1
|
Santos LS, Tonel MZ, Martins MO, dos Santos CL. Theoretical Exploration of Chitosan Nanoparticles Associated with Platinum Compounds for Cancer Treatment: Insights from DFT and Molecular Docking Analyses. BIONANOSCIENCE 2025; 15:79. [DOI: 10.1007/s12668-024-01728-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2024] [Indexed: 01/04/2025]
|
2
|
Zhu Z, Shang Y, Lin C, Zhang D, Ai L, Li Y, Tan W, Liu Y, Zhao Z. Targeted Covalent Nanodrugs Reinvigorate Antitumor Immunity and Kill Tumors via Improving Intratumoral Accumulation and Retention of Doxorubicin. ACS NANO 2025; 19:2315-2333. [PMID: 39760789 DOI: 10.1021/acsnano.4c12447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Specifically improving the intratumoral accumulation and retention and achieving the maximum therapeutic efficacy of small-molecule chemotherapeutics remains a considerable challenge. To address the issue, we here reported near-infrared (NIR) irradiation-activatable targeted covalent nanodrugs by installing diazirine-labeled transferrin receptor 1 (TfR1)-targeted aptamers on PEGylated phospholipid-coated upconversion nanoparticles followed by doxorubicin loading. Targeted covalent nanodrugs recognized and then were activated to covalently cross-link with TfR1 on cancer cells by 980 nm NIR irradiation. Systematic studies revealed that they achieved >6- and >5.5-fold higher intratumoral accumulations of doxorubicin than aptamer-based targeted nanodrugs at 6 and 120 h post intravenous injection, respectively. Based on high drug delivery efficacy, targeted covalent nanodrugs boosted doxorubicin-induced immunogenic cell death, activated antitumor immune responses and shrank the sizes of both primary and distant tumors, and displayed better therapeutic efficacy and less adverse effect than targeted nanodrugs and commercial Doxil in 4T1 syngeneic breast tumor model featuring an immunosuppressive microenvironment. By integrating the specificity of molecular recognition, the reactivity profile of diazirine and the accuracy of light manipulation with nanodrug supremacy, our targeted covalent nanodrugs could be expected as a longer-term and efficient strategy to improve anticancer therapeutic efficacy of chemotherapeutics.
Collapse
Affiliation(s)
- Zhijia Zhu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Yanxue Shang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Chukai Lin
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Dongchen Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Lili Ai
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Youshan Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yanlan Liu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Zilong Zhao
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|
3
|
Zohora FT, Pathmanathan R, Chowdhury EH. Application of Strontium Chloride Hexahydrate to Synthesize Strontium-Substituted Carbonate Apatite as a pH-Sensitive, Biologically Safe, and Highly Efficient siRNA Nanocarrier. ACS APPLIED BIO MATERIALS 2025; 8:348-367. [PMID: 39723844 DOI: 10.1021/acsabm.4c01319] [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] [Indexed: 12/28/2024]
Abstract
Naked siRNAs are sensitive to enzymatic degradation, phagocytic entrapment, quick renal excretion, membrane impermeability, endosomal escape, and off-target effects. Designing a safe and efficient nanocarrier for siRNA delivery to the target site without toxicity remains a significant hurdle in gene therapy. CA is a unique derivative of hydroxyapatite and a highly pH-sensitive nanocarrier with strong particle aggregation and a high polydispersity index. Strontium (Sr2+), a group two divalent metal in the periodic table, has been reported for substituting calcium (Ca2+) ions from the apatite lattice and limiting particle growth/aggregation. This study used strontium chloride hexahydrate (SrCl2·6H2O) salt to develop a Sr-substituted CA (Sr-CA) nanocarrier with ∼30 nm size, spherical shape, less aggregation, homodispersity, and a fair anionic charge. Sr-CA demonstrated a large surface area-to-volume ratio, an improved cargo loading efficiency, and enhanced cellular uptake in HEK-293 cells. Moreover, Sr-CA is a pH-responsive nanocarrier responsible for its long physiological stability, efficient endosomal escape, and optimal cargo delivery within cells. These NPs have differential effects on MAPK1, MAP2K4, PIK3Ca, CAMK4, and p53 gene expression in HEK-293 cells without showing any significant cytotoxicity in cell growth properties. Gene silencing by Sr-CA-mediated siRNA delivery against MAPK1, MAP2K4, PIK3Ca, and CAMK4 genes significantly decreased the level of target gene expression and cell survival, demonstrating successful intracellular siRNA delivery in HEK-293 cells. Additionally, biocompatibility testing confirmed the biological safety of the Sr-CA nanocarrier in mice. These findings suggest that Sr-CA nanocarriers are a promising siRNA delivery system, combining high efficiency with pH-sensitive release and excellent biocompatibility, making them a viable option for future therapeutic applications.
Collapse
Affiliation(s)
- Fatema Tuz Zohora
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Rajadurai Pathmanathan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Ezharul Hoque Chowdhury
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
- Daffodil International University, Daffodil Smart City, Birulia 1216, Bangladesh
- Nanoflex LLC, 31756 Broadwater Avenue, Leesburg, Florida 34748, United States
| |
Collapse
|
4
|
Jiang L, Wu A, Zeng L, Zhou B, Zhao M, Fan M, Jin Z, He Q. A Slimming/Excavating Strategy for Enhanced Intratumoral Penetration of Acid-Disassemblable NO-Releasing Nanomedicines. Adv Healthc Mater 2025:e2404085. [PMID: 39757461 DOI: 10.1002/adhm.202404085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/28/2024] [Indexed: 01/07/2025]
Abstract
Poor tumor penetration is the major predicament of nanomedicines that limits their anticancer efficacy. The dense extracellular matrix (ECM) in the tumor is one of the major barriers against the deep penetration of nanomedicines. In this work, a slimming/excavating strategy is proposed for enhanced intratumoral penetration based on an acid-disassemblable nanomicelles-assembled nanomedicine and the NO-mediated degradation of ECM. The nanomedicine is constructed by cross-linking nanomicelles, which are self-assembled with two kinds of dendrimers containing phenylboronic acid and lactobionic acid, through borate esterification. In the acidic tumor microenvironment, the pH-sensitive borate ester bonds among the nanomicelles are hydrolyzed, triggering the disassembly of nanomedicine (≈150 nm) into small nanomicelles (≈25 nm). In response to the intratumoral over-expressed glutathione (GSH), the NO donor loaded in the nanomicelles produces NO, which mediates the expression of matrix metalloproteinases for the degradation of ECM in the tumor. By collaboration of the disassembling behavior of nanomedicine with the NO-mediated degradation of ECM, the designed nanomedicine can penetrate a long distance in tumors. The proposed slimming/excavating strategy will provide inspiration for overcoming the challenge of nanomedicines in tumor penetration.
Collapse
Affiliation(s)
- Lingdong Jiang
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Anbang Wu
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, 518060, China
| | - Lingting Zeng
- Shanghai Key Laboratory of Hydrogen Science & Center of Hydrogen Science, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Bin Zhou
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, 518060, China
| | - Min Zhao
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, 518060, China
| | - Mingjian Fan
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, 518060, China
| | - Zhaokui Jin
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, 518060, China
| | - Qianjun He
- Shanghai Key Laboratory of Hydrogen Science & Center of Hydrogen Science, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
5
|
Cassani M, Fernandes S, Pagliari S, Cavalieri F, Caruso F, Forte G. Unraveling the Role of the Tumor Extracellular Matrix to Inform Nanoparticle Design for Nanomedicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409898. [PMID: 39629891 PMCID: PMC11727388 DOI: 10.1002/advs.202409898] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/01/2024] [Indexed: 01/14/2025]
Abstract
The extracellular matrix (ECM)-and its mechanobiology-regulates key cellular functions that drive tumor growth and development. Accordingly, mechanotherapy is emerging as an effective approach to treat fibrotic diseases such as cancer. Through restoring the ECM to healthy-like conditions, this treatment aims to improve tissue perfusion, facilitating the delivery of chemotherapies. In particular, the manipulation of ECM is gaining interest as a valuable strategy for developing innovative treatments based on nanoparticles (NPs). However, further progress is required; for instance, it is known that the presence of a dense ECM, which hampers the penetration of NPs, primarily impacts the efficacy of nanomedicines. Furthermore, most 2D in vitro studies fail to recapitulate the physiological deposition of matrix components. To address these issues, a comprehensive understanding of the interactions between the ECM and NPs is needed. This review focuses on the main features of the ECM and its complex interplay with NPs. Recent advances in mechanotherapy are discussed and insights are offered into how its combination with nanomedicine can help improve nanomaterials design and advance their clinical translation.
Collapse
Affiliation(s)
- Marco Cassani
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Soraia Fernandes
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
| | - Stefania Pagliari
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonWC2R 2LSUK
| | - Francesca Cavalieri
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
- Dipartimento di Scienze e Tecnologie ChimicheUniversita di Roma “Tor Vergata”Via della Ricerca Scientifica 1Rome00133Italy
| | - Frank Caruso
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Giancarlo Forte
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonWC2R 2LSUK
| |
Collapse
|
6
|
Sivasubramanian PD, Unnikrishnan G, Kolanthai E, Muthuswamy S. Engineered nanoparticle systems: A review on emerging strategies for enhanced cancer therapeutics. NEXT MATERIALS 2025; 6:100405. [DOI: 10.1016/j.nxmate.2024.100405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
7
|
Lang J, Schorr K, Goepferich A. Towards a switchable nanoparticle behavior using inverse electron-demand Diels-Alder chemistry and ectoenzyme-based ligand activation. Eur J Pharm Sci 2025; 204:106944. [PMID: 39461601 DOI: 10.1016/j.ejps.2024.106944] [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: 07/16/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Nanoparticles (NPs) as drug delivery platforms encounter numerous obstacles on their journey from administration to the target site. Often, diametrically opposing particle properties are desirable to overcome biological and physical barriers. Therefore, stimuli-responsive NPs have been developed to allow for specific particle adaptation. In this work, it was demonstrated that NPs can be rendered switchable with respect to their interaction with a receptor through an external chemical stimulus. A combination of the inverse electron-demand Diels-Alder (iEDDA) reaction for subsequent NP functionalization and ectoenzyme-based ligand activation allowed for specific particle tailoring. Building on this, a two-step process for target cell recognition was developed. First, NPs were functionalized with Angiotensin-I (Ang-I) as inactive ligand using iEDDA chemistry. At the target site, the ligand was enzymatically processed to Angiotensin-ll (Ang-II) by cellular ectoenzymes. Ang-ll binds as active ligand to the angiotensin ll type 1 (AT1) receptor on the target cell surface. This enzymatic activation aims to minimize the biological effect of the ligand prior to particle binding, while the NP target cell specificity is increased by a two-step recognition with enzymatic processing and receptor binding.
Collapse
Affiliation(s)
- Johannes Lang
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, 93053, Germany
| | - Kathrin Schorr
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, 93053, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, 93053, Germany.
| |
Collapse
|
8
|
Richards CJ, Melero Martinez P, Roos WH, Åberg C. High-throughput approach to measure number of nanoparticles associated with cells: size dependence and kinetic parameters. NANOSCALE ADVANCES 2024; 7:185-195. [PMID: 39569330 PMCID: PMC11575555 DOI: 10.1039/d4na00589a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024]
Abstract
Understanding how nanoparticle properties influence uptake by cells is highly important for developing nanomedicine design principles. For this, quantitative studies where actual numbers of cell-associated particles are determined are highly relevant. However, many techniques able to measure particle numbers suffer from low-throughput or place requirements on the types of nanoparticles that can be measured. Here we show the usage of flow cytometry to measure numbers of cell-associated nanoparticles for particles ranging in size from 100-500 nm, and extend this range to 40-500 nm by separate calibration. For the 100 nm particles, we corroborate the numbers by direct, low-throughput, counting using fluorescence microscopy. Applying flow cytometry we subsequently investigated the effect of particle size on the number of cell-associated particles for various timespans up to 5 h and found only a minor effect of size between 40, 100, and 200 nm particles. Next, we measured the kinetic rate constants describing the adsorption, desorption, and internalization for the 100 nm particles specifically. In general, we found values in accordance with previous literature. We foresee the future usage of the methodology applied here to investigate the kinetics of nanoparticle cellular uptake for a variety of particle types.
Collapse
Affiliation(s)
- Ceri J Richards
- Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen 9713 AV Groningen Netherlands
- Molecular Biophysics, Zernike Institute for Advanced Materials, University of Groningen 9747 AG Groningen Netherlands
| | - Paula Melero Martinez
- Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen 9713 AV Groningen Netherlands
| | - Wouter H Roos
- Molecular Biophysics, Zernike Institute for Advanced Materials, University of Groningen 9747 AG Groningen Netherlands
| | - Christoffer Åberg
- Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen 9713 AV Groningen Netherlands
| |
Collapse
|
9
|
Zhang W, Wang X, Wang J, Su C, Bao Y, Xiong M. The Relationship between In Vivo Toxicity and Responsive pH in Transistor-Like pH-Sensitive Nanodetergents. SMALL METHODS 2024:e2401263. [PMID: 39676467 DOI: 10.1002/smtd.202401263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/05/2024] [Indexed: 12/17/2024]
Abstract
Subacidity-responsive materials (saRMs) have attracted considerable attention for disease-specific pH-responsive imaging and therapy. However, the guidance for their pH-responsive design, aimed at achieving effective responses at lesion sites while minimizing unwanted responses in normal tissues, is inadequate and challenged by the subtle pH difference between the desired responsive pH and the pH of normal tissues. Here, the correlation between the responsive pH of 'proton transistor' nanodetergents (pTNTs) is investigated and the in vivo toxicity caused by unwanted responses in normal tissues, taking advantage of their refined responsive pH and the easily characterized membranolytic activity and cytotoxicity following response. It is designed and selected five pTNTs that undergo a refined transition from an inactive "OFF" state with sealed membranolytic activity and cytotoxicity to an active "ON" state with potent membranolytic activity and cytotoxicity within a 0.1 pH perturbation at transition pH (pHt) values of 7.2, 7.1, 6.9, 6.8, and 6.7, respectively. A significant correlation between the in vivo toxicity of these pTNTs and their pHt for membranolytic activity is observed. And non-negligible changes in the organ toxicity of pTNTs are induced by every 0.1 or 0.2 pH shift of pHt. After intravenous administration, pTNTs with a pHt value of 7.2 or 7.1 induced significant hepatotoxicity and cardiotoxicity, while no significant toxicity is detected for pTNTs with pHt values ranging from 6.8 to 6.7. This hepatoxicity is found to be associated with the tissue's pH environment-dependent activation of membranolytic activity. This study can provide guidance for designing pH-responsive membranolytic materials and saRMs to minimize their toxicity and unwanted response in normal tissues.
Collapse
Affiliation(s)
- Weinan Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
- School of Biomedical Engineering, and Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, P. R. China
| | - Xiaochuan Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Jihong Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Chanjuan Su
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, and Medical Research Center, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Yan Bao
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, and Medical Research Center, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Menghua Xiong
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| |
Collapse
|
10
|
Guo Z, Xiu L, Li Y, Tan J, Wei C, Sui J, Zhang S, Zhu R, Li JL. Injectable nanocomposite hydrogel with cascade drug release for treatment of uveal melanoma. J Control Release 2024; 376:1086-1099. [PMID: 39500408 DOI: 10.1016/j.jconrel.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/28/2024] [Accepted: 11/01/2024] [Indexed: 11/11/2024]
Abstract
Uveal melanoma (UM) is the most common malignant intraocular tumor with the trait of distant metastases. Currently, the standard clinical therapy results in suboptimal outcomes due to ineffective inhibition of tumor metastasis. Thus, developing novel therapeutic modalities for UM remains a critical priority. Herein, we have developed an injectable nanocomposite hydrogel (HA-DOX/LAP gel) through integrating hyaluronic acid-based drug-loaded nanoparticles into an alginate-dopamine gel, delivering the chemotherapeutic drugs, lapatinib and doxorubicin for combinational treatment of UM. HA-DOX/LAP gel is fabricated in situ by a simple injection of the mixed precursor solution into tumor sites and maintains in vivo for more than 21 days. The entrapped drug-loaded nanoparticles can gradually release from HA-DOX/LAP gel, enhancing tumor targeting and penetration, and synchronously releasing lapatinib and doxorubicin into the acidic intracellular environment to synergistically destroy UM cells. In vivo anti-tumor studies conducted in MuM-2B tumor models demonstrated that HA-DOX/LAP gel significantly impedes tumor growth, diminishes postoperative recurrence, and prolongs overall survivals of UM tumor-bearing mice through only single injection. Remarkably, the escaped drug-loaded nanoparticles effectively reduce the risk of tumor metastases. Our findings provide new insights for the development of multifunctional nanocomposite-incorporating combination therapy against UM by targeting tumor recurrence and metastases.
Collapse
Affiliation(s)
- Zhihao Guo
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China.
| | - Linyun Xiu
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yumei Li
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jiangcheng Tan
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Cailing Wei
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Junhui Sui
- Beijing Tide Pharmaceutical Co., Ltd., Beijing 100176, China
| | - Shijin Zhang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Ruohua Zhu
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Ji-Liang Li
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; University of Chinese Academy of Sciences Wenzhou Institute, Wenzhou 325001, China.
| |
Collapse
|
11
|
Cao Z, Liu J, Yang X. Deformable nanocarriers for enhanced drug delivery and cancer therapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230037. [PMID: 39439489 PMCID: PMC11491306 DOI: 10.1002/exp.20230037] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 01/28/2024] [Indexed: 10/25/2024]
Abstract
Recently, the field of nanomedicine has witnessed substantial advancements in the development of nanocarriers for targeted drug delivery, emerges as promising platforms to enhance therapeutic efficacy and minimize adverse effects associated with conventional chemotherapy. Notably, deformable nanocarriers have garnered considerable attention due to their unique capabilities of size changeable, tumor-specific aggregation, stimuli-triggered disintegration, and morphological transformations. These deformable nanocarriers present significant opportunities for revolutionizing drug delivery strategies, by responding to specific stimuli or environmental cues, enabling achieved various functions at the tumor site, including size-shrinkage nanocarriers enhance drug penetration, aggregative nanocarriers enhance retention effect, disintegrating nanocarriers enable controlled drug release, and shape-changing nanocarriers improve cellular uptake, allowing for personalized treatment approaches and combination therapies. This review provides an overview of recent developments and applications of deformable nanocarriers for enhancing tumor therapy, underscores the diverse design strategies employed to create deformable nanocarriers and elucidates their remarkable potential in targeted tumor therapy.
Collapse
Affiliation(s)
- Ziyang Cao
- Department of General SurgeryGuangzhou First People's Hospitalthe Second Affiliated HospitalSouth China University of TechnologyGuangzhouPeople's Republic of China
- Center for Medical Research on Innovation and TranslationInstitute of Clinical MedicineSchool of MedicineGuangzhou First People's HospitalSouth China University of TechnologyGuangzhouPeople's Republic of China
| | - Jing Liu
- School of ChemistryChemical Engineering and Biotechnology Nanyang Technological UniversitySingaporeSingapore
| | - Xianzhu Yang
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdongPeople's Republic of China
| |
Collapse
|
12
|
Wang X, Seah HL, Zhang XL, Zhuang Z, Liu XW. Fluorescent Self-Assembled Complexes Based on Glyco-Functionalized G-Quadruplexes as a Targeted Delivery Platform. ACS APPLIED MATERIALS & INTERFACES 2024; 16:50229-50237. [PMID: 39264898 DOI: 10.1021/acsami.4c08079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/14/2024]
Abstract
Targeted delivery systems combined with the stimuli-responsive release of drug molecules hold noteworthy promise for precision medicine, enabling treatments with enhanced effectiveness and reduced adverse effects. An ideal drug delivery platform with versatile targeting moieties, the capability of combinational payloads, and simple preparation is highly desirable. Herein, we developed pH-sensitive fluorescent self-assembled complexes (SACs) of a galactose-functionalized G-quadruplex (G4) and a coumarin carboxamidine derivative as a targeted delivery platform through the nanoprecipitation method. These SACs selectively targeted hepatocellular carcinoma (HepG2) cells in fluorescence imaging after a short incubation and exerted specific anticancer effects in an appropriate dose range. Co-delivery of 1 μM prodrug floxuridine oligomers and 16 μg/mL SACs (minimal hemolytic effect) significantly reduced the cytotoxicity of the nucleoside anticancer drug on normal cells (NIH/3T3), kept up to 70% alive after 72-h incubation, and improved anticancer efficacy compared to SACs alone. This strategy can be extended to ratiometric multidrug delivery through self-assembly for targeted combinational therapy.
Collapse
Affiliation(s)
- Xian Wang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Hui Ling Seah
- School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Xiao-Lin Zhang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Zeyan Zhuang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Xue-Wei Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| |
Collapse
|
13
|
Santos JAV, Silva D, Marques MPM, Batista de Carvalho LAE. Platinum-based chemotherapy: trends in organic nanodelivery systems. NANOSCALE 2024; 16:14640-14686. [PMID: 39037425 DOI: 10.1039/d4nr01483a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Despite the investment in platinum drugs research, cisplatin, carboplatin and oxaliplatin are still the only Pt-based compounds used as first line treatments for several cancers, with a few other compounds being approved for administration in some Asian countries. However, due to the severe and worldwide impact of oncological diseases, there is an urge for improved chemotherapeutic approaches. Furthermore, the pharmaceutical application of platinum complexes is hindered by their inherent toxicity and acquired resistance. Nanodelivery systems rose as a key strategy to overcome these challenges, with recognized versatility and ability towards improving the safety, bioavailability and efficacy of the available drugs. Among the known nanocarriers, organic systems have been widely applied, taking advantage of their potential as drug vehicles. Researchers have mainly focused on the development of lipidic and polymeric carriers, including supramolecular structures, with an overall improvement of encapsulated platinum complexes. Herein, an overview of recent trends and strategies is presented, with the main focus on the encapsulation of platinum compounds into organic nanocarriers, showcasing the evolution in the design and development of these promising systems. This comprehensive review highlights formulation methods as well as characterization procedures, providing insights that may be helpful for the development of novel platinum nanocarriers aiming at future pharmaceutical applications.
Collapse
Affiliation(s)
- João A V Santos
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal.
| | - Daniela Silva
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal.
| | - Maria Paula M Marques
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal.
- Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Luís A E Batista de Carvalho
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal.
| |
Collapse
|
14
|
Iureva AM, Nikitin PI, Tereshina ED, Nikitin MP, Shipunova VO. The influence of various polymer coatings on the in vitro and in vivo properties of PLGA nanoparticles: Comprehensive study. Eur J Pharm Biopharm 2024; 201:114366. [PMID: 38876361 DOI: 10.1016/j.ejpb.2024.114366] [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: 03/09/2024] [Revised: 06/03/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Nanoparticles based on poly(lactic-co-glycolic acid) (PLGA) with various surface chemistry are widely used in biomedicine for theranostic applications. The nature of the external coating of nanoparticles has a significant influence on their efficiency as drug carriers or visualization agents. However, information about the mechanisms of nanoparticle accumulation in tumors and the influence of their surface properties on biodistribution is scarce due to the lack of systematic evaluation. Here we investigate the effect of different polymer coatings of the surface on in vitro and in vivo properties of PLGA nanoparticles. Namely, cell binding efficiency, cytotoxicity, efficiency of fluorescent bioimaging, and tumor accumulation were tested. The highest binding efficiency in vitro and cytotoxicity were observed for positively charged polymers. Interestingly, in vivo fluorescent visualization of tumor-bearing mice and quantitative measurements of biodistribution of magnetite-loaded nanoparticles indicated different dependences of accumulation in tumors on the coating of PLGA nanoparticles. This means that nanoparticle surface properties can simultaneously enhance imaging efficiency and decrease quantitative accumulation in tumors. The obtained data demonstrate the complexity of the dependence of nanoparticles' effectiveness for theranostic applications on surface features. We believe that this study will contribute to the rational design of nanoparticles for effective cancer diagnostics and therapy.
Collapse
Affiliation(s)
- Anna M Iureva
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701 Dolgoprudny, Russia
| | - Petr I Nikitin
- Prokhorov General Physics Institute, Russian Academy of Sciences, 38 Vavilov Street, 119991 Moscow, Russia
| | - Ekaterina D Tereshina
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701 Dolgoprudny, Russia
| | - Maxim P Nikitin
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701 Dolgoprudny, Russia; Nanobiomedicine Division, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sochi, Russia
| | - Victoria O Shipunova
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701 Dolgoprudny, Russia; Nanobiomedicine Division, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sochi, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997 Moscow, Russia.
| |
Collapse
|
15
|
Saadh MJ, Mustafa MA, Kumar A, Alamir HTA, Kumar A, Khudair SA, Faisal A, Alubiady MHS, Jalal SS, Shafik SS, Ahmad I, Khry FAF, Abosaoda MK. Stealth Nanocarriers in Cancer Therapy: a Comprehensive Review of Design, Functionality, and Clinical Applications. AAPS PharmSciTech 2024; 25:140. [PMID: 38890191 DOI: 10.1208/s12249-024-02843-5] [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: 02/17/2024] [Accepted: 05/13/2024] [Indexed: 06/20/2024] Open
Abstract
Nanotechnology has significantly transformed cancer treatment by introducing innovative methods for delivering drugs effectively. This literature review provided an in-depth analysis of the role of nanocarriers in cancer therapy, with a particular focus on the critical concept of the 'stealth effect.' The stealth effect refers to the ability of nanocarriers to evade the immune system and overcome physiological barriers. The review investigated the design and composition of various nanocarriers, such as liposomes, micelles, and inorganic nanoparticles, highlighting the importance of surface modifications and functionalization. The complex interaction between the immune system, opsonization, phagocytosis, and the protein corona was examined to understand the stealth effect. The review carefully evaluated strategies to enhance the stealth effect, including surface coating with polymers, biomimetic camouflage, and targeting ligands. The in vivo behavior of stealth nanocarriers and their impact on pharmacokinetics, biodistribution, and toxicity were also systematically examined. Additionally, the review presented clinical applications, case studies of approved nanocarrier-based cancer therapies, and emerging formulations in clinical trials. Future directions and obstacles in the field, such as advancements in nanocarrier engineering, personalized nanomedicine, regulatory considerations, and ethical implications, were discussed in detail. The review concluded by summarizing key findings and emphasizing the transformative potential of stealth nanocarriers in revolutionizing cancer therapy. This review enhanced the comprehension of nanocarrier-based cancer therapies and their potential impact by providing insights into advanced studies, clinical applications, and regulatory considerations.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan.
| | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Technology, University of Imam Jaafar AL-Sadiq, Baghdad, Iraq
| | - Ashwani Kumar
- Department of Life Sciences, School of Sciences, Jain (Deemed-to-be) University, Bengaluru, Karnataka, India
- Department of Pharmacy, Vivekananda Global University, Jaipur, Rajasthan, India
| | | | - Abhishek Kumar
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, 247341, Uttar Pradesh, India
- Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | | | - Ahmed Faisal
- Department of Pharmacy, Al-Noor University College, Nineveh, Iraq
| | | | - Sarah Salah Jalal
- College of Pharmacy, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | - Shafik Shaker Shafik
- Experimental Nuclear Radiation Group, Scientific Research Center, Al-Ayen University, Thi-Qar, Iraq
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Faeza A F Khry
- Faculty of pharmacy, department of pharmaceutics, Al-Esraa University, Baghdad, Iraq
| | - Munther Kadhim Abosaoda
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Qadisiyyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
16
|
Huayamares SG, Loughrey D, Kim H, Dahlman JE, Sorscher EJ. Nucleic acid-based drugs for patients with solid tumours. Nat Rev Clin Oncol 2024; 21:407-427. [PMID: 38589512 DOI: 10.1038/s41571-024-00883-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/10/2024]
Abstract
The treatment of patients with advanced-stage solid tumours typically involves a multimodality approach (including surgery, chemotherapy, radiotherapy, targeted therapy and/or immunotherapy), which is often ultimately ineffective. Nucleic acid-based drugs, either as monotherapies or in combination with standard-of-care therapies, are rapidly emerging as novel treatments capable of generating responses in otherwise refractory tumours. These therapies include those using viral vectors (also referred to as gene therapies), several of which have now been approved by regulatory agencies, and nanoparticles containing mRNAs and a range of other nucleotides. In this Review, we describe the development and clinical activity of viral and non-viral nucleic acid-based treatments, including their mechanisms of action, tolerability and available efficacy data from patients with solid tumours. We also describe the effects of the tumour microenvironment on drug delivery for both systemically administered and locally administered agents. Finally, we discuss important trends resulting from ongoing clinical trials and preclinical testing, and manufacturing and/or stability considerations that are expected to underpin the next generation of nucleic acid agents for patients with solid tumours.
Collapse
Affiliation(s)
- Sebastian G Huayamares
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Emory University School of Medicine, Atlanta, GA, USA
| | - David Loughrey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Emory University School of Medicine, Atlanta, GA, USA
| | - Hyejin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Emory University School of Medicine, Atlanta, GA, USA
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Emory University School of Medicine, Atlanta, GA, USA.
| | - Eric J Sorscher
- Emory University School of Medicine, Atlanta, GA, USA.
- Department of Pediatrics, Emory University, Atlanta, GA, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
17
|
Yang J, Yang Z, Wang H, Chang Y, Xu JF, Zhang X. A Polymeric Nanoparticle to Co-Deliver Mitochondria-Targeting Peptides and Pt(IV) Prodrug: Toward High Loading Efficiency and Combination Efficacy. Angew Chem Int Ed Engl 2024; 63:e202402291. [PMID: 38380542 DOI: 10.1002/anie.202402291] [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: 01/31/2024] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 02/22/2024]
Abstract
Developing combination chemotherapy systems with high drug loading efficiency at predetermined drug ratios to achieve a synergistic effect is important for cancer therapy. Herein, a polymeric dual-drug nanoparticle composed of a Pt(IV) prodrug derived from oxaliplatin and a mitochondria-targeting cytotoxic peptide is constructed through emulsion interfacial polymerization, which processes high drug loading efficiency and high biocompatibility. The depolymerization of polymeric dual-drug nanoparticle and the activation of Pt prodrug can be effectively triggered by the acidic tumor environment extracellularly and the high levels of glutathione intracellularly in cancer cells, respectively. The utilization of mitochondria-targeting peptide can inhibit ATP-dependent processes including drug efflux and DNA damage repair. This leads to increased accumulation of Pt-drugs within cancer cells. Eventually, the polymeric dual-drug nanoparticle demonstrates appreciable antitumor effects on both cell line derived and patient derived xenograft lung cancer model. It is highly anticipated that the polymeric dual-or multi-drug systems can be applied for combination chemotherapy to achieve enhanced anticancer activity and reduced side effects.
Collapse
Affiliation(s)
- Jinpeng Yang
- Key Lab of Organic Optoelectronics & Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Zhenlin Yang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hua Wang
- Key Lab of Organic Optoelectronics & Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yincheng Chang
- College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jiang-Fei Xu
- Key Lab of Organic Optoelectronics & Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Xi Zhang
- Key Lab of Organic Optoelectronics & Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
18
|
Huang X, Cao Z, Qian J, Ding T, Wu Y, Zhang H, Zhong S, Wang X, Ren X, Zhang W, Xu Y, Yao G, Wang X, Yang X, Wen L, Zhang Y. Nanoreceptors promote mutant p53 protein degradation by mimicking selective autophagy receptors. NATURE NANOTECHNOLOGY 2024; 19:545-553. [PMID: 38216684 DOI: 10.1038/s41565-023-01562-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/01/2023] [Indexed: 01/14/2024]
Abstract
In some cancers mutant p53 promotes the occurrence, development, metastasis and drug resistance of tumours, with targeted protein degradation seen as an effective therapeutic strategy. However, a lack of specific autophagy receptors limits this. Here, we propose the synthesis of biomimetic nanoreceptors (NRs) that mimic selective autophagy receptors. The NRs have both a component for targeting the desired protein, mutant-p53-binding peptide, and a component for enhancing degradation, cationic lipid. The peptide can bind to mutant p53 while the cationic lipid simultaneously targets autophagosomes and elevates the levels of autophagosome formation, increasing mutant p53 degradation. The NRs are demonstrated in vitro and in a patient-derived xenograft ovarian cancer model in vivo. The work highlights a possible direction for treating diseases by protein degradation.
Collapse
Affiliation(s)
- Xiaowan Huang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | - Ziyang Cao
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | - Jieying Qian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China
| | - Tao Ding
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | - Yanxia Wu
- Molecular Cancer Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen, People's Republic of China
| | - Hao Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China
| | - Suqin Zhong
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | - Xiaoli Wang
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | - Xiaoguang Ren
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | - Wang Zhang
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China
| | - Youcui Xu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China
| | - Guangyu Yao
- Breast Center, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xingwu Wang
- Molecular Cancer Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen, People's Republic of China
| | - Xianzhu Yang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China.
- National Engineering Research Centre for Tissue Restoration and Reconstruction and Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, People's Republic of China.
| | - Longping Wen
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People's Republic of China.
| | - Yunjiao Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China.
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China.
- National Engineering Research Centre for Tissue Restoration and Reconstruction and Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, People's Republic of China.
| |
Collapse
|
19
|
Wang AJY, Yan C, Reike MJ, Black PC, Contreras-Sanz A. A systematic review of nanocarriers for treatment of urologic cancers. Urol Oncol 2024; 42:75-101. [PMID: 38161104 DOI: 10.1016/j.urolonc.2023.11.022] [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: 08/28/2023] [Revised: 11/26/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024]
Abstract
Nanocarriers (NCs) are a form of nanotechnology widely investigated in cancer treatment to improve the safety and efficacy of systemic therapies by increasing tumor specificity. Numerous clinical trials have explored the use of NCs in urologic cancers since the approval of the first NCs for cancer treatment over 20 years ago. The objective of this systematic review is to examine the effectiveness and safety of NCs in treating urological cancers. This paper summarizes the state of the field by investigating peer-reviewed, published results from 43 clinical trials involving the use of NCs in bladder, prostate, and kidney cancer patients with a focus on safety and efficacy data. Among the 43 trials, 16 were phase I, 20 phase II, and 4 phase I/II. No phase III trials have been reported. While both novel and classic NCs have been explored in urologic cancers, NCs already approved for the treatment of other cancers were more widely represented. Trials in prostate cancer and mixed trials involving both urologic and non-urologic cancer patients were the most commonly reported trials. Although NCs have demonstrable efficacy with adequate safety in non-urologic cancer patient populations, current clinical stage NC options appear to be less beneficial in the urologic cancer setting. For example, nab-paclitaxel and liposomal doxorubicin have proven ineffective in the treatment of urologic cancers despite successes in other cancers. However, several ongoing pre-clinical studies using targeted and locally applied improved NCs may eventually improve their utility.
Collapse
Affiliation(s)
- Amy J Y Wang
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cathy Yan
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Moritz J Reike
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter C Black
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada..
| | - Alberto Contreras-Sanz
- The Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada..
| |
Collapse
|
20
|
Zhou Q, Liu Q, Wang Y, Chen J, Schmid O, Rehberg M, Yang L. Bridging Smart Nanosystems with Clinically Relevant Models and Advanced Imaging for Precision Drug Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308659. [PMID: 38282076 PMCID: PMC11005737 DOI: 10.1002/advs.202308659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Indexed: 01/30/2024]
Abstract
Intracellular delivery of nano-drug-carriers (NDC) to specific cells, diseased regions, or solid tumors has entered the era of precision medicine that requires systematic knowledge of nano-biological interactions from multidisciplinary perspectives. To this end, this review first provides an overview of membrane-disruption methods such as electroporation, sonoporation, photoporation, microfluidic delivery, and microinjection with the merits of high-throughput and enhanced efficiency for in vitro NDC delivery. The impact of NDC characteristics including particle size, shape, charge, hydrophobicity, and elasticity on cellular uptake are elaborated and several types of NDC systems aiming for hierarchical targeting and delivery in vivo are reviewed. Emerging in vitro or ex vivo human/animal-derived pathophysiological models are further explored and highly recommended for use in NDC studies since they might mimic in vivo delivery features and fill the translational gaps from animals to humans. The exploration of modern microscopy techniques for precise nanoparticle (NP) tracking at the cellular, organ, and organismal levels informs the tailored development of NDCs for in vivo application and clinical translation. Overall, the review integrates the latest insights into smart nanosystem engineering, physiological models, imaging-based validation tools, all directed towards enhancing the precise and efficient intracellular delivery of NDCs.
Collapse
Affiliation(s)
- Qiaoxia Zhou
- Institute of Lung Health and Immunity (LHI), Helmholtz MunichComprehensive Pneumology Center (CPC‐M)Member of the German Center for Lung Research (DZL)85764MunichGermany
- Department of Forensic PathologyWest China School of Preclinical and Forensic MedicineSichuan UniversityNo. 17 Third Renmin Road NorthChengdu610041China
- Burning Rock BiotechBuilding 6, Phase 2, Standard Industrial Unit, No. 7 LuoXuan 4th Road, International Biotech IslandGuangzhou510300China
| | - Qiongliang Liu
- Institute of Lung Health and Immunity (LHI), Helmholtz MunichComprehensive Pneumology Center (CPC‐M)Member of the German Center for Lung Research (DZL)85764MunichGermany
- Department of Thoracic SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| | - Yan Wang
- Qingdao Central HospitalUniversity of Health and Rehabilitation Sciences (Qingdao Central Medical Group)Qingdao266042China
| | - Jie Chen
- Department of Respiratory MedicineNational Key Clinical SpecialtyBranch of National Clinical Research Center for Respiratory DiseaseXiangya HospitalCentral South UniversityChangshaHunan410008China
- Center of Respiratory MedicineXiangya HospitalCentral South UniversityChangshaHunan410008China
- Clinical Research Center for Respiratory Diseases in Hunan ProvinceChangshaHunan410008China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory DiseaseChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalChangshaHunan410008P. R. China
| | - Otmar Schmid
- Institute of Lung Health and Immunity (LHI), Helmholtz MunichComprehensive Pneumology Center (CPC‐M)Member of the German Center for Lung Research (DZL)85764MunichGermany
| | - Markus Rehberg
- Institute of Lung Health and Immunity (LHI), Helmholtz MunichComprehensive Pneumology Center (CPC‐M)Member of the German Center for Lung Research (DZL)85764MunichGermany
| | - Lin Yang
- Institute of Lung Health and Immunity (LHI), Helmholtz MunichComprehensive Pneumology Center (CPC‐M)Member of the German Center for Lung Research (DZL)85764MunichGermany
| |
Collapse
|
21
|
Anitha K, Chenchula S, Surendran V, Shvetank B, Ravula P, Milan R, Chikatipalli R, R P. Advancing cancer theranostics through biomimetics: A comprehensive review. Heliyon 2024; 10:e27692. [PMID: 38496894 PMCID: PMC10944277 DOI: 10.1016/j.heliyon.2024.e27692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 03/02/2024] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
Nanotheranostics, especially those employing biomimetic approaches, are of substantial interest for molecular imaging and cancer therapy. The incorporation of diagnostics and therapeutics, known as cancer theranostics, represents a promising strategy in modern oncology. Biomimetics, inspired by nature, offers a multidisciplinary avenue with potential in advancing cancer theranostics. This review comprehensively analyses recent progress in biomimetics-based cancer theranostics, emphasizing its role in overcoming current treatment challenges, with a focus on breast, prostate, and skin cancers. Biomimetic approaches have been explored to address multidrug resistance (MDR), emphasizing their role in immunotherapy and photothermal therapy. The specific areas covered include biomimetic drug delivery systems bypassing MDR mechanisms, biomimetic platforms for immune checkpoint blockade, immune cell modulation, and photothermal tumor ablation. Pretargeting techniques enhancing radiotherapeutic agent uptake are discussed, along with a comprehensive review of clinical trials of global nanotheranostics. This review delves into biomimetic materials, nanotechnology, and bioinspired strategies for cancer imaging, diagnosis, and targeted drug delivery. These include imaging probes, contrast agents, and biosensors for enhanced specificity and sensitivity. Biomimetic strategies for targeted drug delivery involve the design of nanoparticles, liposomes, and hydrogels for site-specific delivery and improved therapeutic efficacy. Overall, this current review provides valuable information for investigators, clinicians, and biomedical engineers, offering insights into the latest biomimetics applications in cancer theranostics. Leveraging biomimetics aims to revolutionize cancer diagnosis, treatment, and patient outcomes.
Collapse
Affiliation(s)
- Kuttiappan Anitha
- Department of Pharmacology, School of Pharmacy and Technology Management (SPTM), SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-University, Shirpur, 425405, India
| | - Santenna Chenchula
- Department of Clinical Pharmacology, All India Institute of Medical Sciences (AIIMS), Bhopal, 462020, Madhya Pradesh, India
| | - Vijayaraj Surendran
- Dr Kalam College of Pharmacy, Thanjavur District, Tamil Nadu, 614 623, India
| | - Bhatt Shvetank
- School of Health Sciences and Technology, Dr Vishwanath Karad MIT World Peace University, Pune, 411038, Maharashtra, India
| | - Parameswar Ravula
- Amity Institute of Pharmacy, Amity University Madhya Pradesh (AUMP), Gwalior, 474005, Madhya Pradesh, India
| | - Rhythm Milan
- Amity Institute of Pharmacy, Amity University Madhya Pradesh (AUMP), Gwalior, 474005, Madhya Pradesh, India
| | - Radhika Chikatipalli
- Sri Venkateshwara College of Pharmacy, Chittoor District, Andhra Pradesh, 517520, India
| | - Padmavathi R
- SVS Medical College, Mahbubnagar, Telangana, India
| |
Collapse
|
22
|
Jha A, Kumar M, Bharti K, Manjit M, Mishra B. Biopolymer-based tumor microenvironment-responsive nanomedicine for targeted cancer therapy. Nanomedicine (Lond) 2024; 19:633-651. [PMID: 38445583 DOI: 10.2217/nnm-2023-0302] [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] [Indexed: 03/07/2024] Open
Abstract
Nanomedicine has opened up new avenues for cancer treatment by enhancing drug solubility, permeability and targeted delivery to cancer cells. Despite its numerous advantages over conventional therapies, nanomedicine may exhibit off-target drug distribution, harming nontarget regions. The increased permeation and retention effect of nanomedicine in tumor sites also has its limitations, as abnormal tumor vasculature, dense stroma structure and altered tumor microenvironment (TME) may result in limited intratumor distribution and therapeutic failure. However, TME-responsive nanomedicine has exhibited immense potential for efficient, safe and precise delivery of therapeutics utilizing stimuli specific to the TME. This review discusses the mechanistic aspects of various TME-responsive biopolymers and their application in developing various types of TME-responsive nanomedicine.
Collapse
Affiliation(s)
- Abhishek Jha
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Manish Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Kanchan Bharti
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Manjit Manjit
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Brahmeshwar Mishra
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| |
Collapse
|
23
|
Chen Q, Fang C, Xia F, Wang Q, Li F, Ling D. Metal nanoparticles for cancer therapy: Precision targeting of DNA damage. Acta Pharm Sin B 2024; 14:1132-1149. [PMID: 38486992 PMCID: PMC10934341 DOI: 10.1016/j.apsb.2023.08.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/30/2023] [Accepted: 08/15/2023] [Indexed: 03/17/2024] Open
Abstract
Cancer, a complex and heterogeneous disease, arises from genomic instability. Currently, DNA damage-based cancer treatments, including radiotherapy and chemotherapy, are employed in clinical practice. However, the efficacy and safety of these therapies are constrained by various factors, limiting their ability to meet current clinical demands. Metal nanoparticles present promising avenues for enhancing each critical aspect of DNA damage-based cancer therapy. Their customizable physicochemical properties enable the development of targeted and personalized treatment platforms. In this review, we delve into the design principles and optimization strategies of metal nanoparticles. We shed light on the limitations of DNA damage-based therapy while highlighting the diverse strategies made possible by metal nanoparticles. These encompass targeted drug delivery, inhibition of DNA repair mechanisms, induction of cell death, and the cascading immune response. Moreover, we explore the pivotal role of physicochemical factors such as nanoparticle size, stimuli-responsiveness, and surface modification in shaping metal nanoparticle platforms. Finally, we present insights into the challenges and future directions of metal nanoparticles in advancing DNA damage-based cancer therapy, paving the way for novel treatment paradigms.
Collapse
Affiliation(s)
- Qian Chen
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chunyan Fang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fan Xia
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiyue Wang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai 201203, China
| | - Fangyuan Li
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai 201203, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou 310009, China
| | - Daishun Ling
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai 201203, China
| |
Collapse
|
24
|
Wang J, Fang Z, Zhao C, Sun Z, Gao S, Zhang B, Qiu D, Yang M, Sheng F, Gao S, Hou Y. Intelligent Size-Switchable Iron Carbide-Based Nanocapsules with Cascade Delivery Capacity for Hyperthermia-Enhanced Deep Tumor Ferroptosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307006. [PMID: 37924225 DOI: 10.1002/adma.202307006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/31/2023] [Indexed: 11/06/2023]
Abstract
The ferroptosis pathway is recognized as an essential strategy for tumor treatment. However, killing tumor cells in deep tumor regions with ferroptosis agents is still challenging because of distinct size requirements for intratumoral accumulation and deep tumor penetration. Herein, intelligent nanocapsules with size-switchable capability that responds to acid/hyperthermia stimulation to achieve deep tumor ferroptosis are developed. These nanocapsules are constructed using poly(lactic-co-glycolic) acid and Pluronic F127 as carrier materials, with Au-Fe2 C Janus nanoparticles serving as photothermal and ferroptosis agents, and sorafenib (SRF) as the ferroptosis enhancer. The PFP@Au-Fe2 C-SRF nanocapsules, designed with an appropriate size, exhibit superior intratumoral accumulation compared to free Au-Fe2 C nanoparticles, as evidenced by photoacoustic and magnetic resonance imaging. These nanocapsules can degrade within the acidic tumor microenvironment when subjected to laser irradiation, releasing free Au-Fe2 C nanoparticles. This enables them to penetrate deep into tumor regions and disrupt intracellular redox balance. Under the guidance of imaging, these PFP@Au-Fe2 C-SRF nanocapsules effectively inhibit tumor growth when exposed to laser irradiation, capitalizing on the synergistic photothermal and ferroptosis effects. This study presents an intelligent formulation based on iron carbide for achieving deep tumor ferroptosis through size-switchable cascade delivery, thereby advancing the comprehension of ferroptosis in the context of tumor theranostics.
Collapse
Affiliation(s)
- Jingjing Wang
- Beijing Key Laboratory for Magnetoelectric Materials and Devices (BKL-MMD), School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Zhi Fang
- Beijing Key Laboratory for Magnetoelectric Materials and Devices (BKL-MMD), School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Chenyang Zhao
- Department of Ultrasound, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhaoli Sun
- Beijing Key Laboratory for Magnetoelectric Materials and Devices (BKL-MMD), School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Shen Gao
- Department of Radiology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Biao Zhang
- Beijing Key Laboratory for Magnetoelectric Materials and Devices (BKL-MMD), School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Daping Qiu
- Beijing Key Laboratory for Magnetoelectric Materials and Devices (BKL-MMD), School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Meng Yang
- Department of Ultrasound, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Fugeng Sheng
- Department of Radiology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Song Gao
- Institute of Spin-X Science and Technology, South China University of Technology, Guangzhou, 510641, China
| | - Yanglong Hou
- Beijing Key Laboratory for Magnetoelectric Materials and Devices (BKL-MMD), School of Materials Science and Engineering, Peking University, Beijing, 100871, China
- School of Materials, Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
25
|
Ko MJ, Yoo W, Min S, Zhang YS, Joo J, Kang H, Kim DH. Photonic control of image-guided ferroptosis cancer nanomedicine. Coord Chem Rev 2024; 500:215532. [PMID: 38645709 PMCID: PMC11027759 DOI: 10.1016/j.ccr.2023.215532] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Photonic nanomaterials, characterized by their remarkable photonic tunability, empower a diverse range of applications, including cutting-edge advances in cancer nanomedicine. Recently, ferroptosis has emerged as a promising alternative strategy for effectively killing cancer cells with minimizing therapeutic resistance. Novel design of photonic nanomaterials that can integrate photoresponsive-ferroptosis inducers, -diagnostic imaging, and -synergistic components provide significant benefits to effectively trigger local ferroptosis. This review provides a comprehensive overview of recent advancements in photonic nanomaterials for image-guided ferroptosis cancer nanomedicine, offering insights into their strengths, constraints, and their potential as a future paradigm in cancer treatment.
Collapse
Affiliation(s)
- Min Jun Ko
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Woojung Yoo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital Harvard Medical School, Cambridge, MA 02139, USA
| | - Jinmyoung Joo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
- College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
- Department of Biomedical Engineering, University of Illinois, Chicago, IL 60607, USA
| |
Collapse
|
26
|
Shen X, Pan D, Gong Q, Gu Z, Luo K. Enhancing drug penetration in solid tumors via nanomedicine: Evaluation models, strategies and perspectives. Bioact Mater 2024; 32:445-472. [PMID: 37965242 PMCID: PMC10641097 DOI: 10.1016/j.bioactmat.2023.10.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
Effective tumor treatment depends on optimizing drug penetration and accumulation in tumor tissue while minimizing systemic toxicity. Nanomedicine has emerged as a key solution that addresses the rapid clearance of free drugs, but achieving deep drug penetration into solid tumors remains elusive. This review discusses various strategies to enhance drug penetration, including manipulation of the tumor microenvironment, exploitation of both external and internal stimuli, pioneering nanocarrier surface engineering, and development of innovative tactics for active tumor penetration. One outstanding strategy is organelle-affinitive transfer, which exploits the unique properties of specific tumor cell organelles and heralds a potentially transformative approach to active transcellular transfer for deep tumor penetration. Rigorous models are essential to evaluate the efficacy of these strategies. The patient-derived xenograft (PDX) model is gaining traction as a bridge between laboratory discovery and clinical application. However, the journey from bench to bedside for nanomedicines is fraught with challenges. Future efforts should prioritize deepening our understanding of nanoparticle-tumor interactions, re-evaluating the EPR effect, and exploring novel nanoparticle transport mechanisms.
Collapse
Affiliation(s)
- Xiaoding Shen
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Dayi Pan
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, 361021, China
| | - Zhongwei Gu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
27
|
赵 甜, 吴 昊, 陈 世, 王 佳, 刘 贻, 李 亭. [Research Progress on the Influence of Tumor Extracellular Matrix Mechanic Properties on Nanodrug Delivery]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:13-18. [PMID: 38322528 PMCID: PMC10839498 DOI: 10.12182/20240160205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Indexed: 02/08/2024]
Abstract
Nanodrugs are widely utilized in the biomedical fields, exhibiting immense potential in cancer therapy in particular. However, tumors exist in an extremely complicated microenvironment where substances like collagen are continuously deposited and remodeled, leading to significant alterations in the mechanical properties of the extracellular matrix (ECM) during tumor development. Previous research has primarily focused on the specific physicochemical properties of nanodrugs, such as particle size, electric charge, shape, surface chemistry, etc., and their effects on cellular uptake, cytotoxicity, and in vivo pharmacokinetics. Limited studies have been done to explore the impact of ECM mechanical properties on nanodrug delivery. In this review, we systematically summarized the relevant research findings on this topic from the perspective of the characteristics and testing methods of tumor ECM mechanics. Additionally, we made a thorough discussion of the potential mechanical and biological mechanisms involved in nanodrug delivery. We proposed several noteworthy research directions. Regarding the overall strategy, there is a need to emphasize targeted delivery that combines ECM mechanics and nanomechanics to achieve precise drug delivery. Regarding the spatial aspect, attention should be given to the nonlinear spatial mechanical heterogeneity within the interior of solid tumors and the construction of mechanic microenvironment-adaptive nanocarriers to improve the delivery efficiency. Regarding the temporal aspect, emphasis should be placed on the dynamic development and changes in the mechanical microenvironment during solid tumor growth and treatment processes. Based on the stromal mechanical characteristics of the tumor tissues of individual patients, personalized treatment strategies can be formulated, which will enhance treatment specificity and efficacy. In addition, issues such as mechanically targeted nanodrug delivery, degradation, and metabolism under dynamic ECM mechanical conditions warrant further investigation.
Collapse
Affiliation(s)
- 甜 赵
- 电子科技大学生命科学与技术学院 (成都 610054)School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - 昊 吴
- 电子科技大学生命科学与技术学院 (成都 610054)School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - 世桓 陈
- 电子科技大学生命科学与技术学院 (成都 610054)School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - 佳雯 王
- 电子科技大学生命科学与技术学院 (成都 610054)School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - 贻尧 刘
- 电子科技大学生命科学与技术学院 (成都 610054)School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - 亭亭 李
- 电子科技大学生命科学与技术学院 (成都 610054)School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| |
Collapse
|
28
|
Liu S, Sun Y, Ye J, Li C, Wang Q, Liu M, Cui Y, Wang C, Jin G, Fu Y, Xu J, Liang X. Targeted Delivery of Active Sites by Oxygen Vacancy-Engineered Bimetal Silicate Nanozymes for Intratumoral Aggregation-Potentiated Catalytic Therapy. ACS NANO 2024; 18:1516-1530. [PMID: 38172073 DOI: 10.1021/acsnano.3c08780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Biodegradable silicate nanoconstructs have aroused tremendous interest in cancer therapeutics due to their variable framework composition and versatile functions. Nevertheless, low intratumoral retention still limits their practical application. In this study, oxygen vacancy (OV)-enriched bimetallic silicate nanozymes with Fe-Ca dual active sites via modification of oxidized sodium alginate and gallic acid (GA) loading (OFeCaSA-V@GA) were developed for targeted aggregation-potentiated therapy. The band gap of silica markedly decreased from 2.76 to 1.81 eV by codoping of Fe3+ and Ca2+, enabling its excitation by a 650 nm laser to generate reactive oxygen species. The OV that occurred in the hydrothermal synthetic stage of OFeCaSA-V@GA can anchor the metal ions to form an atomic phase, offering a massive fabrication method of single-atom nanozymes. Density functional theory results reveal that the Ca sites can promote the adsorption of H2O2, and Fe sites can accelerate the dissociation of H2O2, thereby realizing a synergetic catalytic effect. More importantly, the targeted delivery of metal ions can induce a morphological transformation at tumor sites, leading to high retention (the highest retention rate is 36.3%) of theranostic components in tumor cells. Thus, this finding may offer an ingenious protocol for designing and engineering highly efficient and long-retention nanodrugs.
Collapse
Affiliation(s)
- Shuang Liu
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, P. R. China
- Guangxi Medical University Cancer Hospital, Nanning, 530021, P. R. China
| | - Yu Sun
- Heilongjiang Vocational Institute Ecological Engineering, Harbin, 150040, P. R. China
| | - Jin Ye
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, P. R. China
| | - Chunsheng Li
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, P. R. China
| | - Qiang Wang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, P. R. China
| | - Mengting Liu
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, P. R. China
| | - Yujie Cui
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, P. R. China
| | - Chen Wang
- Guangxi Medical University Cancer Hospital, Nanning, 530021, P. R. China
| | - Guanqiao Jin
- Guangxi Medical University Cancer Hospital, Nanning, 530021, P. R. China
| | - Yujie Fu
- College of Forestry, Beijing Forestry University, Beijing, 100083, P. R. China
| | - Jiating Xu
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, P. R. China
- Heilongjiang Provincial Key Laboratory of Ecological Utilization of Forestry-Based Active Substances, Northeast Forestry University, Harbin, 150040, P. R. China
| | - Xinqiang Liang
- Guangxi Medical University Cancer Hospital, Nanning, 530021, P. R. China
| |
Collapse
|
29
|
Wu D, Huang Q, Sha S, Xue F, Huang G, Tian Q. Engineering of copper sulfide mediated by phototherapy performance. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1932. [PMID: 37853634 DOI: 10.1002/wnan.1932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/04/2023] [Accepted: 09/20/2023] [Indexed: 10/20/2023]
Abstract
Copper sulfide based phototherapy, including photothermal therapy and photodynamic therapy, is an emerging minimally invasive treatment of tumor, which the light was converted to heat or reactive oxygen to kill the tumor cells. Compared with conventional chemotherapy and radiation therapy, Cu2-x S based phototherapy is more efficient and has fewer side effects. However, considering the dose-dependent toxicity of Cu2-x S, the performance of Cu2-x S based phototherapy still cannot meet the requirement of the clinical application to now. To overcome this limitation, engineering of Cu2-x S to improve the phototherapy performance by increasing light absorption has attracted extensive attention. For better guidance of Cu2-x S engineering, we outline the currently engineering method being explored, including (1) structural engineering, (2) compositional engineering, (3) functional engineering, and (4) performance engineering. Also, the relationship between the engineering method and phototherapy performance was discussed in this review. In addition, the further development of Cu2-x S based phototherapy is prospected, including smart materials based phototherapy, phototherapy induced immune microenvironment modulation et al. This review will provide new ideas and opportunities for engineering of Cu2-x S with better phototherapy performance. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Dan Wu
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Qingqing Huang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Shuang Sha
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Fengfeng Xue
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Qiwei Tian
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
30
|
Elsafy S, Metselaar J, Lammers T. Nanomedicine - Immune System Interactions: Limitations and Opportunities for the Treatment of Cancer. Handb Exp Pharmacol 2024; 284:231-265. [PMID: 37578622 DOI: 10.1007/164_2023_685] [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] [Indexed: 08/15/2023]
Abstract
Nanoparticles interact with immune cells in many different ways. These interactions are crucially important for determining nanoparticles' ability to be used for cancer therapy. Traditionally, strategies such as PEGylation have been employed to reduce (the kinetics of) nanoparticle uptake by immune cells, to endow them with long circulation properties, and to enable them to exploit the Enhanced Permeability and Retention (EPR) effect to accumulate in tumors. More recently, with immunotherapy becoming an increasingly important cornerstone in the clinical management of cancer, ever more research efforts in academia and industry are focusing on specifically targeting immune cells with nanoparticles. In this chapter, we describe the barriers and opportunities of immune cell targeting with nanoparticles, and we discuss how nanoparticle-based drug delivery to specific immune cell populations in tumors as well as in secondary myeloid and lymphoid organs (such as bone marrow, lymph nodes, and spleen) can be leveraged to boost the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Sara Elsafy
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), Center for Biohybrid Medical Systems (CBMS), University Hospital RWTH Aachen, Aachen, Germany
| | - Josbert Metselaar
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), Center for Biohybrid Medical Systems (CBMS), University Hospital RWTH Aachen, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), Center for Biohybrid Medical Systems (CBMS), University Hospital RWTH Aachen, Aachen, Germany.
| |
Collapse
|
31
|
Saeed W, Shahbaz E, Maqsood Q, Ali SW, Mahnoor M. Cutaneous Oncology: Strategies for Melanoma Prevention, Diagnosis, and Therapy. Cancer Control 2024; 31:10732748241274978. [PMID: 39133519 PMCID: PMC11320697 DOI: 10.1177/10732748241274978] [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: 04/21/2024] [Revised: 07/11/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024] Open
Abstract
Skin cancer comprises one-third of all diagnosed cancer cases and remains a major health concern. Genetic and environmental parameters serve as the two main risk factors associated with the development of skin cancer, with ultraviolet radiation being the most common environmental risk factor. Studies have also found fair complexion, arsenic toxicity, indoor tanning, and family history among the prevailing causes of skin cancer. Prevention and early diagnosis play a crucial role in reducing the frequency and ensuring effective management of skin cancer. Recent studies have focused on exploring minimally invasive or non-invasive diagnostic technologies along with artificial intelligence to facilitate rapid and accurate diagnosis. The treatment of skin cancer ranges from traditional surgical excision to various advanced methods such as phototherapy, radiotherapy, immunotherapy, targeted therapy, and combination therapy. Recent studies have focused on immunotherapy, with the introduction of new checkpoint inhibitors and personalized immunotherapy enhancing treatment efficacy. Advancements in multi-omics, nanotechnology, and artificial intelligence have further deepened the understanding of the mechanisms underlying tumoral growth and their interaction with therapeutic effects, which has paved the way for precision oncology. This review aims to highlight the recent advancements in the understanding and management of skin cancer, and provide an overview of existing and emerging diagnostic, prognostic, and therapeutic modalities, while highlighting areas that require further research to bridge the existing knowledge gaps.
Collapse
Affiliation(s)
- Wajeeha Saeed
- Department of Food Sciences, Faculty of Agricultural Sciences, University of the Punjab, Lahore, Pakistan
| | - Esha Shahbaz
- Department of Food Sciences, Faculty of Agricultural Sciences, University of the Punjab, Lahore, Pakistan
| | - Quratulain Maqsood
- Centre for Applied Molecular Biology, University of the Punjab, Lahore Pakistan
| | - Shinawar Waseem Ali
- Department of Food Sciences, Faculty of Agricultural Sciences, University of the Punjab, Lahore, Pakistan
| | - Muhammada Mahnoor
- Sehat Medical Complex Lake City, University of Lahore, Lahore Pakistan
| |
Collapse
|
32
|
Xu H, Guo Z, Li M, Chaves HV, Pinto VDPT, Filho GC, Du M, Bezerra MM. Copper-Based Nanomaterials for Image-Guided Cancer Therapy. BIO INTEGRATION 2024; 5. [DOI: 10.15212/bioi-2024-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
Abstract
Cancer is a significant disease that poses a major threat to human health. Image-guided cancer therapy refers to a series of medical procedures that use imaging technology to precisely locate and treat cancer. Combining the dual characteristics of medical images and functional nanomaterial (NM) drug carriers, various integrated diagnosis and treatment probes have been developed for in vivo dynamic monitoring and therapeutic effect evaluation of drugs based on medical imaging. Copper (Cu)-based NMs have emerged as valuable products of nanotechnology due to their unique physicochemical properties, which are influenced by factors, such as size, shape, and surface properties. In the field of imaging, Cu-based NMs offer a combination of desirable characteristics, including fluorescence emission, contrast enhancement, and radiolabeling stability. These properties form the foundation for a wide range of imaging modalities. In addition, Cu-based NMs can be used as a carrier for diagnostic or therapeutic drugs and the synergistic effect of multiple therapeutic modalities can be realized by doping multiple transition metals into the heterostructures. These properties have become an important basis for imaging-guided therapy with Cu-based NMs. In this review we introduce biocompatible Cu-based NMs for image-guided cancer therapy and provide an overview of the promising outcomes in biomedical research.
Collapse
|
33
|
Mianowska M, Zaremba-Czogalla M, Zygmunt A, Mahmud M, Süss R, Gubernator J. Dual Role of Vitamin C-Encapsulated Liposomal Berberine in Effective Colon Anticancer Immunotherapy. Pharmaceuticals (Basel) 2023; 17:5. [PMID: 38275991 PMCID: PMC10819181 DOI: 10.3390/ph17010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/01/2023] [Accepted: 12/13/2023] [Indexed: 01/27/2024] Open
Abstract
The aim of the study was to achieve effective colon anticancer immunotherapy using the alkaloid berberine. In the presented paper we attempt to develop a formulation of berberine loaded into liposomal carriers using the vitamin C gradient method, characterized by efficient drug encapsulation, high stability during long-term storage, low drug release in human plasma with specific cytotoxicity towards colon cancer cells. Liposomal berberine was responsible for the induction of oxidative stress, the presence of Ca2+ ions in the cytosol, the reduction of Δψm, and ATP depletion with a simultaneous lack of caspase activity. Moreover, treatment with liposomal berberine led to CRT exposure on the surface of cancer cells, extracellular ATP, and HMGB1 release. The above-described mechanism of action was most likely associated with ICD induction, contributing to the increased number of phagocytic cancer cells. We have shown that cancer cells treated with liposomal berberine were phagocytosed more frequently by macrophages compared to the untreated cancer cells. What is more, we have shown that macrophage pre-treatment with liposomal berberine led to a 3-fold change in the number of phagocytosed SW620 cancer cells. The obtained results provide new insights into the role of berberine in maintaining the immune response against colorectal cancer.
Collapse
Affiliation(s)
- Martyna Mianowska
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (M.Z.-C.); (A.Z.); (M.M.)
| | - Magdalena Zaremba-Czogalla
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (M.Z.-C.); (A.Z.); (M.M.)
| | - Adrianna Zygmunt
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (M.Z.-C.); (A.Z.); (M.M.)
| | - Mohamed Mahmud
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (M.Z.-C.); (A.Z.); (M.M.)
- Department of Medical Genetics, Faculty of Health Sciences, University of Misurata, Misurata 2478, Libya
| | - Regine Süss
- Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Albert Ludwig University of Freiburg, Sonnenstr. 5, 79104 Freiburg, Germany;
| | - Jerzy Gubernator
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland; (M.Z.-C.); (A.Z.); (M.M.)
| |
Collapse
|
34
|
Badparvar F, Marjani AP, Salehi R, Ramezani F. pH/redox responsive size-switchable intelligent nanovehicle for tumor microenvironment targeted DOX release. Sci Rep 2023; 13:22475. [PMID: 38110480 PMCID: PMC10728153 DOI: 10.1038/s41598-023-49446-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: 08/10/2023] [Accepted: 12/08/2023] [Indexed: 12/20/2023] Open
Abstract
Tumor microenvironment (TME) targeted strategy could control the drug release in tumor cells more accurately and creates a new opportunity for enhanced site-specific targeted delivery. In this study, (PAA-b-PCL-S-S-PCL-b-PAA) copolymeric nanoparticles (NPs) with size-switchable ability and dual pH/redox-triggered drug release behavior were designed to significantly promote cancer uptake (cell internalization of around 100% at 30 min) and site-specific targeted doxorubicin (DOX) delivery in MDA-MB-231 tumor cells. NPs surface charge was shifted from - 17.8 to - 2.4 and their size shrunk from 170.3 to 93 nm in TME. The cell cycle results showed that DOX-loaded NPs showed G2/M (68%) arrest, while free DOX showed sub-G1 arrest (22%). Apoptosis tests confirmed that the cells treated with DOX-loaded NPs showed a higher amount of apoptosis (71.6%) than the free DOX (49.8%). Western blot and RT-PCR assays revealed that the apoptotic genes and protein levels were significantly upregulated using the DOX-loaded NPs vs. the free DOX (Pvalue < 0.001). In conclusion, dual pH/redox-responsive and size-switchable DOX-loaded NPs developed here showed outstanding anti-tumoral features compared with free DOX that might present a prospective platform for tumor site-specific accumulation and drug release that suggest further in vivo research.
Collapse
Affiliation(s)
- Fahimeh Badparvar
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia, Iran
| | | | - Roya Salehi
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Fatemeh Ramezani
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
35
|
Bauri S, Tripathi S, Choudhury AM, Mandal SS, Raj H, Maiti P. Nanomaterials as Theranostic Agents for Cancer Therapy. ACS APPLIED NANO MATERIALS 2023; 6:21462-21495. [DOI: 10.1021/acsanm.3c04235] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Affiliation(s)
- Sudepta Bauri
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Swikriti Tripathi
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Avishek Mallick Choudhury
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Subham Sekhar Mandal
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Hans Raj
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Pralay Maiti
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| |
Collapse
|
36
|
Li X, Gao Y, Li H, Majoral JP, Shi X, Pich A. Smart and bioinspired systems for overcoming biological barriers and enhancing disease theranostics. PROGRESS IN MATERIALS SCIENCE 2023; 140:101170. [DOI: 10.1016/j.pmatsci.2023.101170] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
37
|
Niu B, Wu Y, Zhou M, Lin R, Ge P, Chen X, Zhou H, Zhang X, Xie J. Precise delivery of celastrol by PEGylated aptamer dendrimer nanoconjugates for enormous therapeutic effect via superior intratumor penetration over antibody counterparts. Cancer Lett 2023; 579:216461. [PMID: 37898358 DOI: 10.1016/j.canlet.2023.216461] [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: 06/22/2023] [Revised: 10/05/2023] [Accepted: 10/17/2023] [Indexed: 10/30/2023]
Abstract
Antibody-coated nanoparticles have been reported to have the extremely low delivery efficiency in solid tumors in preclinical trials. Though aptamers were considered to be superior over antibodies in cancer theranostics, whether PEGylated aptamer nanoparticles are better than antibody nanoparticles in improving delivery specificity and penetration efficiency of chemotherapeutics is still unknown. Here, we conjugate celastrol, a natural product with anti-tumor effect, onto PEGylated EpCAM aptamer or antibody dendrimers to obtain two nanoconjugates, and for the first time, conduct a comprehensive study to compare their performance in delivery specificity, intratumoral penetration ability and therapeutic outcomes. Our results showed that compared to antibody counterparts, PEGylated aptamer nanoconjugates exhibited the enhanced accumulation and retention specificities at tumor sites and the stronger intratumoral penetration capabilities by reducing the macrophage reservoir effects in solid tumors. When delivered celastrol to a colorectal xenograft tumor mice model by PEGylated aptamer dendrimers, 20 % of enhanced therapeutic efficiency was achieved compared to that by antibody-modified ones. Moreover, celastrol at 2 mg/kg delivered by PEGylated aptamer dendrimers showed the prominent anticancer efficiency (nearly 92 %) but without obvious side effects. These data firstly provide the proof-of-concept implementation that PEGylated aptamer nanoconjugates will display the great potential in the effective and safe cancer treatment with regard to the superiority over antibody ones in penetration abilities.
Collapse
Affiliation(s)
- Boning Niu
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China; Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuehuang Wu
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Min Zhou
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Ruimiao Lin
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Pengjin Ge
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Xiaohui Chen
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Hu Zhou
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China.
| | - Xiaokun Zhang
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Jingjing Xie
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China.
| |
Collapse
|
38
|
Guo L, Yang J, Wang H, Yi Y. Multistage Self-Assembled Nanomaterials for Cancer Immunotherapy. Molecules 2023; 28:7750. [PMID: 38067480 PMCID: PMC10707962 DOI: 10.3390/molecules28237750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Advances in nanotechnology have brought innovations to cancer therapy. Nanoparticle-based anticancer drugs have achieved great success from bench to bedside. However, insufficient therapy efficacy due to various physiological barriers in the body remains a key challenge. To overcome these biological barriers and improve the therapeutic efficacy of cancers, multistage self-assembled nanomaterials with advantages of stimuli-responsiveness, programmable delivery, and immune modulations provide great opportunities. In this review, we describe the typical biological barriers for nanomedicines, discuss the recent achievements of multistage self-assembled nanomaterials for stimuli-responsive drug delivery, highlighting the programmable delivery nanomaterials, in situ transformable self-assembled nanomaterials, and immune-reprogramming nanomaterials. Ultimately, we perspective the future opportunities and challenges of multistage self-assembled nanomaterials for cancer immunotherapy.
Collapse
Affiliation(s)
- Lamei Guo
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, 391 Binshui Xidao, Xiqing District, Tianjin 300384, China; (L.G.); (J.Y.)
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| | - Jinjun Yang
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, 391 Binshui Xidao, Xiqing District, Tianjin 300384, China; (L.G.); (J.Y.)
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| | - Yu Yi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| |
Collapse
|
39
|
Liu Z, Ji P, Liu H, Yu L, Zhang SM, Liu P, Zhang XZ, Luo GF, Shang Z. FNIII14 Peptide-Enriched Membrane Nanocarrier to Disrupt Stromal Barriers through Reversing CAFs for Augmenting Drug Penetration in Tumors. NANO LETTERS 2023; 23:9963-9971. [PMID: 37729438 DOI: 10.1021/acs.nanolett.3c02983] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Given the key roles of cancer associated fibroblasts (CAFs) in shaping tumor stroma, this study shows a CAF-associated ITGB1-inactivating peptide-enriched membrane nanodelivery system (designated as PMNPs-D) to simultaneously target CAFs and tumor cells for boosted chemotherapy through promoted drug perfusion. In the structure of PMNPs-D, the PLGA-based inner core is loaded with the chemotherapeutic drug doxorubicin, and the outer surface is cloaked by hybrid biomembranes with the insertion of integrin β1 (ITGB1) inhibiting peptide (i.e., FNIII14). After prolonged blood circulation and actively targeting in tumor sites, PMNPs-D can respond to CAF-overexpressed fibroblast activation protein-α (FAP-α) to trigger the release of FNIII14, which will bind to ITGB1 and inhibit CAFs' biological function in producing the stromal matrix, thereby loosening the condensed stromal structure and enhancing the permeability of nanotherapeutics in tumors. As a result, this tailor-designed nanosystem shows substantial tumor inhibition and metastasis retardation in aggressive adenoid cystic carcinoma (ACC) tumor-harboring mice.
Collapse
Affiliation(s)
- Zhenan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Ping Ji
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Hanzhe Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Lili Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Shi-Man Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Pan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Guo-Feng Luo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| |
Collapse
|
40
|
Doustmihan A, Fathi M, Mazloomi M, Salemi A, Hamblin MR, Jahanban-Esfahlan R. Molecular targets, therapeutic agents and multitasking nanoparticles to deal with cancer stem cells: A narrative review. J Control Release 2023; 363:57-83. [PMID: 37739017 DOI: 10.1016/j.jconrel.2023.09.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 09/08/2023] [Accepted: 09/17/2023] [Indexed: 09/24/2023]
Abstract
There is increasing evidence that malignant tumors are initiated and maintained by a sub-population of tumor cells that have similar biological properties to normal adult stem cells. This very small population of Cancer Stem Cells (CSC) comprises tumor initiating cells responsible for cancer recurrence, drug resistance and metastasis. Conventional treatments such as chemotherapy, radiotherapy and surgery, in addition to being potentially toxic and non-specific, may paradoxically increase the population, spread and survival of CSCs. Next-generation sequencing and omics technologies are increasing our understanding of the pathways and factors involved in the development of CSCs, and can help to discover new therapeutic targets against CSCs. In addition, recent advances in nanomedicine have provided hope for the development of optimal specific therapies to eradicate CSCs. Moreover, the use of artificial intelligence and nano-informatics can elucidate new drug targets, and help to design drugs and nanoparticles (NPs) to deal with CSCs. In this review, we first summarize the properties of CSCs and describe the signaling pathways and molecular characteristics responsible for the emergence and survival of CSCs. Also, the location of CSCs within the tumor and the effect of host factors on the creation and maintenance of CSCs are discussed. Newly discovered molecular targets involved in cancer stemness and some novel therapeutic compounds to combat CSCs are highlighted. The optimum properties of anti-CSC NPs, including blood circulation and stability, tumor accumulation and penetration, cellular internalization, drug release, endosomal escape, and aptamers designed for specific targeting of CSCs are covered. Finally, some recent smart NPs designed for therapeutic and theranostic purposes to overcome CSCs are discussed.
Collapse
Affiliation(s)
- Abolfazl Doustmihan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marziyeh Fathi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - MirAhmad Mazloomi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysan Salemi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa.
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
41
|
Kon E, Ad-El N, Hazan-Halevy I, Stotsky-Oterin L, Peer D. Targeting cancer with mRNA-lipid nanoparticles: key considerations and future prospects. Nat Rev Clin Oncol 2023; 20:739-754. [PMID: 37587254 DOI: 10.1038/s41571-023-00811-9] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2023] [Indexed: 08/18/2023]
Abstract
Harnessing mRNA-lipid nanoparticles (LNPs) to treat patients with cancer has been an ongoing research area that started before these versatile nanoparticles were successfully used as COVID-19 vaccines. Currently, efforts are underway to harness this platform for oncology therapeutics, mainly focusing on cancer vaccines targeting multiple neoantigens or direct intratumoural injections of mRNA-LNPs encoding pro-inflammatory cytokines. In this Review, we describe the opportunities of using mRNA-LNPs in oncology applications and discuss the challenges for successfully translating the findings of preclinical studies of these nanoparticles into the clinic. We critically appraise the potential of various mRNA-LNP targeting and delivery strategies, considering physiological, technological and manufacturing challenges. We explore these approaches in the context of the potential clinical applications best suited to each approach and highlight the obstacles that currently need to be addressed to achieve these applications. Finally, we provide insights from preclinical and clinical studies that are leading to this powerful platform being considered the next frontier in oncology treatment.
Collapse
Affiliation(s)
- Edo Kon
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Nitay Ad-El
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Inbal Hazan-Halevy
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Lior Stotsky-Oterin
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel.
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
42
|
Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, Tang J, Zhou Z, Shen Y. Tumor Abnormality-Oriented Nanomedicine Design. Chem Rev 2023; 123:10920-10989. [PMID: 37713432 DOI: 10.1021/acs.chemrev.3c00062] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.
Collapse
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yechun Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
43
|
Kiaie SH, Salehi-Shadkami H, Sanaei MJ, Azizi M, Shokrollahi Barough M, Nasr MS, Sheibani M. Nano-immunotherapy: overcoming delivery challenge of immune checkpoint therapy. J Nanobiotechnology 2023; 21:339. [PMID: 37735656 PMCID: PMC10512572 DOI: 10.1186/s12951-023-02083-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/26/2023] [Indexed: 09/23/2023] Open
Abstract
Immune checkpoint (ICP) molecules expressed on tumor cells can suppress immune responses against tumors. ICP therapy promotes anti-tumor immune responses by targeting inhibitory and stimulatory pathways of immune cells like T cells and dendritic cells (DC). The investigation into the combination therapies through novel immune checkpoint inhibitors (ICIs) has been limited due to immune-related adverse events (irAEs), low response rate, and lack of optimal strategy for combinatorial cancer immunotherapy (IMT). Nanoparticles (NPs) have emerged as powerful tools to promote multidisciplinary cooperation. The feasibility and efficacy of targeted delivery of ICIs using NPs overcome the primary barrier, improve therapeutic efficacy, and provide a rationale for more clinical investigations. Likewise, NPs can conjugate or encapsulate ICIs, including antibodies, RNAs, and small molecule inhibitors. Therefore, combining the drug delivery system (DDS) with ICP therapy could provide a profitable immunotherapeutic strategy for cancer treatment. This article reviews the significant NPs with controlled DDS using current data from clinical and pre-clinical trials on mono- and combination IMT to overcome ICP therapeutic limitations.
Collapse
Affiliation(s)
- Seyed Hossein Kiaie
- Department of Formulation Development, ReNAP Therapeutics, Tehran, Iran.
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Hossein Salehi-Shadkami
- Department of Formulation Development, ReNAP Therapeutics, Tehran, Iran
- Department of Medical Science, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Sanaei
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, 8815713471, Iran
| | - Marzieh Azizi
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | | | - Mohammad Sadegh Nasr
- Department of Computer Science and Engineering Multi-Interprofessional Center for Health Informatics (MICHI), The University of Texas at Arlington, Arlington, TX, USA
| | - Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Razi Drug Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
44
|
Zheng S, Li G, Shi J, Liu X, Li M, He Z, Tian C, Kamei KI. Emerging platinum(IV) prodrug nanotherapeutics: A new epoch for platinum-based cancer therapy. J Control Release 2023; 361:819-846. [PMID: 37597809 DOI: 10.1016/j.jconrel.2023.08.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
Owing to the unique DNA damaging cytotoxicity, platinum (Pt)-based chemotherapy has long been the first-line choice for clinical oncology. Unfortunately, Pt drugs are restricted by the severe dose-dependent toxicity and drug resistance. Correspondingly, Pt(IV) prodrugs are developed with the aim to improve the antitumor performance of Pt drugs. However, as "free" molecules, Pt(IV) prodrugs are still subject to unsatisfactory in vivo destiny and antitumor efficacy. Recently, Pt(IV) prodrug nanotherapeutics, inheriting both the merits of Pt(IV) prodrugs and nanotherapeutics, have emerged and demonstrated the promise to address the underexploited dilemma of Pt-based cancer therapy. Herein, we summarize the latest fronts of emerging Pt(IV) prodrug nanotherapeutics. First, the basic outlines of Pt(IV) prodrug nanotherapeutics are overviewed. Afterwards, how versatile Pt(IV) prodrug nanotherapeutics overcome the multiple biological barriers of antitumor drug delivery is introduced in detail. Moreover, advanced combination therapies based on multimodal Pt(IV) prodrug nanotherapeutics are discussed with special emphasis on the synergistic mechanisms. Finally, prospects and challenges of Pt(IV) prodrug nanotherapeutics for future clinical translation are spotlighted.
Collapse
Affiliation(s)
- Shunzhe Zheng
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Guanting Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jianbin Shi
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinying Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Meng Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chutong Tian
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, Hangzhou 310058, China.
| | - Ken-Ichiro Kamei
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
45
|
Lu Q, Yu H, Zhao T, Zhu G, Li X. Nanoparticles with transformable physicochemical properties for overcoming biological barriers. NANOSCALE 2023; 15:13202-13223. [PMID: 37526946 DOI: 10.1039/d3nr01332d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
In recent years, tremendous progress has been made in the development of nanomedicines for advanced therapeutics, yet their unsatisfactory targeting ability hinders the further application of nanomedicines. Nanomaterials undergo a series of processes, from intravenous injection to precise delivery at target sites. Each process faces different or even contradictory requirements for nanoparticles to pass through biological barriers. To overcome biological barriers, researchers have been developing nanomedicines with transformable physicochemical properties in recent years. Physicochemical transformability enables nanomedicines to responsively switch their physicochemical properties, including size, shape, surface charge, etc., thus enabling them to cross a series of biological barriers and achieve maximum delivery efficiency. In this review, we summarize recent developments in nanomedicines with transformable physicochemical properties. First, the biological dilemmas faced by nanomedicines are analyzed. Furthermore, the design and synthesis of nanomaterials with transformable physicochemical properties in terms of size, charge, and shape are summarized. Other switchable physicochemical parameters such as mobility, roughness and mechanical properties, which have been sought after most recently, are also discussed. Finally, the prospects and challenges for nanomedicines with transformable physicochemical properties are highlighted.
Collapse
Affiliation(s)
- Qianqian Lu
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai 200433, P. R. China.
| | - Hongyue Yu
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai 200433, P. R. China.
| | - Tiancong Zhao
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai 200433, P. R. China.
| | - Guanjia Zhu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, P. R. China.
| | - Xiaomin Li
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai 200433, P. R. China.
| |
Collapse
|
46
|
Fan D, Cao Y, Cao M, Wang Y, Cao Y, Gong T. Nanomedicine in cancer therapy. Signal Transduct Target Ther 2023; 8:293. [PMID: 37544972 PMCID: PMC10404590 DOI: 10.1038/s41392-023-01536-y] [Citation(s) in RCA: 145] [Impact Index Per Article: 72.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 08/08/2023] Open
Abstract
Cancer remains a highly lethal disease in the world. Currently, either conventional cancer therapies or modern immunotherapies are non-tumor-targeted therapeutic approaches that cannot accurately distinguish malignant cells from healthy ones, giving rise to multiple undesired side effects. Recent advances in nanotechnology, accompanied by our growing understanding of cancer biology and nano-bio interactions, have led to the development of a series of nanocarriers, which aim to improve the therapeutic efficacy while reducing off-target toxicity of the encapsulated anticancer agents through tumor tissue-, cell-, or organelle-specific targeting. However, the vast majority of nanocarriers do not possess hierarchical targeting capability, and their therapeutic indices are often compromised by either poor tumor accumulation, inefficient cellular internalization, or inaccurate subcellular localization. This Review outlines current and prospective strategies in the design of tumor tissue-, cell-, and organelle-targeted cancer nanomedicines, and highlights the latest progress in hierarchical targeting technologies that can dynamically integrate these three different stages of static tumor targeting to maximize therapeutic outcomes. Finally, we briefly discuss the current challenges and future opportunities for the clinical translation of cancer nanomedicines.
Collapse
Affiliation(s)
- Dahua Fan
- Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, 528300, China.
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
| | - Yongkai Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Meiqun Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Yajun Wang
- Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, 528300, China
| | | | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
47
|
Yan M, Chen Q, Liu T, Li X, Pei P, Zhou L, Zhou S, Zhang R, Liang K, Dong J, Wei X, Wang J, Terasaki O, Chen P, Gu Z, Jiang L, Kong B. Site-selective superassembly of biomimetic nanorobots enabling deep penetration into tumor with stiff stroma. Nat Commun 2023; 14:4628. [PMID: 37532754 PMCID: PMC10397308 DOI: 10.1038/s41467-023-40300-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 06/24/2023] [Indexed: 08/04/2023] Open
Abstract
Chemotherapy remains as the first-choice treatment option for triple-negative breast cancer (TNBC). However, the limited tumor penetration and low cellular internalization efficiency of current nanocarrier-based systems impede the access of anticancer drugs to TNBC with dense stroma and thereby greatly restricts clinical therapeutic efficacy, especially for TNBC bone metastasis. In this work, biomimetic head/hollow tail nanorobots were designed through a site-selective superassembly strategy. We show that nanorobots enable efficient remodeling of the dense tumor stromal microenvironments (TSM) for deep tumor penetration. Furthermore, the self-movement ability and spiky head markedly promote interfacial cellular uptake efficacy, transvascular extravasation, and intratumoral penetration. These nanorobots, which integrate deep tumor penetration, active cellular internalization, near-infrared (NIR) light-responsive release, and photothermal therapy capacities into a single nanodevice efficiently suppress tumor growth in a bone metastasis female mouse model of TNBC and also demonstrate potent antitumor efficacy in three different subcutaneous tumor models.
Collapse
Affiliation(s)
- Miao Yan
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials, iChEM, Fudan University, 200438, Shanghai, P. R. China
| | - Qing Chen
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, 200032, Shanghai, P. R. China
| | - Tianyi Liu
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials, iChEM, Fudan University, 200438, Shanghai, P. R. China
| | - Xiaofeng Li
- Department of Chemistry, The University of Hong Kong, Hong Kong, 999077, P. R. China
| | - Peng Pei
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials, iChEM, Fudan University, 200438, Shanghai, P. R. China
| | - Lei Zhou
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, 200032, Shanghai, P. R. China
| | - Shan Zhou
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials, iChEM, Fudan University, 200438, Shanghai, P. R. China
| | - Runhao Zhang
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials, iChEM, Fudan University, 200438, Shanghai, P. R. China
| | - Kang Liang
- School of Chemical Engineering and Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Jian Dong
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, 200032, Shanghai, P. R. China
| | - Xunbin Wei
- Biomedical Engineering Department and Cancer Hospital and Institute, Key Laboratory of Carcinogenesis and Translational Research, Peking University, 100081, Beijing, P. R. China
| | - Jinqiang Wang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, 310063, Hangzhou, P. R. China
| | - Osamu Terasaki
- School of Physical Science and Technology, ShanghaiTech University, 201210, Shanghai, P. R. China
| | - Pu Chen
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Zhen Gu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, 310063, Hangzhou, P. R. China
| | - Libo Jiang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, 200032, Shanghai, P. R. China.
| | - Biao Kong
- Department of Chemistry, Shanghai Key Lab of Molecular Catalysis and Innovative Materials, iChEM, Fudan University, 200438, Shanghai, P. R. China.
- Yiwu Research Institute of Fudan University, 322000, Yiwu, Zhejiang, P. R. China.
- Shandong Research Institute, Fudan University, 250103, Jinan, Shandong, P. R. China.
| |
Collapse
|
48
|
Wang X, Zhang H, Chen X, Wu C, Ding K, Sun G, Luo Y, Xiang D. Overcoming tumor microenvironment obstacles: Current approaches for boosting nanodrug delivery. Acta Biomater 2023; 166:42-68. [PMID: 37257574 DOI: 10.1016/j.actbio.2023.05.043] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/12/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
In order to achieve targeted delivery of anticancer drugs, efficacy improvement, and side effect reduction, various types of nanoparticles are employed. However, their therapeutic effects are not ideal. This phenomenon is caused by tumor microenvironment abnormalities such as abnormal blood vessels, elevated interstitial fluid pressure, and dense extracellular matrix that affect nanoparticle penetration into the tumor's interstitium. Furthermore, nanoparticle properties including size, charge, and shape affect nanoparticle transport into tumors. This review comprehensively goes over the factors hindering nanoparticle penetration into tumors and describes methods for improving nanoparticle distribution by remodeling the tumor microenvironment and optimizing nanoparticle physicochemical properties. Finally, a critical analysis of future development of nanodrug delivery in oncology is further discussed. STATEMENT OF SIGNIFICANCE: This article reviews the factors that hinder the distribution of nanoparticles in tumors, and describes existing methods and approaches for improving the tumor accumulation from the aspects of remodeling the tumor microenvironment and optimizing the properties of nanoparticles. The description of the existing methods and approaches is followed by highlighting their advantages and disadvantages and put forward possible directions for the future researches. At last, the challenges of improving tumor accumulation in nanomedicines design were also discussed. This review will be of great interest to the broad readers who are committed to delivering nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Xiaohui Wang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Hong Zhang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Xiaohui Chen
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Chunrong Wu
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Ke Ding
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Guiyin Sun
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China.
| | - Yang Luo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Debing Xiang
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China.
| |
Collapse
|
49
|
Yang Y, Wang B, Zhang X, Li H, Yue S, Zhang Y, Yang Y, Liu M, Ye C, Huang P, Zhou X. Activatable Graphene Quantum-Dot-Based Nanotransformers for Long-Period Tumor Imaging and Repeated Photodynamic Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2211337. [PMID: 37025038 DOI: 10.1002/adma.202211337] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/06/2023] [Indexed: 06/09/2023]
Abstract
Photodynamic therapy (PDT) is considered as an emerging therapeutic modality against cancer with high spatiotemporal selectivity because the utilized photosensitizers (PSs) are only active and toxic upon light irradiation. To maximize its effectiveness, PDT is usually applied repetitively for ablating various tumors. However, the total overdose of PSs from repeated administrations causes severe side effects. Herein, acidity-activated graphene quantum dots-based nanotransformers (GQD NT) are developed as PS vehicles for long-period tumor imaging and repeated PDT. Under the guidance of Arg-Gly-Asp peptide, GQD NT targets to tumor tissues actively, and then loosens and enlarges in tumor acidity, thus promising long tumor retention. Afterwards, GQD NT transforms into small pieces for better penetration in tumor. Upon laser irradiation, GQD NT generates mild hyperthermia that enhances cell membrane permeability and further promotes the PSs uptake. Most intriguingly, the as-prepared GQD NT not only "turns-on" fluorescence/magnetic resonance signals, but also achieves efficient repeated PDT. Notably, the total PSs dose is reduced to 3.5 µmol kg-1 , which is 10-30 times lower than that of other reported works. Overall, this study exploits a smart vehicle to enhance accumulation, retention, and release of PSs in tumors through programmed deformation, thus overcoming the overdose obstacle in repeated PDT.
Collapse
Affiliation(s)
- Yuqi Yang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences - Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
- Optics Valley Laboratory, Wuhan, Hubei, 430073, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Baolong Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences - Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
- Optics Valley Laboratory, Wuhan, Hubei, 430073, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xu Zhang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences - Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
- Optics Valley Laboratory, Wuhan, Hubei, 430073, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hongchuang Li
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences - Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
- Optics Valley Laboratory, Wuhan, Hubei, 430073, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Sen Yue
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences - Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
- Optics Valley Laboratory, Wuhan, Hubei, 430073, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yifan Zhang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Yunhuang Yang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences - Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
- Optics Valley Laboratory, Wuhan, Hubei, 430073, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Maili Liu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences - Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
- Optics Valley Laboratory, Wuhan, Hubei, 430073, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chaohui Ye
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences - Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
- Optics Valley Laboratory, Wuhan, Hubei, 430073, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Xin Zhou
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences - Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
- Optics Valley Laboratory, Wuhan, Hubei, 430073, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
50
|
Azizi M, Jahanban-Esfahlan R, Samadian H, Hamidi M, Seidi K, Dolatshahi-Pirouz A, Yazdi AA, Shavandi A, Laurent S, Be Omide Hagh M, Kasaiyan N, Santos HA, Shahbazi MA. Multifunctional nanostructures: Intelligent design to overcome biological barriers. Mater Today Bio 2023; 20:100672. [PMID: 37273793 PMCID: PMC10232915 DOI: 10.1016/j.mtbio.2023.100672] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/24/2023] [Accepted: 05/18/2023] [Indexed: 06/06/2023] Open
Abstract
Over the past three decades, nanoscience has offered a unique solution for reducing the systemic toxicity of chemotherapy drugs and for increasing drug therapeutic efficiency. However, the poor accumulation and pharmacokinetics of nanoparticles are some of the key reasons for their slow translation into the clinic. The is intimately linked to the non-biological nature of nanoparticles and the aberrant features of solid cancer, which together significantly compromise nanoparticle delivery. New findings on the unique properties of tumors and their interactions with nanoparticles and the human body suggest that, contrary to what was long-believed, tumor features may be more mirage than miracle, as the enhanced permeability and retention based efficacy is estimated to be as low as 1%. In this review, we highlight the current barriers and available solutions to pave the way for approved nanoformulations. Furthermore, we aim to discuss the main solutions to solve inefficient drug delivery with the use of nanobioengineering of nanocarriers and the tumor environment. Finally, we will discuss the suggested strategies to overcome two or more biological barriers with one nanocarrier. The variety of design formats, applications and implications of each of these methods will also be evaluated.
Collapse
Affiliation(s)
- Mehdi Azizi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Dental Implants Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Samadian
- Dental Implants Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Masoud Hamidi
- Université Libre de Bruxelles (ULB), École Polytechnique de Bruxelles-BioMatter Unit, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Khaled Seidi
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Amirhossein Ahmadieh Yazdi
- Department of Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amin Shavandi
- Université Libre de Bruxelles (ULB), École Polytechnique de Bruxelles-BioMatter Unit, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Sophie Laurent
- General, Organic and Biomedical Chemistry Unit, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons – UMONS, Mons, Belgium
| | - Mahsa Be Omide Hagh
- Immunology Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nahid Kasaiyan
- Department of Nephrology and Hypertension, University Medical Center Utrecht, 3508 GA, Utrecht, Netherlands
| | - Hélder A. Santos
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
| |
Collapse
|