potential for enhancing therapeutic efficacy and personalized medicine in cancer therapy.
Collapse
Affiliation(s)
- Prasanta Panja
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vibhudutta Awasthi
- Department of Pharmaceutical Sciences, University of Oklahoma Health Science Center, Suite 309, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Priyabrata Mukherjee
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
4
|
Xia W, Tan Y, Liu Y, Xie N, Zhu H. Prospect of extracellular vesicles in tumor immunotherapy. Front Immunol 2025; 16:1525052. [PMID: 40078996 PMCID: PMC11897508 DOI: 10.3389/fimmu.2025.1525052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/28/2025] [Indexed: 03/14/2025] Open
Abstract
Extracellular vesicles (EVs), as cell-derived small vesicles, facilitate intercellular communication within the tumor microenvironment (TME) by transporting biomolecules. EVs from different sources have varied contents, demonstrating differentiated functions that can either promote or inhibit cancer progression. Thus, regulating the formation, secretion, and intake of EVs becomes a new strategy for cancer intervention. Advancements in EV isolation techniques have spurred interest in EV-based therapies, particularly for tumor immunotherapy. This review explores the multifaceted functions of EVs from various sources in tumor immunotherapy, highlighting their potential in cancer vaccines and adoptive cell therapy. Furthermore, we explore the potential of EVs as nanoparticle delivery systems in tumor immunotherapy. Finally, we discuss the current state of EVs in clinical settings and future directions, aiming to provide crucial information to advance the development and clinical application of EVs for cancer treatment.
Collapse
Affiliation(s)
- Wenbo Xia
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yunhan Tan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yongen Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Huili Zhu
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Tang Y, Shen Q, Lin P, Chen Z, Fan D, Zhuo M, Gan Y, Su Y, Qian Q, Lin L, Xue E, Chen Z. aPD-L1-facilitated theranostic and tumor microenvironment remodeling of pancreatic cancer via docetaxel-loaded phase-transformation nanoparticles triggered by low-intensity pulsed ultrasound. J Nanobiotechnology 2025; 23:48. [PMID: 39871305 PMCID: PMC11773723 DOI: 10.1186/s12951-025-03105-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025] Open
Abstract
Early diagnosis of pancreatic ductal adenocarcinoma (PDAC) is challenging because of its depth, which often leads to misdiagnosis during ultrasound examinations. The unique PDAC tumor microenvironment (TME) is characterized by significant fibrous tissue growth, and high interstitial pressure hinders drug penetration into tumors. Additionally, hypoxia and immune suppression within the tumor contribute to poor responses to radiotherapy and chemotherapy, ultimately leading to an unfavorable prognosis. In this study, aPD-L1-modified docetaxel and perfluoropentane-loaded liquid‒vapor phase-transformation lipid nanoparticles (aPDL1-DTX/PFP@Lipid) were synthesized and had an average diameter of 61.63 nm with 84.3% antibody modification. We demonstrated that the nanoparticles (NPs) exhibited excellent PDAC-targeting capabilities both in vitro and in vivo. Upon exposure to low-intensity pulsed ultrasound (LIPUS) stimulation, the NPs underwent a phase transformation to form microbubbles with substantial molecular ultrasound diagnostic effects, and combined treatment resulted in a tumor growth inhibition rate of 88.91%. This treatment strategy also led to the infiltration of CD8+ T cells, the downregulation of Treg cells, the promotion of M1 macrophage polarization, the inhibition of fibrosis to reduce tumor stromal pressure, and the facilitation of perfluoropentane (PFP) gasification to release O2 and improve tumor hypoxia. In conclusion, aPD-L1-modified liquid‒vapor phase-transformation nanoparticles loaded with docetaxel (DTX) and PFP were successfully combined with ultrasound for the molecular diagnosis and targeted treatment of PDAC. aPDL1-DTX/PFP@Lipid could reshape the PDAC TME, offering a new approach for ultrasound-mediated diagnosis and treatment with promising clinical applications.
Collapse
Affiliation(s)
- Yi Tang
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
- Fujian Institute of Ultrasonic Medicine Laboratory, 29 Xinquan Road, Fuzhou, China
| | - Qingling Shen
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
- Fujian Institute of Ultrasonic Medicine Laboratory, 29 Xinquan Road, Fuzhou, China
| | - Peng Lin
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Zhixin Chen
- Fujian College Association Instrumental Analysis Center, Fuzhou University, 2 Xueyuan Road, Fuzhou, China
| | - Denghui Fan
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, China
| | - Minling Zhuo
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Yajiao Gan
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Yixi Su
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Qingfu Qian
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Liwu Lin
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Ensheng Xue
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China
| | - Zhikui Chen
- Department of Ultrasound, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, China.
- Fujian Institute of Ultrasonic Medicine Laboratory, 29 Xinquan Road, Fuzhou, China.
| |
Collapse
|
6
|
Zhang X, Dong T, Li X, Xu C, Chen F, Wang S, Wang X. Design, Synthesis, and Evaluation of Doxifluridine Derivatives as Nitroreductase-Responsive Anticancer Prodrugs. Molecules 2024; 29:5077. [PMID: 39519718 PMCID: PMC11547703 DOI: 10.3390/molecules29215077] [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: 09/19/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Antimetabolite antitumor drugs interfere with nucleic acid and DNA synthesis, causing cancer cell death. However, they also affect rapidly dividing normal cells and cause serious side effects. Doxifluridine (5'-deoxy-5-fluorouridine [5'-DFUR]), a 5-fluorouracil (5-FU) prodrug converted to 5-FU by thymidine phosphorylase (TP), exerts antitumor effects. Since TP is distributed in tumor and normal tissues, 5'-DFUR features side effects. Here we designed a series of novel 5'-DFUR derivatives based on high nitroreductase (NTR) levels in the hypoxic microenvironment of tumor tissues by introducing nitro-containing moieties into the 5'-DFUR structure. These derivatives exert their antitumor effects by producing 5-FU under the dual action of TP and NTR in the tumor microenvironment. The derivatives were synthesized and their stability, release, and cytotoxicity evaluated in vitro and antitumor activity evaluated in vivo. Compound 2c, featuring nitrofuran fragments, was stable in phosphate-buffered saline and plasma at different pH values and reduced rapidly in the presence of NTR. The in vitro cytotoxicity evaluation indicated that compound 2c showed excellent selectivity in the MCF-7 and HT29 cell lines. Moreover, it exhibited antitumor effects comparable to those of 5'-DFUR in vivo without significant toxic side effects. These results suggest that compound 2c is a promising antitumor prodrug.
Collapse
Affiliation(s)
| | | | | | | | | | - Shiben Wang
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, 1 Hunan Street, Liaocheng 252059, China; (X.Z.); (T.D.); (X.L.); (C.X.); (F.C.)
| | - Xuekun Wang
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, 1 Hunan Street, Liaocheng 252059, China; (X.Z.); (T.D.); (X.L.); (C.X.); (F.C.)
| |
Collapse
|
7
|
Xiao S, Mu M, Feng C, Pan S, Chen N. The application of bacteria-nanomaterial hybrids in antitumor therapy. J Nanobiotechnology 2024; 22:536. [PMID: 39227831 PMCID: PMC11373302 DOI: 10.1186/s12951-024-02793-x] [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: 07/01/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024] Open
Abstract
Adverse effects and multidrug resistance remain significant obstacles in conventional cancer therapy. Nanomedicines, with their intrinsic properties such as nano-sized dimensions and tunable surface characteristics, have the potential to mitigate the side effects of traditional cancer treatments. While nanomaterials have been widely applied in cancer treatment, challenges such as low targeting efficiency and poor tumor penetration persist. Recent research has shown that anaerobic bacteria exhibit high selectivity for primary tumors and metastatic cancers, offering good safety and superior tumor penetration capabilities. This suggests that combining nanomaterials with bacteria could complement their respective limitations, opening vast potential applications in cancer therapy. The use of bacteria in combination with nanomaterials for anticancer treatments, including chemotherapy, radiotherapy, and photothermal/photodynamic therapy, has contributed to the rapid development of the field of bacterial oncology treatments. This review explores the mechanisms of bacterial tumor targeting and summarizes strategies for synthesizing bacterial-nanomaterial and their application in cancer therapy. The combination of bacterial-nanomaterial hybrids with modern therapeutic approaches represents a promising avenue for future cancer treatment research, with the potential to improve treatment outcomes for cancer patients.
Collapse
Affiliation(s)
- Susu Xiao
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Min Mu
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chenqian Feng
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shulin Pan
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Nianyong Chen
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
8
|
de Roode KE, Hashemi K, Verdurmen WPR, Brock R. Tumor-On-A-Chip Models for Predicting In Vivo Nanoparticle Behavior. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402311. [PMID: 38700060 DOI: 10.1002/smll.202402311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Indexed: 05/05/2024]
Abstract
Nanosized drug formulations are broadly explored for the improvement of cancer therapy. Prediction of in vivo nanoparticle (NP) behavior, however, is challenging, given the complexity of the tumor and its microenvironment. Microfluidic tumor-on-a-chip models are gaining popularity for the in vitro testing of nanoparticle targeting under conditions that simulate the 3D tumor (microenvironment). In this review, following a description of the tumor microenvironment (TME), the state of the art regarding tumor-on-a-chip models for investigating nanoparticle delivery to solid tumors is summarized. The models are classified based on the degree of compartmentalization (single/multi-compartment) and cell composition (tumor only/tumor microenvironment). The physiological relevance of the models is critically evaluated. Overall, microfluidic tumor-on-a-chip models greatly improve the simulation of the TME in comparison to 2D tissue cultures and static 3D spheroid models and contribute to the understanding of nanoparticle behavior. Interestingly, two interrelated aspects have received little attention so far which are the presence and potential impact of a protein corona as well as nanoparticle uptake through phagocytosing cells. A better understanding of their relevance for the predictive capacity of tumor-on-a-chip systems and development of best practices will be a next step for the further refinement of advanced in vitro tumor models.
Collapse
Affiliation(s)
- Kim E de Roode
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Khadijeh Hashemi
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Wouter P R Verdurmen
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Roland Brock
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
- Department of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, 329, Bahrain
| |
Collapse
|
9
|
Gabriel EM, Sukniam K, Popp K, Kowkabany G, Manaise HK, Attwood K, George A, Zhai Q, Bagaria SP, Knutson KL, Skitzki JJ, Robertson MW, Dinh TA. Human tumour vessel heterogeneity in ovarian cancer and its association with response to neoadjuvant chemotherapy. Clin Transl Med 2024; 14:e1633. [PMID: 38616706 PMCID: PMC11016937 DOI: 10.1002/ctm2.1633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 04/16/2024] Open
Affiliation(s)
- Emmanuel M. Gabriel
- Department of General SurgeryDivision of Surgical OncologyMayo ClinicJacksonvilleFloridaUSA
| | - Kulkaew Sukniam
- Department of SurgeryPhiladelphia College of Osteopathic MedicineSuwaneeGeorgiaUSA
| | - Kyle Popp
- Department of BiologyFlorida State UniversityTallahasseeFloridaUSA
| | | | - Harsheen K. Manaise
- Department of MedicineGovernment Medical College and HospitalChandigarhIndia
| | - Kristopher Attwood
- Department of BiostatisticsRoswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Anthony George
- Department of BiostatisticsRoswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Qihui Zhai
- Department of PathologyMayo ClinicJacksonvilleFloridaUSA
| | - Sanjay P. Bagaria
- Department of General SurgeryDivision of Surgical OncologyMayo ClinicJacksonvilleFloridaUSA
| | | | - Joseph J. Skitzki
- Department of Surgical OncologyRoswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | | | - Tri A. Dinh
- Department of Gynecological OncologyMayo ClinicJacksonvilleFloridaUSA
| |
Collapse
|
10
|
Yang J, Shi X, Kuang Y, Wei R, Feng L, Chen J, Wu X. Cell-nanocarrier drug delivery system: a promising strategy for cancer therapy. Drug Deliv Transl Res 2024; 14:581-596. [PMID: 37721694 DOI: 10.1007/s13346-023-01429-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 09/19/2023]
Abstract
Tumor targeting has been a great challenge for drug delivery systems. A number of nanotechnology-derived drug carriers have been developed for cancer treatment to improve efficacy and biocompatibility. Among them, the emergence of cell-nanocarriers has attracted great attention, which simulates cell function and has good biocompatibility. They can also escape the clearance of reticuloendothelial system, showing a long-cycle effect. The inherent tumor migration and tumor homing ability of cells increase their significance as tumor-targeting vectors. In this review, we focus on the combination of stem cells, immune cells, red blood cells, and cell membranes to nanocarriers, which enable chemotherapy agents to efficiently target lesion sites and improve drug distribution while being low toxic and safe. In addition, we discuss the pros and cons of these nanoparticles as well as the challenges and opportunities that lie ahead. Although research to address these limitations is still ongoing, this promising tumor-targeted drug delivery system will provide a safe and effective platform against cancer.
Collapse
Affiliation(s)
- Jiefen Yang
- Fujian University of Traditional Chinese Medicine, No. 1, Qiuyang Road, Fuzhou, Fujian, People's Republic of China
- Shanghai Wei Er Lab, Shanghai, China
| | - Xiongxi Shi
- Fujian University of Traditional Chinese Medicine, No. 1, Qiuyang Road, Fuzhou, Fujian, People's Republic of China
- Shanghai Wei Er Lab, Shanghai, China
| | - Yanting Kuang
- Shanghai Wei Er Lab, Shanghai, China
- Inner Mongolia Medical University, No. 5, Xinhua Road, Hohhot, Inner Mongolia, People's Republic of China
| | - Ruting Wei
- Fujian University of Traditional Chinese Medicine, No. 1, Qiuyang Road, Fuzhou, Fujian, People's Republic of China
- Shanghai Wei Er Lab, Shanghai, China
| | - Lanni Feng
- Fujian University of Traditional Chinese Medicine, No. 1, Qiuyang Road, Fuzhou, Fujian, People's Republic of China
- Shanghai Wei Er Lab, Shanghai, China
| | - Jianming Chen
- Fujian University of Traditional Chinese Medicine, No. 1, Qiuyang Road, Fuzhou, Fujian, People's Republic of China.
- Shanghai Wei Er Lab, Shanghai, China.
| | - Xin Wu
- Fujian University of Traditional Chinese Medicine, No. 1, Qiuyang Road, Fuzhou, Fujian, People's Republic of China.
- Shanghai Wei Er Lab, Shanghai, China.
| |
Collapse
|
11
|
Wu L, Liu Y, Zeng W, Ishigaki Y, Zhou S, Wang X, Sun Y, Zhang Y, Jiang X, Suzuki T, Ye D. Smart Lipid Nanoparticle that Remodels Tumor Microenvironment for Activatable H 2S Gas and Photodynamic Immunotherapy. J Am Chem Soc 2023; 145:27838-27849. [PMID: 38059465 DOI: 10.1021/jacs.3c11328] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Hydrogen sulfide (H2S) has shown promise for gas therapy. However, it is still controversial whether H2S can remodel the tumor microenvironment (TME) and induce robust antitumor immunity. Here, a tumor-targeting and TME-responsive "smart" lipid nanoparticle (1-JK-PS-FA) is presented, which is capable of delivering and releasing H2S specifically in tumor tissues for on-demand H2S gas and photodynamic immunotherapy. 1-JK-PS-FA enables a burst release of H2S in the acidic TME, which promptly reduces the embedded organic electrochromic materials and consequently switches on near-infrared fluorescence and photodynamic activity. Furthermore, we found that high levels of H2S can reprogram the TME by reducing tumor interstitial fluid pressure, promoting angiogenesis, increasing vascular permeability, ameliorating hypoxia, and reducing immunosuppressive conditions. This leads to increased tumor uptake of 1-JK-PS-FA, thereby enhancing PDT efficacy and eliciting strong immunogenic cell death during 808 nm laser irradiation. Therefore, 1-JK-PS-FA permits synergistic H2S gas and photodynamic immunotherapy, effectively eradicating orthotopic breast tumors and preventing tumor metastasis and recurrence. This work showcases the capacity of H2S to reprogram the TME to enhance H2S gas and immunotherapy.
Collapse
Affiliation(s)
- Luyan Wu
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), School of Materials Science and Engineering, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Yili Liu
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Wenhui Zeng
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yusuke Ishigaki
- Department of Chemistry, Faculty of Science, Hokkaido University, N10 W8, North-ward, Sapporo 060-0810, Japan
| | - Sensen Zhou
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- MOE Key Laboratory of High Performance Polymer Materials and Technology, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xingxing Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yidan Sun
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yan Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xiqun Jiang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- MOE Key Laboratory of High Performance Polymer Materials and Technology, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Takanori Suzuki
- Department of Chemistry, Faculty of Science, Hokkaido University, N10 W8, North-ward, Sapporo 060-0810, Japan
| | - Deju Ye
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
12
|
Lu H, Niu L, Yu L, Jin K, Zhang J, Liu J, Zhu X, Wu Y, Zhang Y. Cancer phototherapy with nano-bacteria biohybrids. J Control Release 2023; 360:133-148. [PMID: 37315693 DOI: 10.1016/j.jconrel.2023.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/24/2023] [Accepted: 06/03/2023] [Indexed: 06/16/2023]
Abstract
The utilization of light for therapeutic interventions, also known as phototherapy, has been extensively employed in the treatment of a wide range of illnesses, including cancer. Despite the benefits of its non-invasive nature, phototherapy still faces challenges pertaining to the delivery of phototherapeutic agents, phototoxicity, and light delivery. The incorporation of nanomaterials and bacteria in phototherapy has emerged as a promising approach that leverages the unique properties of each component. The resulting nano-bacteria biohybrids exhibit enhanced therapeutic efficacy when compared to either component individually. In this review, we summarize and discuss the various strategies for assembling nano-bacteria biohybrids and their applications in phototherapy. We provide a comprehensive overview of the properties and functionalities of nanomaterials and cells in the biohybrids. Notably, we highlight the roles of bacteria beyond their function as drug vehicles, particularly their capacity to produce bioactive molecules. Despite being in its early stage, the integration of photoelectric nanomaterials and genetically engineered bacteria holds promise as an effective biosystem for antitumor phototherapy. The utilization of nano-bacteria biohybrids in phototherapy is a promising avenue for future investigation, with the potential to enhance treatment outcomes for cancer patients.
Collapse
Affiliation(s)
- Hongfei Lu
- Department of Chemical and Environmental Engineering, Shanghai University, Shanghai 200433, China
| | - Luqi Niu
- Department of Chemical and Environmental Engineering, Shanghai University, Shanghai 200433, China
| | - Lin Yu
- School of Medicine, Shanghai University, Shanghai 200433, China
| | - Kai Jin
- Department of Chemical and Environmental Engineering, Shanghai University, Shanghai 200433, China
| | - Jing Zhang
- Department of Chemical and Environmental Engineering, Shanghai University, Shanghai 200433, China
| | - Jinliang Liu
- Department of Chemical and Environmental Engineering, Shanghai University, Shanghai 200433, China
| | - Xiaohui Zhu
- Department of Chemical and Environmental Engineering, Shanghai University, Shanghai 200433, China
| | - Yihan Wu
- Department of Chemical and Environmental Engineering, Shanghai University, Shanghai 200433, China.
| | - Yong Zhang
- Department of Biomedical Engineering, National University of Singapore, 119077, Singapore; National University of Singapore Research Institute, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
13
|
Yun WS, Kim J, Lim DK, Kim DH, Jeon SI, Kim K. Recent Studies and Progress in the Intratumoral Administration of Nano-Sized Drug Delivery Systems. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2225. [PMID: 37570543 PMCID: PMC10421122 DOI: 10.3390/nano13152225] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/23/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023]
Abstract
Over the last 30 years, diverse types of nano-sized drug delivery systems (nanoDDSs) have been intensively explored for cancer therapy, exploiting their passive tumor targetability with an enhanced permeability and retention effect. However, their systemic administration has aroused some unavoidable complications, including insufficient tumor-targeting efficiency, side effects due to their undesirable biodistribution, and carrier-associated toxicity. In this review, the recent studies and advancements in intratumoral nanoDDS administration are generally summarized. After identifying the factors to be considered to enhance the therapeutic efficacy of intratumoral nanoDDS administration, the experimental results on the application of intratumoral nanoDDS administration to various types of cancer therapies are discussed. Subsequently, the reports on clinical studies of intratumoral nanoDDS administration are addressed in short. Intratumoral nanoDDS administration is proven with its versatility to enhance the tumor-specific accumulation and retention of therapeutic agents for various therapeutic modalities. Specifically, it can improve the efficacy of therapeutic agents with poor bioavailability by increasing their intratumoral concentration, while minimizing the side effect of highly toxic agents by restricting their delivery to normal tissues. Intratumoral administration of nanoDDS is considered to expand its application area due to its potent ability to improve therapeutic effects and relieve the systemic toxicities of nanoDDSs.
Collapse
Affiliation(s)
- Wan Su Yun
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Jeongrae Kim
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Dong-Kwon Lim
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Dong-Hwee Kim
- Korea Institute of Science and Technology (KU-KIST), Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Seong Ik Jeon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Kwangmeyung Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
14
|
Banerjee M, Devi Rajeswari V. A novel cross-communication of HIF-1α and HIF-2α with Wnt signaling in TNBC and influence of hypoxic microenvironment in the formation of an organ-on-chip model of breast cancer. Med Oncol 2023; 40:245. [PMID: 37454033 DOI: 10.1007/s12032-023-02112-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 07/01/2023] [Indexed: 07/18/2023]
Abstract
The microenvironment role is very important in cancer development. The epithelial-mesenchymal transition of the cancer cells depends upon specific signaling and microenvironmental conditions, such as hypoxic conditions. The crosstalk between hypoxia and Wnt signaling through some molecular mechanism in TNBC is related. Cross-communication between hypoxia and Wnt signaling in cancer cells is known, but the detailed mechanism in TNBC is unknown. This review includes the role of the hypoxia microenvironment in TNBC and the novel crosstalk of the Wnt signaling and hypoxia. When targeted, the new pathway and crosstalk link may be a solution for metastatic TNBC and chemoresistance. The microenvironment influences cancer's metastasis, which changes from person to person. Therefore, organ-on-a-chip is a very novel model to test the drugs clinically before going for human trials, focusing on personalized medications can be done. The effect of the hypoxia microenvironment on breast cancer stem cells is still unknown. Apart from all the published papers, this paper mainly focuses only on the hypoxic microenvironment and its association with the growth of TNBC. The medicines or small proteins, drugs, mimics, and inhibitors targeting wnt and hypoxia genes are consolidated in this review paper.
Collapse
Affiliation(s)
- Manosi Banerjee
- Department of Biomedical Science, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India
| | - V Devi Rajeswari
- Department of Biomedical Science, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
15
|
Kansız S, Elçin YM. Advanced liposome and polymersome-based drug delivery systems: Considerations for physicochemical properties, targeting strategies and stimuli-sensitive approaches. Adv Colloid Interface Sci 2023; 317:102930. [PMID: 37290380 DOI: 10.1016/j.cis.2023.102930] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 06/10/2023]
Abstract
Liposomes and polymersomes are colloidal vesicles that are self-assembled from lipids and amphiphilic polymers, respectively. Because of their ability to encapsulate both hydrophilic and hydrophobic therapeutics, they are of great interest in drug delivery research. Today, the applications of liposomes and polymersomes have expanded to a wide variety of complex therapeutic molecules, including nucleic acids, proteins and enzymes. Thanks to their chemical versatility, they can be tailored to different drug delivery applications to achieve maximum therapeutic index. This review article evaluates liposomes and polymersomes from a perspective that takes into account the physical and biological barriers that reduce the efficiency of the drug delivery process. In this context, the design approaches of liposomes and polymersomes are discussed with representative examples in terms of their physicochemical properties (size, shape, charge, mechanical), targeting strategies (passive and active) and response to different stimuli (pH, redox, enzyme, temperature, light, magnetic field, ultrasound). Finally, the challenges limiting the transition from laboratory to practice, recent clinical developments, and future perspectives are addressed.
Collapse
Affiliation(s)
- Seyithan Kansız
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, Department of Chemistry, Ankara, Turkey
| | - Yaşar Murat Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, Department of Chemistry, Ankara, Turkey; Biovalda Health Technologies, Inc., Ankara, Turkey.
| |
Collapse
|
16
|
Law SSY, Miyamoto T, Numata K. Organelle-targeted gene delivery in plants by nanomaterials. Chem Commun (Camb) 2023. [PMID: 37183975 DOI: 10.1039/d3cc00962a] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Genetic engineering of plants has revolutionized agriculture and has had a significant impact on our everyday life. It has allowed for the production of crops with longer shelf lives, enhanced yields and resistance to pests and disease. The application of nanomaterials in plant genetic engineering has further augmented these programs with higher delivery efficiencies, biocompatibility and the potential for plant regeneration. In particular, subcellular targeting using nanomaterials has recently become possible with the cutting-edge developments within nanomaterials, but remains challenging despite the promise in organellar engineering for the introduction of useful traits and the elucidation of subcellular interactions. This feature article provides an overview of nanomaterial delivery within plants and highlights the application of recent progress in nanomaterials for subcellular organelle-targeted delivery.
Collapse
Affiliation(s)
- Simon Sau Yin Law
- Biomacromolecules Research Team, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan.
| | - Takaaki Miyamoto
- Biomacromolecules Research Team, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan.
| | - Keiji Numata
- Biomacromolecules Research Team, RIKEN Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan.
- Department of Material Chemistry, Kyoto University, Nishikyo-ku, Kyoto 615-8510, Japan
| |
Collapse
|
17
|
Yang J, Xu Y, Fu Z, Chen J, Fan W, Wu X. Progress in research and development of temozolomide brain-targeted preparations: a review. J Drug Target 2023; 31:119-133. [PMID: 36039767 DOI: 10.1080/1061186x.2022.2119243] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gliomas are a heterogeneous group of brain tumours with high malignancy, for which surgical resection remains the mainstay of treatment at present. However, the overall prognosis of gliomas remains poor because of their aggressiveness and high recurrence. Temozolomide (TMZ) has anti-proliferative and cytotoxic effects and is indicated for glioblastoma multiforme and recurrent mesenchymal astrocytoma. However, TMZ is disadvantaged by low efficacy and drug resistance, and therefore it is necessary to enhance the brain drug concentration of TMZ to improve its effectiveness and reduce the toxic and adverse effects from systemic administration. There have been many nano-formulations developed for the delivery of TMZ to gliomas that overcome the limitations of TMZ penetration to tumours and increase brain targeting. In this paper, we review the research progress of TMZ nano-formulations, and also discuss challenges and opportunities in the research and development of drug delivery systems, hoping that the data and information summarised herein could provide assistance for the clinical treatment of gliomas.
Collapse
Affiliation(s)
- Jiefen Yang
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Youfa Xu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Department of Pharmacy, Shanghai Wei Er Biopharmaceutical Technology Co., Ltd, Shanghai, China
| | - Zhiqin Fu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Department of Pharmacy, Shanghai Wei Er Biopharmaceutical Technology Co., Ltd, Shanghai, China
| | - Jianming Chen
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Wei Fan
- Department of Pharmacy, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Wu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Department of Pharmacy, Shanghai Wei Er Biopharmaceutical Technology Co., Ltd, Shanghai, China
| |
Collapse
|
18
|
Khan RU, Shao J, Liao JY, Qian L. pH-triggered cancer-targeting polymers: From extracellular accumulation to intracellular release. NANO RESEARCH 2023; 16:5155-5168. [PMID: 36618069 PMCID: PMC9807988 DOI: 10.1007/s12274-022-5252-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/23/2022] [Accepted: 10/27/2022] [Indexed: 05/25/2023]
Abstract
Stimuli-responsive polymers are promising to achieve targeted delivery, improved stability during circulation, and controlled release of therapeutic and diagnostic agents. Among them, pH-responsive polymeric nanocarriers have attracted significant attention as pH varies in different body fluids (e.g., stomach, intestine, and colon) and intracellular organelles (e.g., endosome, lysosome, and mitochondria) to maintain homeostasis, while distinctive pH changes are also found in certain pathological states. For example, the extracellular environment of the tumor is acidic, which can be employed to drive selective delivery. During the internalization process, since most nanocarriers enter cells upon endocytosis where a drop of pH from 6.5 to 5.0 can occur from endosome to lysosome, pH-sensitive groups have been developed for enhanced cargo release. In this review, both non-covalent and covalent interactions responsive to pH changes are introduced, with a focus on the structure-property relationship and their applications in cancer targeting and endosomal escape.
Collapse
Affiliation(s)
- Rizwan Ullah Khan
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310058 China
| | - Jinning Shao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310058 China
| | - Jia-Yu Liao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310058 China
| | - Linghui Qian
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310058 China
| |
Collapse
|
19
|
Wan Z, Huang H, West RE, Zhang M, Zhang B, Cai X, Zhang Z, Luo Z, Chen Y, Zhang Y, Xie W, Yang D, Nolin TD, Wang J, Li S, Sun J. Overcoming pancreatic cancer immune resistance by codelivery of CCR2 antagonist using a STING-activating gemcitabine-based nanocarrier. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2023; 62:33-50. [PMID: 38239407 PMCID: PMC10795849 DOI: 10.1016/j.mattod.2022.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2024]
Abstract
STING agonist has recently gained much attention for cancer treatment, but the therapeutic potential of STING agonist is hampered by STING-associated tumor immune resistance. In this work, guided by both bioinformatics and computer modeling, we rationally designed a "one stone hits two birds" nanoparticle-based strategy to simultaneously activate STING innate immune response while eliminating STING-associated immune resistance for the treatment of pancreatic ductal adenocarcinoma (PDAC). We discovered that the ultra-small sized micellar system based on gemcitabine-conjugated polymer (PGEM), which showed superior capacity of penetration in pancreatic tumor spheroid model and orthotopic tumor model, could serve as a novel "STING agonist". The activation of STING signaling in dendritic cells (DCs) by PGEM increased both innate nature killer (NK) and adaptive anti-tumor T cell response. However, activation of STING signaling by PGEM in tumor cells also drove the induction of chemokines CCL2 and CCL7, resulting in immune resistance by recruiting tumor associated macrophage (TAM) and myeloid-derived suppressor cells (MDSCs). Through the combination of computer modeling and experimental screening, we developed a dual delivery modality by incorporating a CCR2 (the receptor shared by both CCL2 and CCL7) antagonist PF-6309 (PF) into PGEM micellar system. Our studies demonstrated that PGEM/PF formulation significantly reduced pancreatic tumor burden and induced potent anti-tumor immunity through reversing the CCL2/CCL7-mediated immunosuppression. Moreover, PGEM/PF sensitized PDAC tumors to anti-PD-1 therapy, leading to complete suppression/eradication of the tumors. Our work has shed light to the multi-faceted role of STING activation and provided a novel immunotherapy regimen to maximize the benefit of STING activation for PDAC treatment. In addition, this work paved a new way for bioinformatics and computer modeling-guided rational design of nanomedicine.
Collapse
Affiliation(s)
- Zhuoya Wan
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Haozhe Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Raymond E West
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Min Zhang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Bei Zhang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Xinran Cai
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Ziqian Zhang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Zhangyi Luo
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Yuang Chen
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Yue Zhang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Wen Xie
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Da Yang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Thomas D Nolin
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Junmei Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Jingjing Sun
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| |
Collapse
|
20
|
Allahyari E, Velaei K, Sanaat Z, Jalilzadeh N, Mehdizadeh A, Rahmati M. RNA interference: Promising approach for breast cancer diagnosis and treatment. Cell Biol Int 2022; 47:833-847. [PMID: 36571107 DOI: 10.1002/cbin.11979] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/15/2022] [Accepted: 12/11/2022] [Indexed: 12/27/2022]
Abstract
Today, cancer is one of the main health-related challenges, and in the meantime, breast cancer (BC) is one of the most common cancers among women, with an alarming number of incidences and deaths every year. For this reason, the discovery of novel and more effective approaches for the diagnosis, treatment, and monitoring of the disease are very important. In this regard, scientists are looking for diagnostic molecules to achieve the above-mentioned goals with higher accuracy and specificity. RNA interference (RNAi) is a posttranslational regulatory process mediated by microRNA intervention and small interfering RNAs. After transcription and edition, these two noncoding RNAs are integrated and activated with the RNA-induced silencing complex (RISC) and AGO2 to connect the target mRNA by their complementary sequence and suppress their translation, thus reducing the expression of their target genes. These two RNAi categories show different patterns in different BC types and stages compared to healthy cells, and hence, these molecules have high diagnostic, monitoring, and therapeutic potentials. This article aims to review the RNAi pathway and diagnostic and therapeutic potentials with a special focus on BC.
Collapse
Affiliation(s)
- Elham Allahyari
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kobra Velaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical, Sciences, Tabriz, Iran
| | - Zohreh Sanaat
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Jalilzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Rahmati
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
Armenia I, Cuestas Ayllón C, Torres Herrero B, Bussolari F, Alfranca G, Grazú V, Martínez de la Fuente J. Photonic and magnetic materials for on-demand local drug delivery. Adv Drug Deliv Rev 2022; 191:114584. [PMID: 36273514 DOI: 10.1016/j.addr.2022.114584] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/26/2022] [Accepted: 10/16/2022] [Indexed: 02/06/2023]
Abstract
Nanomedicine has been considered a promising tool for biomedical research and clinical practice in the 21st century because of the great impact nanomaterials could have on human health. The generation of new smart nanomaterials, which enable time- and space-controlled drug delivery, improve the limitations of conventional treatments, such as non-specific targeting, poor biodistribution and permeability. These smart nanomaterials can respond to internal biological stimuli (pH, enzyme expression and redox potential) and/or external stimuli (such as temperature, ultrasound, magnetic field and light) to further the precision of therapies. To this end, photonic and magnetic nanoparticles, such as gold, silver and iron oxide, have been used to increase sensitivity and responsiveness to external stimuli. In this review, we aim to report the main and most recent systems that involve photonic or magnetic nanomaterials for external stimulus-responsive drug release. The uniqueness of this review lies in highlighting the versatility of integrating these materials within different carriers. This leads to enhanced performance in terms of in vitro and in vivo efficacy, stability and toxicity. We also point out the current regulatory challenges for the translation of these systems from the bench to the bedside, as well as the yet unresolved matter regarding the standardization of these materials.
Collapse
Affiliation(s)
- Ilaria Armenia
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain.
| | - Carlos Cuestas Ayllón
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain
| | - Beatriz Torres Herrero
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain
| | - Francesca Bussolari
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain
| | - Gabriel Alfranca
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain
| | - Valeria Grazú
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain; Centro de Investigación Biomédica em Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| | - Jesús Martínez de la Fuente
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain; Centro de Investigación Biomédica em Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| |
Collapse
|
22
|
Li X, Qi H, Cui W, Wang Z, Fu X, Li T, Ma H, Yang Y, Yu T. Recent advances in targeted delivery of non-coding RNA-based therapeutics for atherosclerosis. Mol Ther 2022; 30:3118-3132. [PMID: 35918894 PMCID: PMC9552813 DOI: 10.1016/j.ymthe.2022.07.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 10/16/2022] Open
Abstract
Cardiovascular disease (CVD) has overtaken infectious illnesses as the leading cause of mortality and disability worldwide. The pathology that underpins CVD is atherosclerosis, characterized by chronic inflammation caused by the accumulation of plaques in the arteries. As our knowledge about the microenvironment of blood vessel walls deepens, there is an opportunity to fine-tune treatments to target the mechanisms driving atherosclerosis more directly. The application of non-coding RNAs (ncRNAs) as biomarkers or intervention targets is increasing. Although these ncRNAs play an important role in driving atherosclerosis and vascular dysfunction, the cellular and extracellular environments pose a challenge for targeted transmission and therapeutic regulation of ncRNAs. Specificity, delivery, and tolerance have hampered the clinical translation of ncRNA-based therapeutics. Nanomedicine is an emerging field that uses nanotechnology for targeted drug delivery and advanced imaging. Recently, nanoscale carriers have shown promising results and have introduced new possibilities for nucleic acid targeted drug delivery, particularly for atherosclerosis. In this review, we discuss the latest developments in nanoparticles to aid ncRNA-based drug development, particularly miRNA, and we analyze the current challenges in ncRNA targeted delivery. In particular, we highlight the emergence of various kinds of nanotherapeutic approaches based on ncRNAs, which can improve treatment options for atherosclerosis.
Collapse
Affiliation(s)
- Xiaoxin Li
- Center for Regenerative Medicine, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People's Republic of China
| | - Hongzhao Qi
- Center for Regenerative Medicine, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People's Republic of China
| | - Weigang Cui
- Department of Cardiology, People's Hospital of Rizhao, No. 126 Taian Road, Rizhao 276827, People's Republic of China
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266000, China
| | - Xiuxiu Fu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266000, China
| | - Tianxiang Li
- Center for Regenerative Medicine, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People's Republic of China
| | - Huibo Ma
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanyan Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao 266021, People's Republic of China.
| | - Tao Yu
- Center for Regenerative Medicine, Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People's Republic of China; Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266000, China.
| |
Collapse
|
23
|
Zhou Q, Li J, Xiang J, Shao S, Zhou Z, Tang J, Shen Y. Transcytosis-enabled active extravasation of tumor nanomedicine. Adv Drug Deliv Rev 2022; 189:114480. [PMID: 35952830 DOI: 10.1016/j.addr.2022.114480] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/11/2022] [Accepted: 08/01/2022] [Indexed: 01/24/2023]
Abstract
Extravasation is the first step for nanomedicines in circulation to reach targeted solid tumors. Traditional nanomedicines have been designed to extravasate into tumor interstitium through the interendothelial gaps previously assumed rich in tumor blood vessels, i.e., the enhanced permeability and retention (EPR) effect. While the EPR effect has been validated in animal xenograft tumor models, accumulating evidence implies that the EPR effect is very limited and highly heterogeneous in human tumors, leading to highly unpredictable and inefficient extravasation and thus limited therapeutic efficacy of nanomedicines, including those approved in clinics. Enabling EPR-independent extravasation is the key to develop new generation of nanomedicine with enhanced efficacy. Transcytosis of tumor endothelial cells can confer nanomedicines to actively extravasate into solid tumors without relying on the EPR effect. Here, we review and prospectthe development of transcytosis-inducing nanomedicines, in hope of providing instructive insights for design of nanomedicines that can undergo selective transcellular transport across tumor endothelial cells, and thus inspiring the development of next-generation nanomedicines for clinical translation.
Collapse
Affiliation(s)
- Quan Zhou
- Key Laboratory of Smart Biomaterials of Zhejiang Province and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Junjun Li
- Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Key Laboratory of Smart Biomaterials of Zhejiang Province and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311215, China
| | - Shiqun Shao
- Key Laboratory of Smart Biomaterials of Zhejiang Province and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311215, China
| | - Zhuxian Zhou
- Key Laboratory of Smart Biomaterials of Zhejiang Province and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jianbin Tang
- Key Laboratory of Smart Biomaterials of Zhejiang Province and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311215, China.
| | - Youqing Shen
- Key Laboratory of Smart Biomaterials of Zhejiang Province and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
24
|
Embelin and Its Derivatives: Design, Synthesis, and Potential Delivery Systems for Cancer Therapy. Pharmaceuticals (Basel) 2022; 15:ph15091131. [PMID: 36145352 PMCID: PMC9505931 DOI: 10.3390/ph15091131] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Embelin is a naturally occurring benzoquinone that inhibits the growth of cancer cells, making it a potent anticancer drug. However, the low water solubility of embelin restricts its clinical applicability. This review provides a concise summary and in-depth analysis of the published literature on the design and synthesis of embelin derivatives possessing increased aqueous solubility and superior therapeutic efficacy. In addition, the potential of drug delivery systems to improve the anticancer capabilities of embelin and its derivatives is discussed.
Collapse
|
25
|
Flores de los Rios PA, Casañas Pimentel RG, San Martín Martínez E. Nanodrugs against cancer: biological considerations in its redesign. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2097680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- P. A. Flores de los Rios
- Instituto Politécnico Nacional, Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada Legaria 694, Irrigación, Ciudad de México, México
| | - R. G. Casañas Pimentel
- Instituto Politécnico Nacional, Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada Legaria 694, Irrigación, Ciudad de México, México
| | - E. San Martín Martínez
- Instituto Politécnico Nacional, Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada Legaria 694, Irrigación, Ciudad de México, México
| |
Collapse
|
26
|
Xie H, Li W, Liu H, Chen Y, Ma M, Wang Y, Luo Y, Song D, Hou Q, Lu W, Bai Y, Li B, Ma J, Huang C, Yang T, Liu Z, Zhao X, Ding P. Erythrocyte Membrane-Coated Invisible Acoustic-Sensitive Nanoparticle for Inducing Tumor Thrombotic Infarction by Precisely Damaging Tumor Vascular Endothelium. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201933. [PMID: 35789094 DOI: 10.1002/smll.202201933] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/16/2022] [Indexed: 06/15/2023]
Abstract
Selective induction of tumor thrombus infarction is a promising antitumor strategy. Non-persistent embolism due to non-compacted thrombus and activated fibrinolytic system within the tumor large blood vessels and tumor margin recurrence are the main therapeutic bottlenecks. Herein, an erythrocyte membrane-coated invisible acoustic-sensitive nanoparticle (TXA+DOX/PFH/RBCM@cRGD) is described, which can induce tumor thrombus infarction by precisely damaging tumor vascular endothelium. It is revealed that TXA+DOX/PFH/RBCM@cRGD can effectively accumulate on the endothelial surface of tumor vessels with the help of the red blood cell membrane (RBCM) stealth coating and RGD cyclic peptide (cRGD), which can be delivered in a targeted manner as nanoparticle missiles. As a kind of phase-change material, perfluorohexane (PFH) nanodroplets possess excellent acoustic responsiveness. Acoustic-sensitive missiles can undergo an acoustic phase transition and intense cavitation with response to low-intensity focused ultrasound (LIFU), damaging the tumor vascular endothelium, rapidly initiating the coagulation cascade, and forming thromboembolism in the tumor vessels. The drugs loaded in the inner water phase are released explosively. Tranexamic acid (TXA) inhibits the fibrinolytic system, and doxorubicin (DOX) eliminates the margin survival. In summary, a stealthy and acoustically responsive multifunctional nanoparticle delivery platform is successfully developed for inducing thrombus infarction by precisely damaging tumor vascular endothelium.
Collapse
Affiliation(s)
- Huichao Xie
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Wan Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hui Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yongfeng Chen
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Mengrui Ma
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yichen Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yucen Luo
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Di Song
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Qianqian Hou
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Wenwen Lu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yu Bai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Bao Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jizhuang Ma
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Chi Huang
- Ultrasound Department of Shengjing Hospital, China Medical University, Shenyang, 110016, China
| | - Tianzhi Yang
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, ME, 04401, USA
| | - Zhining Liu
- Ultrasound Department, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Xiaoyun Zhao
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Pingtian Ding
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| |
Collapse
|
27
|
Wang K, Yan S, Han T, Wu Q, Yan N, Kang M, Ge J, Wang D, Tang BZ. Cascade C-H-Activated Polyannulations toward Ring-Fused Heteroaromatic Polymers for Intracellular pH Mapping and Cancer Cell Killing. J Am Chem Soc 2022; 144:11788-11801. [PMID: 35736562 DOI: 10.1021/jacs.2c04032] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The development of straightforward and efficient synthetic methods toward ring-fused heteroaromatic polymers with attractive functionalities has great significance in both chemistry and materials science. Herein, we develop a facile cascade C-H-activated polyannulation route that can in situ generate multiple ring-fused aza-heteroaromatic polymers from readily available monomers in an atom-economical manner. A series of complex polybenzimidazole derivatives with high absolute molecular weights of up to 24 000 are efficiently produced in high yields within 2 h. Benefiting from their unique imidazole-containing ring-fused structures with multiple aryl pendants, the obtained polymers show excellent thermal and morphological stability, good solution processability, high refractive index, small chromic dispersion, as well as remarkable acid-base-responsive fluorescence. Taking advantage of the ratiometric fluorescence response of the triphenylamine-substituted heteroaromatic polymer to pH variations, we successfully apply it as a sensitive fluorescence probe for the mapping and quantitative analysis of intracellular pH in live cells. Furthermore, through the simple N-methylation reaction of the ring-fused polybenzimidazoles, diverse azonia-containing polyelectrolytes are readily produced, which can efficiently kill cancer cells via the synergistic effects of dark toxicity and phototoxicity.
Collapse
Affiliation(s)
- Kang Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China.,College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Saisai Yan
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China.,College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Ting Han
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Qian Wu
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China.,Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong 999077, China
| | - Neng Yan
- Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong 999077, China
| | - Miaomiao Kang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jinyin Ge
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China.,College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Ben Zhong Tang
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong 999077, China.,Shenzhen Institute of Aggregate Science and Technology, School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| |
Collapse
|
28
|
Vascular bursts-mediated tumor accumulation and deep penetration of spherical nucleic acids for synergistic radio-immunotherapy. JOURNAL OF CONTROLLED RELEASE : OFFICIAL JOURNAL OF THE CONTROLLED RELEASE SOCIETY 2022; 348:1050-1065. [PMID: 35750133 DOI: 10.1016/j.jconrel.2022.06.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/13/2022] [Accepted: 06/17/2022] [Indexed: 11/22/2022]
Abstract
While nanomedicines have attracted great interests for tumor therapy, their targeting and intra-tumoral penetrating efficiencies have been questioned. Here, we report a two-step low-dose radiotherapy (RT) strategy to realize significant accumulation and deep penetration of spherical nucleic acids (SNAs)-based nanomedicine for synergistic radio-immunotherapy. The first step RT was employed to recruit large amounts of macrophages into tumor. The tumor infiltrated macrophages not only served as nanoparticles drug depots, but also elicited dynamic bursts extravasation to enhance nanoparticles accumulation. We optimized the spatiotemporal combination of RT and SNAs administration for higher level of SNAs delivery, and the delivered SNAs promote M2-to-M1 phenotype switch of macrophages to increase phagocytosis of nanoparticles by 6-fold, resulting in positive feedback with even higher accumulation and intra-tumor penetration of SNAs. Through vascular bursts and macrophage repolarization, as high as 25-fold enhancement of nanoparticles accumulation was achieved as compared to passive targeting of nanoparticles, and the nanoparticles were eventually distributed throughout the tumor tissue with efficient deep penetration. Finally, SNAs in tumor simultaneously sensitized the second dose of RT and remodeled tumor immune microenvironment, resulting in a synergistic anticancer therapy in combination of anti-PD-L1 antibody (αPD-L1) with no noticeable side effects caused by either RT or αPD-L1.
Collapse
|
29
|
Delvaux NA, Mathew B, Rice KG. Fluorescent labeling of plasmid DNA for gene delivery: Implications of dye hydrophobicity on labeling efficiencies and nanoparticle size. Anal Biochem 2022; 644:113895. [PMID: 32783899 PMCID: PMC7870724 DOI: 10.1016/j.ab.2020.113895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/27/2020] [Indexed: 11/16/2022]
Abstract
Covalent fluorescent labels are important tools for monitoring the in vitro and in vivo localization of plasmid DNA nanoparticles, but must meet several criteria including high DNA labeling efficiencies and minimal impact on nanoparticle size. We developed a novel fluorescent labeling strategy utilizing an aryl azide photolabel conjugated to a short cationic peptide to label plasmid DNA with Cyanine 5 and sulfo-Cyanine 5. Using a simple camera flash apparatus, photolabel-peptide-dyes can be conjugated to DNA in minutes with preservation of DNA structure and minimal dye photobleaching. The addition of two anionic sulfonates to the Cyanine 5 core greatly improved labeling efficiencies from ~13 to ~53% and mitigated PEGylated polyacridine peptide-DNA nanoparticle size increases over a range of labeling densities. Comparison of our sulfo-Cyanine 5 peptide label to the Mirus Bio Label IT-Cy5 kit revealed that while both did not affect nanoparticle sizes appreciably, labeling efficiencies with our conjugate were higher, possibly due to the higher positive charge density on the peptide linker. The results from this work provide important considerations for choosing fluorophore tags to track DNA nanoparticles.
Collapse
Affiliation(s)
- Nathan A Delvaux
- Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, 52242, Iowa City, IA, USA
| | - Basil Mathew
- Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, 52242, Iowa City, IA, USA
| | - Kevin G Rice
- Division of Medicinal and Natural Products Chemistry, College of Pharmacy, University of Iowa, 52242, Iowa City, IA, USA.
| |
Collapse
|
30
|
Chu S, Shi X, Tian Y, Gao F. pH-Responsive Polymer Nanomaterials for Tumor Therapy. Front Oncol 2022; 12:855019. [PMID: 35392227 PMCID: PMC8980858 DOI: 10.3389/fonc.2022.855019] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/24/2022] [Indexed: 12/24/2022] Open
Abstract
The complexity of the tumor microenvironment presents significant challenges to cancer therapy, while providing opportunities for targeted drug delivery. Using characteristic signals of the tumor microenvironment, various stimuli-responsive drug delivery systems can be constructed for targeted drug delivery to tumor sites. Among these, the pH is frequently utilized, owing to the pH of the tumor microenvironment being lower than that of blood and healthy tissues. pH-responsive polymer carriers can improve the efficiency of drug delivery in vivo, allow targeted drug delivery, and reduce adverse drug reactions, enabling multifunctional and personalized treatment. pH-responsive polymers have gained increasing interest due to their advantageous properties and potential for applicability in tumor therapy. In this review, recent advances in, and common applications of, pH-responsive polymer nanomaterials for drug delivery in cancer therapy are summarized, with a focus on the different types of pH-responsive polymers. Moreover, the challenges and future applications in this field are prospected.
Collapse
Affiliation(s)
- Shunli Chu
- Department of Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xiaolu Shi
- Department of Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Ye Tian
- Department of Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Fengxiang Gao
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| |
Collapse
|
31
|
Zhao Y, Shi D, Shang M, Sun X, Guo L, Meng D, Liu X, Zhou X, Li J. GRP78-targeted and doxorubicin-loaded nanodroplets combined with ultrasound: a potential novel theranostics for castration-resistant prostate cancer. Drug Deliv 2022; 29:203-213. [PMID: 34985396 PMCID: PMC8741251 DOI: 10.1080/10717544.2021.2023698] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The construction of multifunctional oncotherapy nanoplatforms that combine diagnosis and treatment remains challenging. Nanodroplets (NDs), which simultaneously enhance ultrasound imaging and therapeutic effects, are a potential strategy for non-invasive drug delivery. To achieve the goals of precise medicine, novel SP94 peptide-modified and doxorubicin-loaded ultrasonic NDs (SP94-DOX-NDs) for castration-resistant prostate cancer (CRPC) targeting and treatment were constructed in this study. The characteristics, contrast-enhanced ultrasound imaging (CEUI), targeting ability to glucose-regulated protein 78 (GRP78)-overexpressing CRPC and anticancer effect of the SP94-DOX-NDs were assessed. The desired SP94-NDs were successfully prepared using the nanoemulsification method using a certain proportion of SP94-PEG-chitosan, perfluoropentane (PFP), Tween 20, and lecithin. SP94-NDs with a size of ∼300 nm showed great biocompatibility and CEUI ability. Compared with blank NDs, SP94-NDs exhibited higher tumor-specific targeting ability due to conjugation between the SP94 peptide and GRP78-overexpressing 22RV1 cells. Most importantly, in vitro and in vivo investigations showed that SP94-DOX-NDs combined with ultrasound could specifically deliver DOX into 22RV1 cells and thereby demonstrated a stronger anticancer effect than DOX-NDs and DOX. Thus, SP94-DOX-NDs may provide an efficient approach for the real-time imaging of tumors and triggered, accurate drug delivery to tumors.
Collapse
Affiliation(s)
- Yading Zhao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, China
| | - Dandan Shi
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, China
| | - Mengmeng Shang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao Sun
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, China
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, China
| | - Dong Meng
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, China
| | - Xinxin Liu
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoying Zhou
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, China
| | - Jie Li
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
32
|
Yadav N, Dahiya T, Chhillar AK, Rana JS, Mohan H. Promising Applications of Nanotechnology in Cancer Diagnostics and Therapeutics. Curr Pharm Biotechnol 2021; 23:1556-1568. [PMID: 34951360 DOI: 10.2174/1389201023666211222165508] [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: 08/25/2021] [Revised: 11/12/2021] [Accepted: 11/19/2021] [Indexed: 11/22/2022]
Abstract
Cancer is characterized by the accumulation of genetic mutations in cells by different types of mutagens such as physical, chemical, and biological. Consequently, normal cell cycles get interrupted. Conventional techniques used for diagnosis include. Various conventional techniques used for cancer diagnosis include immunological assays, histopathogical tests, polymerase chain reaction, computed tomography, magnetic resonance, radiation therapy, and many more. These techniques are expensive, time consuming, tedious, adverse effects to healthy cells and requirement of skilled personnel for their operation. Therefore nanomaterials based biosensors have been used for the sensitive, selective, economic and quick detection of cancer biomarkers. Electrochemical biosensors have shown profound impact in efficient diagnosis of cancers that facilitate the effective treatment of patient in acute stage. Nanomaterials including inorganic, organic and polymeric nanomaterials have been used in the treatment of different types of cancers. Nanoapproaches have offered several merits including site-specific, require traces amount of therapeutic molecules, limited toxicity, avoid drug resistance, more efficient, sensitive and reliable than conventional chemotherapeutics and radiation therapies. Therefore, future research should be focussed on development of highly inventive nanotools for the diagnosis and therapeutics of cancers.
Collapse
Affiliation(s)
- Neelam Yadav
- Department of Biotechnology, Deenbandhu Chhotu Ram University of Science and Technology, -131039, Haryana. India
| | - Twinkle Dahiya
- Department of Biotechnology, Deenbandhu Chhotu Ram University of Science and Technology, -131039, Haryana. India
| | - Anil Kumar Chhillar
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| | - Jogender Singh Rana
- Department of Biotechnology, Deenbandhu Chhotu Ram University of Science and Technology, -131039, Haryana. India
| | - Hari Mohan
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| |
Collapse
|
33
|
Roy SM, Garg V, Barman S, Ghosh C, Maity AR, Ghosh SK. Kinetics of Nanomedicine in Tumor Spheroid as an In Vitro Model System for Efficient Tumor-Targeted Drug Delivery With Insights From Mathematical Models. Front Bioeng Biotechnol 2021; 9:785937. [PMID: 34926430 PMCID: PMC8671936 DOI: 10.3389/fbioe.2021.785937] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/27/2021] [Indexed: 12/25/2022] Open
Abstract
Numerous strategies have been developed to treat cancer conventionally. Most importantly, chemotherapy shows its huge promise as a better treatment modality over others. Nonetheless, the very complex behavior of the tumor microenvironment frequently impedes successful drug delivery to the tumor sites that further demands very urgent and effective distribution mechanisms of anticancer drugs specifically to the tumor sites. Hence, targeted drug delivery to tumor sites has become a major challenge to the scientific community for cancer therapy by assuring drug effects to selective tumor tissue and overcoming undesired toxic side effects to the normal tissues. The application of nanotechnology to the drug delivery system pays heed to the design of nanomedicine for specific cell distribution. Aiming to limit the use of traditional strategies, the adequacy of drug-loaded nanocarriers (i.e., nanomedicine) proves worthwhile. After systemic blood circulation, a typical nanomedicine follows three levels of disposition to tumor cells in order to exhibit efficient pharmacological effects induced by the drug candidates residing within it. As a result, nanomedicine propounds the assurance towards the improved bioavailability of anticancer drug candidates, increased dose responses, and enhanced targeted efficiency towards delivery and distribution of effective therapeutic concentration, limiting toxic concentration. These aspects emanate the proficiency of drug delivery mechanisms. Understanding the potential tumor targeting barriers and limiting conditions for nanomedicine extravasation, tumor penetration, and final accumulation of the anticancer drug to tumor mass, experiments with in vivo animal models for nanomedicine screening are a key step before it reaches clinical translation. Although the study with animals is undoubtedly valuable, it has many associated ethical issues. Moreover, individual experiments are very expensive and take a longer time to conclude. To overcome these issues, nowadays, multicellular tumor spheroids are considered a promising in vitro model system that proposes better replication of in vivo tumor properties for the future development of new therapeutics. In this review, we will discuss how tumor spheroids could be used as an in vitro model system to screen nanomedicine used in targeted drug delivery, aiming for better therapeutic benefits. In addition, the recent proliferation of mathematical modeling approaches gives profound insight into the underlying physical principles and produces quantitative predictions. The hierarchical tumor structure is already well decorous to be treated mathematically. To study targeted drug delivery, mathematical modeling of tumor architecture, its growth, and the concentration gradient of oxygen are the points of prime focus. Not only are the quantitative models circumscribed to the spheroid, but also the role of modeling for the nanoparticle is equally inevitable. Abundant mathematical models have been set in motion for more elaborative and meticulous designing of nanomedicine, addressing the question regarding the objective of nanoparticle delivery to increase the concentration and the augmentative exposure of the therapeutic drug molecule to the core. Thus, to diffuse the dichotomy among the chemistry involved, biological data, and the underlying physics, the mathematical models play an indispensable role in assisting the experimentalist with further evaluation by providing the admissible quantitative approach that can be validated. This review will provide an overview of the targeted drug delivery mechanism for spheroid, using nanomedicine as an advantageous tool.
Collapse
Affiliation(s)
| | - Vrinda Garg
- Department of Physics, National Institute of Technology, Warangal, India
| | - Sourav Barman
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| | - Chitrita Ghosh
- Department of Pharmacology, Burdwan Medical College and Hospital, Burdwan, India
| | | | - Surya K. Ghosh
- Department of Physics, National Institute of Technology, Warangal, India
| |
Collapse
|
34
|
Design and Optimization of the Circulatory Cell-Driven Drug Delivery Platform. Stem Cells Int 2021; 2021:8502021. [PMID: 34603454 PMCID: PMC8481068 DOI: 10.1155/2021/8502021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/17/2021] [Indexed: 01/14/2023] Open
Abstract
Achievement of high targeting efficiency for a drug delivery system remains a challenge of tumor diagnoses and nonsurgery therapies. Although nanoparticle-based drug delivery systems have made great progress in extending circulation time, improving durability, and controlling drug release, the targeting efficiency remains low. And the development is limited to reducing side effects since overall survival rates are mostly unchanged. Therefore, great efforts have been made to explore cell-driven drug delivery systems in the tumor area. Cells, particularly those in the blood circulatory system, meet most of the demands that the nanoparticle-based delivery systems do not. These cells possess extended circulation times and innate chemomigration ability and can activate an immune response that exerts therapeutic effects. However, new challenges have emerged, such as payloads, cell function change, cargo leakage, and in situ release. Generally, employing cells from the blood circulatory system as cargo carriers has achieved great benefits and paved the way for tumor diagnosis and therapy. This review specifically covers (a) the properties of red blood cells, monocytes, macrophages, neutrophils, natural killer cells, T lymphocytes, and mesenchymal stem cells; (b) the loading strategies to balance cargo amounts and cell function balance; (c) the cascade strategies to improve cell-driven targeting delivery efficiency; and (d) the features and applications of cell membranes, artificial cells, and extracellular vesicles in cancer treatment.
Collapse
|
35
|
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: 111] [Impact Index Per Article: 27.8] [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
|
36
|
Liu M, Wu C, Ke L, Li Z, Wu YL. Emerging Biomaterials-Based Strategies for Inhibiting Vasculature Function in Cancer Therapy. SMALL METHODS 2021; 5:e2100347. [PMID: 34927997 DOI: 10.1002/smtd.202100347] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/20/2021] [Indexed: 06/14/2023]
Abstract
The constant feeding of oxygen and nutrients through the blood vasculature has a vital role in maintaining tumor growth. Interestingly, recent endeavors have shown that nanotherapeutics with the strategy to block tumor blood vessels feeding nutrients and oxygen for starvation therapy can be helpful in cancer treatment. However, this field has not been detailed. Hence, this review will present an exhaustive summary of the existing biomaterial based strategies to disrupt tumor vascular function for effective cancer treatment, including hydrogel or nanogel-mediated local arterial embolism, thrombosis activator loaded nano-material-mediated vascular occlusion and anti-vascular drugs that block tumor vascular function, which may be beneficial to the design of anti-cancer nanomedicine by targeting the tumor vascular system.
Collapse
Affiliation(s)
- Minting Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Lingjie Ke
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Zhiguo Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
37
|
Abstract
"There's plenty of room at the bottom" (Richard Feynman, 1959): an invitation for (metalla)carboranes to enter the (new) field of nanomedicine. For two decades, the number of publications on boron cluster compounds designed for potential applications in medicine has been constantly increasing. Hundreds of compounds have been screened in vitro or in vivo for a variety of biological activities (chemotherapeutics, radiotherapeutics, antiviral, etc.), and some have shown rather promising potential for further development. However, until now, no boron cluster compounds have made it to the clinic, and even clinical trials have been very sparse. This review introduces a new perspective in the field of medicinal boron chemistry, namely that boron-based drugs should be regarded as nanomedicine platforms, due to their peculiar self-assembly behaviour in aqueous solutions, and treated as such. Examples for boron-based 12- and 11-vertex clusters and appropriate comparative studies from medicinal (in)organic chemistry and nanomedicine, highlighting similarities, differences and gaps in physicochemical and biological characterisation methods, are provided to encourage medicinal boron chemists to fill in the gaps between chemistry laboratory and real applications in living systems by employing bioanalytical and biophysical methods for characterising and controlling the aggregation behaviour of the clusters in solution.
Collapse
Affiliation(s)
- Marta Gozzi
- Institute of Inorganic ChemistryFaculty of Chemistry and MineralogyLeipzig UniversityJohannisallee 2904103LeipzigGermany
- Institute of Analytical ChemistryFaculty of Chemistry and MineralogyLeipzig UniversityLinnéstr. 304103LeipzigGermany
- Institute of Medicinal Physics and BiophysicsFaculty of MedicineLeipzig UniversityHärtelstr. 16–1804107LeipzigGermany
| | - Benedikt Schwarze
- Institute of Medicinal Physics and BiophysicsFaculty of MedicineLeipzig UniversityHärtelstr. 16–1804107LeipzigGermany
| | - Evamarie Hey‐Hawkins
- Institute of Inorganic ChemistryFaculty of Chemistry and MineralogyLeipzig UniversityJohannisallee 2904103LeipzigGermany
| |
Collapse
|
38
|
Li JX, Huang QY, Zhang JY, Du JZ. Engineering nanoparticles to tackle tumor barriers. J Mater Chem B 2021; 8:6686-6696. [PMID: 32579660 DOI: 10.1039/d0tb00967a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Engineering nanoparticles (NPs) as delivery systems of anticancer therapeutics has attracted tremendous attention in recent decades, and some nanoscale drug formulations have been approved for clinical use. However, their therapeutic efficacies are still limited by the presence of a series of biological barriers during the delivery process. Among these obstacles, tumor barriers are generally recognized as the bottleneck, because they dominate the NP extravasation from the tumor vasculature and penetration into the tumor parenchyma. Therefore, this review first discussed tumor barriers from two aspects: tumor vascular barriers and tumor stromal barriers. Pathological features of the two sets of barriers as well as their influence on the delivery efficacy were described. Then, we outlined strategies for engineering NPs to overcome these challenges: increasing extravasation through physical property optimization and tumor vascular targeting; and facilitating deep penetration through particle size manipulation, modulation of the tumor extracellular matrix, and some new mechanisms. This review will provide a critical perspective on engineering strategies for more efficient nanomedicine in oncology.
Collapse
Affiliation(s)
- Jia-Xian Li
- Guangzhou First People's Hospital, and Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| | | | | | | |
Collapse
|
39
|
Subia B, Dahiya UR, Mishra S, Ayache J, Casquillas GV, Caballero D, Reis RL, Kundu SC. Breast tumor-on-chip models: From disease modeling to personalized drug screening. J Control Release 2021; 331:103-120. [PMID: 33417986 PMCID: PMC8172385 DOI: 10.1016/j.jconrel.2020.12.057] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 02/06/2023]
Abstract
Breast cancer is one of the leading causes of mortality worldwide being the most common cancer among women. Despite the significant progress obtained during the past years in the understanding of breast cancer pathophysiology, women continue to die from it. Novel tools and technologies are needed to develop better diagnostic and therapeutic approaches, and to better understand the molecular and cellular players involved in the progression of this disease. Typical methods employed by the pharmaceutical industry and laboratories to investigate breast cancer etiology and evaluate the efficiency of new therapeutic compounds are still based on traditional tissue culture flasks and animal models, which have certain limitations. Recently, tumor-on-chip technology emerged as a new generation of in vitro disease model to investigate the physiopathology of tumors and predict the efficiency of drugs in a native-like microenvironment. These microfluidic systems reproduce the functional units and composition of human organs and tissues, and importantly, the rheological properties of the native scenario, enabling precise control over fluid flow or local gradients. Herein, we review the most recent works related to breast tumor-on-chip for disease modeling and drug screening applications. Finally, we critically discuss the future applications of this emerging technology in breast cancer therapeutics and drug development.
Collapse
Affiliation(s)
- Bano Subia
- Elvesys Microfluidics Innovation Centre, Paris 75011, France..
| | | | - Sarita Mishra
- CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India..
| | - Jessica Ayache
- Elvesys Microfluidics Innovation Centre, Paris 75011, France..
| | | | - David Caballero
- 3B's Research Group, I3Bs-Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Barco, Guimarãaes 4805-017, Portugal; ICVS/3B's - PT Government Associate Laboratory, 4805-017, Braga/Guimarães, Portugal.
| | - Rui L Reis
- 3B's Research Group, I3Bs-Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Barco, Guimarãaes 4805-017, Portugal; ICVS/3B's - PT Government Associate Laboratory, 4805-017, Braga/Guimarães, Portugal.
| | - Subhas C Kundu
- 3B's Research Group, I3Bs-Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Barco, Guimarãaes 4805-017, Portugal; ICVS/3B's - PT Government Associate Laboratory, 4805-017, Braga/Guimarães, Portugal.
| |
Collapse
|
40
|
Huang D, Sun L, Huang L, Chen Y. Nanodrug Delivery Systems Modulate Tumor Vessels to Increase the Enhanced Permeability and Retention Effect. J Pers Med 2021; 11:124. [PMID: 33672813 PMCID: PMC7917988 DOI: 10.3390/jpm11020124] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 12/24/2022] Open
Abstract
The use of nanomedicine for antitumor therapy has been extensively investigated for a long time. Enhanced permeability and retention (EPR) effect-mediated drug delivery is currently regarded as an effective way to bring drugs to tumors, especially macromolecular drugs and drug-loaded pharmaceutical nanocarriers. However, a disordered vessel network, and occluded or embolized tumor blood vessels seriously limit the EPR effect. To augment the EPR effect and improve curative effects, in this review, we focused on the perspective of tumor blood vessels, and analyzed the relationship among abnormal angiogenesis, abnormal vascular structure, irregular blood flow, extensive permeability of tumor vessels, and the EPR effect. In this commentary, nanoparticles including liposomes, micelles, and polymers extravasate through the tumor vasculature, which are based on modulating tumor vessels, to increase the EPR effect, thereby increasing their therapeutic effect.
Collapse
Affiliation(s)
- Dong Huang
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; (D.H.); (L.S.)
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Lingna Sun
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; (D.H.); (L.S.)
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA;
| | - Yanzuo Chen
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; (D.H.); (L.S.)
- Engineering Research Centre of Pharmaceutical Process Chemistry, Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
41
|
Fakhri KU, Sultan A, Mushtaque M, Hasan MR, Nafees S, Hafeez ZB, Zafaryab M, Rizwanullah M, Sharma D, Bano F, AlMalki WH, Ahmad FJ, Rizvi MMA. Obstructions in Nanoparticles Conveyance, Nano-Drug Retention, and EPR Effect in Cancer Therapies. HANDBOOK OF RESEARCH ON ADVANCEMENTS IN CANCER THERAPEUTICS 2021. [DOI: 10.4018/978-1-7998-6530-8.ch026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this chapter, the authors first review nano-devices that are mixtures of biologic molecules and synthetic polymers like nano-shells and nano-particles for the most encouraging applications for different cancer therapies. Nano-sized medications additionally spill especially into tumor tissue through penetrable tumor vessels and are then held in the tumor bed because of diminished lymphatic drainage. This procedure is known as the enhanced penetrability and retention (EPR) impact. Nonetheless, while the EPR impact is generally held to improve conveyance of nano-medications to tumors, it in certainty offers not exactly a 2-overlay increment in nano-drug conveyance contrasted with basic ordinary organs, bringing about medication concentration that is not adequate for restoring most malignant growths. In this chapter, the authors likewise review different obstructions for nano-sized medication conveyance and to make the conveyance of nano-sized medications to tumors progressively successful by expanding on the EPR impact..
Collapse
Affiliation(s)
| | | | | | | | | | | | - Md Zafaryab
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Md Rizwanullah
- School of Pharmaceutical Education and Research, Jamia Hamdard, India
| | - Deepti Sharma
- Institute of Nuclear Medicine and Allied Sciences, India
| | - Farhad Bano
- National Institute of Immunology, New Delhi, India
| | | | - Farhan Jalees Ahmad
- School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | | |
Collapse
|
42
|
Li J, Kataoka K. Chemo-physical Strategies to Advance the in Vivo Functionality of Targeted Nanomedicine: The Next Generation. J Am Chem Soc 2020; 143:538-559. [PMID: 33370092 DOI: 10.1021/jacs.0c09029] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The past few decades have witnessed an evolution of nanomedicine from biologically inert entities to more smart systems, aimed at advancing in vivo functionality. However, we should recognize that most systems still rely on reasonable explanation-including some over-explanation-rather than definitive evidence, which is a watershed radically determining the speed and extent of advancing nanomedicine. Probing nano-bio interactions and desirable functionality at the tissue, cellular, and molecular levels is most frequently overlooked. Progress toward answering these questions will provide instructive insight guiding more effective chemo-physical strategies. Thus, in the next generation, we argue that much effort should be made to provide definitive evidence for proof-of-mechanism, in lieu of creating many new and complicated systems for similar proof-of-concept.
Collapse
Affiliation(s)
- Junjie Li
- Innovation Center of NanoMedicne, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicne, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan.,Institute for Future Initiatives, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
43
|
Abstract
Bacteria possess many unique properties in treating cancer that are unachievable with standard methods, including specific tumor targeting, deep tissue penetration, and programmable therapeutic efficacy. Bacteria species such as Salmonella, Escherichia, Clostridium, and Listeria have been demonstrated to restrict tumor growth with improved prognosis in mice models. Moreover, some bacterial strains were advanced to clinical trials. This Spotlight on Applications summarizes general strategies for engineering living bacteria to fight cancer and provides examples to illustrate different approaches to engineer bacteria for safety and therapeutic index improvement.
Collapse
Affiliation(s)
- Lei Rong
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China.,Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Qi Lei
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China.,School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, People's Republic of China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China
| |
Collapse
|
44
|
Lu D, Wang J, Li Y, Zhang Y, Yu L, Xu T, Guo H, Zhang Y, Wang X, Wang X, Teng G, Lei Z. Tumor Noninvasive and Target Embolization Therapy Platform by Intravenous Injection Based on Acidic Microenvironment-Responsive Hyperbranched Poly(amino acid)s. ACS CENTRAL SCIENCE 2020; 6:1977-1986. [PMID: 33274275 PMCID: PMC7706070 DOI: 10.1021/acscentsci.0c00506] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Indexed: 05/05/2023]
Abstract
Transcatheter arterial embolization (TAE) has been widely applied in treatments of unresectable or hypervascular tumors, but the procedure of TAE is complicated possibly brings inherent risks. Here, inspired by pH-responsive drug delivery systems, a new method of noninvasive and target embolization therapy by intravenous injection was developed. This method is based on a type of acidic microenvironment-responsive hyperbranched poly(amino acid) (HPTTG) to avoid using catheterization and real-time image guidance angiography, simplifying the procedure, elevating compliance and general applicability of embolization therapy. The pH value of the sol-to-gel phase transition with decreasing pH of HPTTG was controlled by adjusting the ratio of acidic amino acids in copolymers. The results of the tumor-bearing animal experiment indicate that the HPTTG have an excellent target and embolic ability; they accumulate the most at the tumor site in 8 h postinjection. Blood vessels of the tumors were occluded, and the tumors were inhibited and necrotized in about 20 days. Therefore, it is expected that HPTTG not only can be used as novel embolic materials for efficient noninvasive embolization therapy of many solid tumors but also can be used as a multifunctional platform for combined theranostics, for example, combination with controlled release, thermal ablation, multimodal imaging, synergistic therapy, etc.
Collapse
Affiliation(s)
- Dedai Lu
- Key
Laboratory of Eco-Functional Polymer Materials of the Ministry of
Education, Key Laboratory of Eco-Environmental Polymer Materials of
Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, China
| | - Jiachen Wang
- Key
Laboratory of Eco-Functional Polymer Materials of the Ministry of
Education, Key Laboratory of Eco-Environmental Polymer Materials of
Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, China
| | - Yunfei Li
- Key
Laboratory of Eco-Functional Polymer Materials of the Ministry of
Education, Key Laboratory of Eco-Environmental Polymer Materials of
Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, China
| | - Yongyong Zhang
- Key
Laboratory of Eco-Functional Polymer Materials of the Ministry of
Education, Key Laboratory of Eco-Environmental Polymer Materials of
Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, China
| | - Lili Yu
- Key
Laboratory of Eco-Functional Polymer Materials of the Ministry of
Education, Key Laboratory of Eco-Environmental Polymer Materials of
Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, China
| | - Tingting Xu
- Jiangsu
Key Laboratory of Molecular Imaging and Function Imaging, Department
of Radiology, Zhongda Hospital Southeast
University, School of Medicine, Southeast University, Nanjing 210009, China
| | - Hongyun Guo
- Institute
of Gansu Medical Science Research, Gansu
Provincial Cancer Hospital, Lanzhou 730050, China
| | - Yongdong Zhang
- Institute
of Gansu Medical Science Research, Gansu
Provincial Cancer Hospital, Lanzhou 730050, China
| | - Xingdong Wang
- Institute
of Gansu Medical Science Research, Gansu
Provincial Cancer Hospital, Lanzhou 730050, China
| | - Xiaoqi Wang
- Institute
of Gansu Medical Science Research, Gansu
Provincial Cancer Hospital, Lanzhou 730050, China
| | - Gaojun Teng
- Jiangsu
Key Laboratory of Molecular Imaging and Function Imaging, Department
of Radiology, Zhongda Hospital Southeast
University, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ziqiang Lei
- Key
Laboratory of Eco-Functional Polymer Materials of the Ministry of
Education, Key Laboratory of Eco-Environmental Polymer Materials of
Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, China
| |
Collapse
|
45
|
Yang H, Tong Z, Sun S, Mao Z. Enhancement of tumour penetration by nanomedicines through strategies based on transport processes and barriers. J Control Release 2020; 328:28-44. [PMID: 32858072 DOI: 10.1016/j.jconrel.2020.08.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
Nanomedicines for antitumour therapy have been widely studied in recent decades, but only a few have been used in clinical applications. One of the most important reasons is the poor tumour permeability of the nanomedicines. In this three-part review, intravascular, transvascular and extravascular transport were introduced one by one according to their roles in the overall process of nanomedicine transport into tumours. Transportation obstacles, such as elevated interstitial fluid pressure (IFP), abnormal blood vessels, dense tumour extracellular matrix (ECM) and binding site barriers (BSB), were each discussed in the context of the respective transport processes. Furthermore, homologous resolution strategies were summarized on the basis of each transportation obstacle, such as the normalization of blood vessels, regulation of the tumour microenvironment (TME) and application of transformable nanoparticles. At the end of this review, we propose holistic, concrete, and innovative views for better tumour penetration of nanomedicines.
Collapse
Affiliation(s)
- Huang Yang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China.
| | - Zongrui Tong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Shichao Sun
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| |
Collapse
|
46
|
Sun J, Wan Z, Chen Y, Xu J, Luo Z, Parise RA, Diao D, Ren P, Beumer JH, Lu B, Li S. Triple drugs co-delivered by a small gemcitabine-based carrier for pancreatic cancer immunochemotherapy. Acta Biomater 2020; 106:289-300. [PMID: 32004652 PMCID: PMC7183357 DOI: 10.1016/j.actbio.2020.01.039] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 12/15/2022]
Abstract
Poor tumor penetration and highly immunosuppressive tumor microenvironment are two major factors that limit the therapeutic efficacy for the treatment of pancreatic ductal adenocarcinoma (PDA). In this work, a redox-responsive gemcitabine (GEM)-conjugated polymer, PGEM, was employed as a tumor penetrating nanocarrier to co-load an immunomodulating agent (NLG919, an inhibitor of indoleamine 2,3-dioxygenase 1 (IDO1) and a chemotherapeutic drug (paclitaxel (PTX)) for immunochemo combination therapy. The NLG919/PTX co-loaded micelles showed very small size of ~15 nm. In vivo tumor imaging study indicated that PGEM was much more effective than the relatively large-sized POEG-co-PVD nanoparticles (~160 nm) in deep tumor penetration and could reach the core of the pancreatic tumor. PTX formulated in the PGEM carrier showed improved tumor inhibition effect compared with PGEM alone. Incorporation of NLG919 in the formulation led to a more immunoactive tumor microenvironment with significantly decreased percentage of Treg cells, and increased percentages of CD4+ IFNγ+ T and CD8+ IFNγ+ T cells. PGEM micelles co-loaded with PTX and NLG919 showed the best anti-tumor activity in pancreatic (PANC02) as well as two other tumor models compared to PGEM micelles loaded with PTX or NLG919 alone, suggesting that codelivery of NLG919 and PTX via PGEM may represent an effective strategy for immunochemotherapy of PDA as well as other types of cancers. STATEMENT OF SIGNIFICANCE: In order to effectively accumulate and penetrate the PDA that is poorly vascularized and enriched with dense fibrotic stroma, the size of nanomedicine has to be well controlled. Here, we reported an immunochemotherapy regimen based on co-delivery of GEM, PTX and IDO1 inhibitor NLG919 through an ultra-small sized GEM-based nanocarrier (PGEM). We demonstrated that the PGEM carrier was effective in accumulating and penetrating into PDA tumors. Besides, PGEM co-loaded with PTX and NLG9 induced an improved anti-tumor immune response and was highly efficacious in inhibiting tumor growth as well as in prolonging the survival rate in PANC02 xenograft model. Our work represents a potential strategy for enhancing PDA tumor penetration and immunochemotherapy.
Collapse
Affiliation(s)
- Jingjing Sun
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Zhuoya Wan
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yichao Chen
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jieni Xu
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhangyi Luo
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert A Parise
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dingwei Diao
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Pengfei Ren
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jan H Beumer
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Song Li
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
47
|
Tumor extravasation and infiltration as barriers of nanomedicine for high efficacy: The current status and transcytosis strategy. Biomaterials 2020; 240:119902. [PMID: 32105817 DOI: 10.1016/j.biomaterials.2020.119902] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/15/2020] [Accepted: 02/15/2020] [Indexed: 12/12/2022]
Abstract
Nanotechnology-based drug delivery platforms have been explored for cancer treatments and resulted in several nanomedicines in clinical uses and many in clinical trials. However, current nanomedicines have not met the expected clinical therapeutic efficacy. Thus, improving therapeutic efficacy is the foremost pressing task of nanomedicine research. An effective nanomedicine must overcome biological barriers to go through at least five steps to deliver an effective drug into the cytosol of all the cancer cells in a tumor. Of these barriers, nanomedicine extravasation into and infiltration throughout the tumor are the two main unsolved blockages. Up to now, almost all the nanomedicines are designed to rely on the high permeability of tumor blood vessels to extravasate into tumor interstitium, i.e., the enhanced permeability and retention (EPR) effect or so-called "passive tumor accumulation"; however, the EPR features are not so characteristic in human tumors as in the animal tumor models. Following extravasation, the large size nanomedicines are almost motionless in the densely packed tumor microenvironment, making them restricted in the periphery of tumor blood vessels rather than infiltrating in the tumors and thus inaccessible to the distal but highly malignant cells. Recently, we demonstrated using nanocarriers to induce transcytosis of endothelial and cancer cells to enable nanomedicines to actively extravasate into and infiltrate in solid tumors, which led to radically increased anticancer activity. In this perspective, we make a brief discussion about how active transcytosis can be employed to overcome the difficulties, as mentioned above, and solve the inherent extravasation and infiltration dilemmas of nanomedicines.
Collapse
|
48
|
Kokkinos J, Ignacio RMC, Sharbeen G, Boyer C, Gonzales-Aloy E, Goldstein D, Australian Pancreatic Cancer Genome Initiative Apgi, McCarroll JA, Phillips PA. Targeting the undruggable in pancreatic cancer using nano-based gene silencing drugs. Biomaterials 2020; 240:119742. [PMID: 32088410 DOI: 10.1016/j.biomaterials.2019.119742] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/03/2019] [Accepted: 12/25/2019] [Indexed: 12/20/2022]
Abstract
Pancreatic cancer is predicted to be the second leading cause of cancer-related death by 2025. The best chemotherapy only extends survival by an average of 18 weeks. The extensive fibrotic stroma surrounding the tumor curbs therapeutic options as chemotherapy drugs cannot freely penetrate the tumor. RNA interference (RNAi) has emerged as a promising approach to revolutionize cancer treatment. Small interfering RNA (siRNA) can be designed to inhibit the expression of any gene which is important given the high degree of genetic heterogeneity present in pancreatic tumors. Despite the potential of siRNA therapies, there are hurdles limiting their clinical application such as poor transport across biological barriers, limited cellular uptake, degradation, and rapid clearance. Nanotechnology can address these challenges. In fact, the past few decades have seen the conceptualization, design, pre-clinical testing and recent clinical approval of a RNAi nanodrug to treat disease. In this review, we comment on the current state of play of clinical trials evaluating siRNA nanodrugs and review pre-clinical studies investigating the efficacy of siRNA therapeutics in pancreatic cancer. We assess the physiological barriers unique to pancreatic cancer that need to be considered when designing and testing new nanomedicines for this disease.
Collapse
Affiliation(s)
- John Kokkinos
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW, Sydney, NSW, 2052, Australia; Australian Centre for Nanomedicine, UNSW, Sydney, NSW, 2052, Australia
| | - Rosa Mistica C Ignacio
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW, Sydney, NSW, 2052, Australia
| | - George Sharbeen
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW, Sydney, NSW, 2052, Australia
| | - Cyrille Boyer
- Australian Centre for Nanomedicine, UNSW, Sydney, NSW, 2052, Australia; Centre for Advanced Macromolecular Design, School of Chemical Engineering, UNSW, Sydney, NSW, 2052, Australia
| | - Estrella Gonzales-Aloy
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW, Sydney, NSW, 2052, Australia
| | - David Goldstein
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW, Sydney, NSW, 2052, Australia; Prince of Wales Hospital, Prince of Wales Clinical School, Sydney, NSW, 2052, Australia
| | | | - Joshua A McCarroll
- Australian Centre for Nanomedicine, UNSW, Sydney, NSW, 2052, Australia; Tumour Biology & Targeting Program, Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia, 2031; School of Women's and Children's Health, Faculty of Medicine, UNSW, Sydney, NSW, 2052, Australia.
| | - Phoebe A Phillips
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW, Sydney, NSW, 2052, Australia; Australian Centre for Nanomedicine, UNSW, Sydney, NSW, 2052, Australia.
| |
Collapse
|
49
|
Alanazi SA, Alanazi F, Haq N, Shakeel F, Badran MM, Harisa GI. Lipoproteins-Nanocarriers as a Promising Approach for Targeting Liver Cancer: Present Status and Application Prospects. Curr Drug Deliv 2020; 17:826-844. [PMID: 32026776 DOI: 10.2174/1567201817666200206104338] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/27/2019] [Accepted: 01/28/2020] [Indexed: 12/14/2022]
Abstract
The prevalence of liver cancer is increasing over the years and it is the fifth leading cause of mortality worldwide. The intrusive features and burden of low survival rate make it a global health issue in both developing and developed countries. The recommended chemotherapy drugs for patients in the intermediate and advanced stages of various liver cancers yield a low response rate due to the nonspecific nature of drug delivery, thus warranting the search for new therapeutic strategies and potential drug delivery carriers. There are several new drug delivery methods available to ferry the targeted molecules to the specific biological environment. In recent years, the nano assembly of lipoprotein moieties (lipidic nanoparticles) has emerged as a promising and efficiently tailored drug delivery system in liver cancer treatment. This increased precision of nano lipoproteins conjugates in chemotherapeutic targeting offers new avenues for the treatment of liver cancer with high specificity and efficiency. This present review is focused on concisely outlining the knowledge of liver cancer diagnosis, existing treatment strategies, lipoproteins, their preparation, mechanism and their potential application in the treatment of liver cancer.
Collapse
Affiliation(s)
- Saleh A Alanazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Fars Alanazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nazrul Haq
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Faiyaz Shakeel
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed M Badran
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Gamaleldin I Harisa
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
50
|
Kang H, Rho S, Stiles WR, Hu S, Baek Y, Hwang DW, Kashiwagi S, Kim MS, Choi HS. Size-Dependent EPR Effect of Polymeric Nanoparticles on Tumor Targeting. Adv Healthc Mater 2020; 9:e1901223. [PMID: 31794153 DOI: 10.1002/adhm.201901223] [Citation(s) in RCA: 276] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/14/2019] [Indexed: 12/13/2022]
Abstract
Passive targeting of large nanoparticles by the enhanced permeability and retention (EPR) effect is a crucial concept for solid tumor targeting in cancer nanomedicine. There is, however, a trade-off between the long-term blood circulation of nanoparticles and their nonspecific background tissue uptake. To define this size-dependent EPR effect, near-infrared fluorophore-conjugated polyethylene glycols (PEG-ZW800s; 1-60 kDa) are designed and their biodistribution, pharmacokinetics, and renal clearance are evaluated in tumor-bearing mice. The targeting efficiency of size-variant PEG-ZW800s is investigated in terms of tumor-to-background ratio (TBR). Interestingly, smaller sized PEGs (≤20 kDa, 12 nm) exhibit significant tumor targeting with minimum to no nonspecific uptakes, while larger sized PEGs (>20 kDa, 13 nm) accumulate highly in major organs, including the lungs, liver, and pancreas. Among those tested, 20 kDa PEG-ZW800 exhibits the highest TBR, while excreting unbound molecules to the urinary bladder. This result lays a foundation for engineering tumor-targeted nanoparticles and therapeutics based on the size-dependent EPR effect.
Collapse
Affiliation(s)
- Homan Kang
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical School Boston MA 02114 USA
| | - Sunghoon Rho
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical School Boston MA 02114 USA
| | - Wesley R. Stiles
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical School Boston MA 02114 USA
| | - Shuang Hu
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical School Boston MA 02114 USA
| | - Yoonji Baek
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical School Boston MA 02114 USA
| | - Do Won Hwang
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical School Boston MA 02114 USA
| | - Satoshi Kashiwagi
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical School Boston MA 02114 USA
| | - Moon Suk Kim
- Department of Molecular Science and TechnologyAjou University Suwon 16499 South Korea
| | - Hak Soo Choi
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical School Boston MA 02114 USA
| |
Collapse
|