1
|
Hazra RS, Dutta D, Khan MRH, Tani S, Abdullah CS, Remex NS, Aishwarya R, Islam T, Yang Z, Mallik S, Bhuiyan MS, Choi Y, Quadir M. Mechanistic Underpinnings of Epigenetic Enzyme-Responsive Nanoparticles. ACS APPLIED BIO MATERIALS 2025. [PMID: 40307668 DOI: 10.1021/acsabm.4c02007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Epigenetic enzyme-responsive nanoparticles (ERPs), which destabilize under the influence of histone deacetylase 8 (HDAC8), have been characterized in terms of their physicochemical, nanomechanical, and in vitro attributes. These nanoparticles were synthesized from amphiphilic block copolymers composed of poly(acetyl l-lysine) as the hydrophobic and poly(ethylene glycol) as the hydrophilic block. Upon optimizing the particle assembly process, we demonstrated that the nanoscale properties of ERPs, including hydrodynamic diameter (DH), size distribution, and stiffness, are selectively influenced by the HDAC8 enzyme. Using 1H NMR and circular dichroism spectroscopy, we identified that deacetylation of the lysine residues located within the hydrophobic block acts as a molecular driver for the enzyme-induced destabilization of ERPs. Comprehensive in vitro studies demonstrated that ERPs are biocompatible and noncytotoxic and can accumulate in perinuclear and several cellular organelles, including lysosomes and mitochondria. Collectively, this study attempts to establish the mechanistic background of ERP functions, providing the baseline for designing epigenetic-enzyme-sensitive nanoscale platforms.
Collapse
Affiliation(s)
- Raj Shankar Hazra
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Debasmita Dutta
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Md Rakib Hasan Khan
- Biomedical Engineering Program, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Sakurako Tani
- Deapartment of Physics, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71103, United States
| | - Naznin Sultana Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71103, United States
| | - Richa Aishwarya
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71103, United States
| | - Tamjid Islam
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71103, United States
| | - Zhongyu Yang
- Department of Chemical and Biomedical Engineering, University of Misouri, Columbia, Missouri 65211, United States
| | - Sanku Mallik
- College of Pharmacy and Allied Health Professions, South Dakota State University, Brooking, South Dakota 57007, United States
| | - Md Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71103, United States
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71103, United States
| | - Yongki Choi
- Deapartment of Physics, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58102, United States
- Biomedical Engineering Program, North Dakota State University, Fargo, North Dakota 58102, United States
| |
Collapse
|
2
|
Yamada S, Sasaki E, Ohno H, Hanaoka K. Heat-guided drug delivery via thermally induced crosslinking of polymeric micelles. Commun Chem 2024; 7:287. [PMID: 39627351 PMCID: PMC11615195 DOI: 10.1038/s42004-024-01383-0] [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: 05/21/2024] [Accepted: 11/28/2024] [Indexed: 12/06/2024] Open
Abstract
Targeted drug delivery in response to external stimuli is therapeutically desirable, but long-term drug retention at the target site after stimulation is turned off remains a challenge. Herein, we present a targeted-delivery strategy via irreversible aggregation of drug carriers in response to mild external heating. We constructed two types of polymeric micelles, DBCO-TRM and Az-TRM, having a thermo-responsive polymer shell based on N-isopropylacrylamide (NIPAAm) and incorporating alkyne and azide moieties, respectively. Upon heating at 42 °C, the micelles aggregated through hydrophobic interaction between their dehydrated shells. Further, the azide moieties of Az-TRM become exposed on the surface due to the thermally shrinkage of the shells, thereby enabling crosslinking between the two types of micelles via azide-alkyne click chemistry to form irreversible aggregates. These aggregates were efficiently accumulated at tumor sites in mice by local heating after intravenous administration of a mixture of the micelles, and were well retained after cessation of heating due to their increased size. As proof of concept, we show that delivery of doxorubicin in this heat-guided drug delivery system dramatically improved the anti-tumor effect in a mouse model after a single treatment. Our results suggest that this platform could be an efficient tool for on-demand drug delivery.
Collapse
Affiliation(s)
- Sota Yamada
- Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, 105-8512, Japan
| | - Eita Sasaki
- Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, 105-8512, Japan
| | - Hisashi Ohno
- Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, 105-8512, Japan
| | - Kenjiro Hanaoka
- Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, 105-8512, Japan.
| |
Collapse
|
3
|
Ray P, Sedigh A, Confeld M, Alhalhooly L, Iduoku K, Casanola-Martin GM, Pham-The H, Rasulev B, Choi Y, Yang Z, Mallik S, Quadir M. Design and evaluation of nanoscale materials with programmed responsivity towards epigenetic enzymes. J Mater Chem B 2024; 12:9905-9920. [PMID: 39021201 DOI: 10.1039/d4tb00514g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Self-assembled materials capable of modulating their assembly properties in response to specific enzymes play a pivotal role in advancing 'intelligent' encapsulation platforms for biotechnological applications. Here, we introduce a previously unreported class of synthetic nanomaterials that programmatically interact with histone deacetylase (HDAC) as the triggering stimulus for disassembly. These nanomaterials consist of co-polypeptides comprising poly(acetyl L-lysine) and poly(ethylene glycol) blocks. Under neutral pH conditions, they self-assemble into particles. The hydrodynamic diameters of particles were typically withing the range of 108-190 nm, depending on degree of acetylation of the hydrophobic block. However, their stability is compromised upon exposure to HDACs, depending on enzyme concentration and exposure time. Our investigation, utilizing HDAC8 as the model enzyme, revealed that the primary mechanism behind disassembly involves a decrease in amphiphilicity within the block copolymer due to the deacetylation of lysine residues within the particles' hydrophobic domains. To elucidate the response mechanism, we encapsulated a fluorescent dye within these nanoparticles. Upon incubation with HDAC, the nanoparticle structure collapsed, leading to controlled release of the dye over time. Notably, this release was not triggered by denatured HDAC8, other proteolytic enzymes like trypsin, or the co-presence of HDAC8 and its inhibitor. We also demonstrated the biocompatibility and cellular effects of these materials in the context of drug delivery in different types of anticancer cell lines, such as MIA PaCa-2, PANC-1, cancer like stem cells (CSCs), and non-cancerous HPNE cells. We observed that the release of a model drug (such as a STAT3 pathway inhibitor, Napabucasin) can be loaded into these nanoparticles, with >90% of the dosage can be released over 3 h under the influence of HDAC8 enzyme in a controlled fashion. Further, we conducted a comprehensive computational study to unveil the possible interaction mechanism between enzymes and particles. By drawing parallels to the mechanism of naturally occurring histone proteins, this research represents a pioneering step toward developing functional materials capable of harnessing the activity of epigenetic enzymes such as HDACs.
Collapse
Affiliation(s)
- Priyanka Ray
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND 58102, USA.
| | - Abbas Sedigh
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58102, USA
| | - Matthew Confeld
- Deapartment of Physics, North Dakota State University, Fargo, ND 58102, USA
| | - Lina Alhalhooly
- Department of Chemistry and Biochemistry, North Dakota State University, Fargo, ND 58102, USA
| | - Kweeni Iduoku
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND 58102, USA.
| | - Gerardo M Casanola-Martin
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND 58102, USA.
| | - Hai Pham-The
- University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi 10000, Vietnam
| | - Bakhtiyor Rasulev
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND 58102, USA.
| | - Yongki Choi
- Department of Chemistry and Biochemistry, North Dakota State University, Fargo, ND 58102, USA
| | - Zhongyu Yang
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58102, USA
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58102, USA
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND 58102, USA.
| |
Collapse
|
4
|
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
|
5
|
Chu Z, Wang W, Zheng W, Fu W, Wang Y, Wang H, Qian H. Biomaterials with cancer cell-specific cytotoxicity: challenges and perspectives. Chem Soc Rev 2024; 53:8847-8877. [PMID: 39092634 DOI: 10.1039/d4cs00636d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Significant advances have been made in materials for biomedical applications, including tissue engineering, bioimaging, cancer treatment, etc. In the past few decades, nanostructure-mediated therapeutic strategies have been developed to improve drug delivery, targeted therapy, and diagnosis, maximizing therapeutic effectiveness while reducing systemic toxicity and side effects by exploiting the complicated interactions between the materials and the cell and tissue microenvironments. This review briefly introduces the differences between the cells and tissues of tumour or normal cells. We summarize recent advances in tumour microenvironment-mediated therapeutic strategies using nanostructured materials. We then comprehensively discuss strategies for fabricating nanostructures with cancer cell-specific cytotoxicity by precisely controlling their composition, particle size, shape, structure, surface functionalization, and external energy stimulation. Finally, we present perspectives on the challenges and future opportunities of nanotechnology-based toxicity strategies in tumour therapy.
Collapse
Affiliation(s)
- Zhaoyou Chu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, P. R. China.
- The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China.
| | - Wanni Wang
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, P. R. China.
| | - Wang Zheng
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, P. R. China.
| | - Wanyue Fu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, P. R. China.
| | - Yujie Wang
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, P. R. China.
| | - Hua Wang
- The First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China.
| | - Haisheng Qian
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, P. R. China.
- Anhui Engineering Research Center for Medical Micro-Nano Devices, Anhui Medical University, Hefei 230011, P. R. China
| |
Collapse
|
6
|
Qian C, Zhao G, Huo M, Su M, Hu X, Liu Q, Wang L. Tumor microenvironment-regulated drug delivery system combined with sonodynamic therapy for the synergistic treatment of breast cancer. RSC Adv 2024; 14:17612-17626. [PMID: 38828276 PMCID: PMC11141688 DOI: 10.1039/d4ra00539b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 05/16/2024] [Indexed: 06/05/2024] Open
Abstract
Co-loading of sonosensitizers and chemotherapeutic drugs into nanocarriers can improve the biocompatibilities, stabilities, and targeting of drugs and reduce the adverse reactions of drugs, providing a robust platform to orchestrate the synergistic interplay between chemotherapy and sonodynamic therapy (SDT) in cancer treatment. In this regard, biodegradable manganese dioxide (MnO2) has attracted widespread attention because of its unique properties in the tumor microenvironment (TME). Accordingly, herein, MnO2 nanoshells with hollow mesoporous structures (H-MnO2) were etched to co-load hematoporphyrin monomethyl ether (HMME) and doxorubicin (DOX), and DOX/HMME-HMnO2@bovine serum albumin (BSA) obtained after simple BSA modification of DOX/HMME-HMnO2 exhibited excellent hydrophilicity and dispersibility. H-MnO2 rapidly degraded in the weakly acidic TME, releasing loaded HMME and DOX, and catalysed the decomposition of H2O2 abundantly present in TME, producing oxygen (O2) in situ, significantly increasing O2 concentration and downregulating the hypoxia-inducible factor 1α (HIF-1α). After irradiation of the tumor area with low-frequency ultrasound, the drug delivery efficiency of DOX/HMME-HMnO2@BSA substantially increased, and the excited HMME generated a large amount of reactive oxygen species (ROS), which caused irreversible damage to tumor cells. Moreover, the cell death rate exceeded 60% after synergistic SDT-chemotherapy. Therefore, the pH-responsive nanoshells designed in this study can realize drug accumulation in tumor regions by responding to TME and augment SDT-chemotherapy potency for breast cancer treatment by improving hypoxia in tumors. Thus, this study provides theoretical support for the development of multifunctional nanocarriers and scientific evidence for further exploration of safer and more efficient breast cancer treatments.
Collapse
Affiliation(s)
- Chao Qian
- Shandong Provincial Hospital, Shandong University Jinan 250000 China
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan 250021 China
| | - Guoliang Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University Jinan 250014 China
| | - Mengping Huo
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan 250021 China
| | - Meixia Su
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan 250021 China
| | - Xuexue Hu
- School of Clinical Medicine, Shandong First Medical University Jinan 250117 China
| | - Qiang Liu
- Shandong Provincial Hospital, Shandong University Jinan 250000 China
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan 250021 China
| | - Lei Wang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan 250021 China
| |
Collapse
|
7
|
Zhou Y, Zhang J, Sun S, Chen W, Wang Y, Shi H, Yang R, Qing Z. Amplified Biosensors Powered by Endogenous Molecules for Intracellular Fluorescence Imaging. Anal Chem 2024; 96:8078-8090. [PMID: 38622818 DOI: 10.1021/acs.analchem.4c00329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Affiliation(s)
- Yibo Zhou
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Chemical Engineering, Changsha University of Science and Technology, Changsha 410114, P.R. China
| | - Jun Zhang
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Chemical Engineering, Changsha University of Science and Technology, Changsha 410114, P.R. China
| | - Shuanghong Sun
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Chemical Engineering, Changsha University of Science and Technology, Changsha 410114, P.R. China
| | - Weiju Chen
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Chemical Engineering, Changsha University of Science and Technology, Changsha 410114, P.R. China
| | - Yuping Wang
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Chemical Engineering, Changsha University of Science and Technology, Changsha 410114, P.R. China
| | - Huiqiu Shi
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Chemical Engineering, Changsha University of Science and Technology, Changsha 410114, P.R. China
| | - Ronghua Yang
- Laboratory of Chemical Biology & Traditional Chinese Medicine Research, Ministry of Education, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, P.R. China
| | - Zhihe Qing
- Hunan Provincial Key Laboratory of Cytochemistry, School of Chemistry and Chemical Engineering, Changsha University of Science and Technology, Changsha 410114, P.R. China
| |
Collapse
|
8
|
Ray P, Sedigh A, Confeld M, Alhalhooly L, Iduoku K, Casanola-Martin GM, Pham-The H, Rasulev B, Choi Y, Yang Z, Mallik S, Quadir M. Design and Evaluation of Nanoscale Materials with Programmed Responsivity towards Epigenetic Enzymes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.585429. [PMID: 38586020 PMCID: PMC10996597 DOI: 10.1101/2024.03.26.585429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Self-assembled materials capable of modulating their assembly properties in response to specific enzymes play a pivotal role in advancing 'intelligent' encapsulation platforms for biotechnological applications. Here, we introduce a previously unreported class of synthetic nanomaterials that programmatically interact with histone deacetylase (HDAC) as the triggering stimulus for disassembly. These nanomaterials consist of co-polypeptides comprising poly (acetyl L-lysine) and poly(ethylene glycol) blocks. Under neutral pH conditions, they self-assemble into particles. However, their stability is compromised upon exposure to HDACs, depending on enzyme concentration and exposure time. Our investigation, utilizing HDAC8 as the model enzyme, revealed that the primary mechanism behind disassembly involves a decrease in amphiphilicity within the block copolymer due to the deacetylation of lysine residues within the particles' hydrophobic domains. To elucidate the response mechanism, we encapsulated a fluorescent dye within these nanoparticles. Upon incubation with HDAC, the nanoparticle structure collapsed, leading to controlled release of the dye over time. Notably, this release was not triggered by denatured HDAC8, other proteolytic enzymes like trypsin, or the co-presence of HDAC8 and its inhibitor. We further demonstrated the biocompatibility and cellular effects of these materials and conducted a comprehensive computational study to unveil the possible interaction mechanism between enzymes and particles. By drawing parallels to the mechanism of naturally occurring histone proteins, this research represents a pioneering step toward developing functional materials capable of harnessing the activity of epigenetic enzymes such as HDACs.
Collapse
|
9
|
Dey S, Mondal A, Aash A, Mukherjee R, Kolay S, Murmu N, Murmu N, Giri B, Molla MR. Poly-β-thioester-Based Cross-Linked Nanocarrier for Cancer Cell Selectivity over Normal Cells and Cellular Apoptosis by Triggered Release of Parthenolide, an Anticancer Drug. ACS APPLIED BIO MATERIALS 2024; 7:1214-1228. [PMID: 38326023 DOI: 10.1021/acsabm.3c01121] [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: 02/09/2024]
Abstract
Breast cancer is the most prevalent and aggressive type of cancer, causing high mortality rates in women globally. Many drawbacks and side effects of the current chemotherapy force us to develop a robust chemotherapeutic system that can deal with off-target hazards and selectively combat cancer growth, invasiveness, and cancer-initiating cells. Here, a pH-responsive cross-linked nanocarrier (140-160 nm) endowed with poly-β-thioester functionality (CBAPTL) has been sketched and fabricated for noncovalent firm encapsulation of anticancer drug, parthenolide (PTL) at physiological pH (7.4), which enables sustain release of PTL at relevant endosomal pH (∼5.0-5.3). For this, a bolaamphiphilic molecule integrated with β-thioester and acrylate functionality was synthesized to fabricate the pH-responsive poly-β-thioester-based cross-linked nanocarrier via Michael addition click reactions in water. The poly-β-thioester functionality of CBAPTL hydrolyzes at endosomal acidic conditions, thus leading to the selective release of PTL inside the cancer cell. Cross-linked nanocarriers exhibit high serum stability, dilution insensitivity, and targeted cellular uptake at tumor microenvironment (TME), contrasting normal cells. In vitro study using human MCF-7 breast cancer cells demonstrated that CBAPTL exhibited selective cytotoxicity, reduced clonogenic potential, increased reactive oxygen species (ROS) generation, and arrested the progression of the cell cycle at the G0/G1 phase efficiently. CBAPTL induced apoptosis via downregulating pro-proliferative protein Bcl-2 and upregulating proapoptotic proteins p53, BAD, p21, and cleaved PARP-1. CBAPTL inhibited proliferating signaling by suppressing AKT phosphorylation and p38 expression. CBAPTL also blocked the invasion and migration of MCF-7 cells. CBAPTL effectively inhibits primary and secondary mammosphere formation, thereby preventing cancer-initiating cells' growth. Conversely, CBAPTL has negligible effect on human red blood cells (RBCs) and peripheral blood mononuclear cells (PBMCs). These findings highlight the superior efficacy of CBAPTL compared to PTL alone in suppressing cancer cell growth, inducing apoptosis, and preventing invasiveness of MCF-7 cells. Thus, CBAPTL could be considered a possible selective chemotherapeutic cargo against breast cancer without affecting normal cells.
Collapse
Affiliation(s)
- Sananda Dey
- Department of Physiology, University of Gour Banga, Malda 732103, West Bengal, India
| | - Arun Mondal
- Department of Chemistry, University of Calcutta, Kolkata 700009, West Bengal, India
| | - Asmita Aash
- Department of Chemistry, University of Calcutta, Kolkata 700009, West Bengal, India
| | - Rimi Mukherjee
- Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, Kolkata 700026, West Bengal, India
| | - Soumya Kolay
- Department of Chemistry, University of Calcutta, Kolkata 700009, West Bengal, India
| | - Nensina Murmu
- Department of Physiology, University of Gour Banga, Malda 732103, West Bengal, India
| | - Nabendu Murmu
- Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, Kolkata 700026, West Bengal, India
| | - Biplab Giri
- Department of Physiology, University of Gour Banga, Malda 732103, West Bengal, India
| | | |
Collapse
|
10
|
Park D, Lee SJ, Park JW. Aptamer-Based Smart Targeting and Spatial Trigger-Response Drug-Delivery Systems for Anticancer Therapy. Biomedicines 2024; 12:187. [PMID: 38255292 PMCID: PMC10813750 DOI: 10.3390/biomedicines12010187] [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: 12/15/2023] [Revised: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
In recent years, the field of drug delivery has witnessed remarkable progress, driven by the quest for more effective and precise therapeutic interventions. Among the myriad strategies employed, the integration of aptamers as targeting moieties and stimuli-responsive systems has emerged as a promising avenue, particularly in the context of anticancer therapy. This review explores cutting-edge advancements in targeted drug-delivery systems, focusing on the integration of aptamers and stimuli-responsive platforms for enhanced spatial anticancer therapy. In the aptamer-based drug-delivery systems, we delve into the versatile applications of aptamers, examining their conjugation with gold, silica, and carbon materials. The synergistic interplay between aptamers and these materials is discussed, emphasizing their potential in achieving precise and targeted drug delivery. Additionally, we explore stimuli-responsive drug-delivery systems with an emphasis on spatial anticancer therapy. Tumor microenvironment-responsive nanoparticles are elucidated, and their capacity to exploit the dynamic conditions within cancerous tissues for controlled drug release is detailed. External stimuli-responsive strategies, including ultrasound-mediated, photo-responsive, and magnetic-guided drug-delivery systems, are examined for their role in achieving synergistic anticancer effects. This review integrates diverse approaches in the quest for precision medicine, showcasing the potential of aptamers and stimuli-responsive systems to revolutionize drug-delivery strategies for enhanced anticancer therapy.
Collapse
Affiliation(s)
- Dongsik Park
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Su Jin Lee
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Jee-Woong Park
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| |
Collapse
|
11
|
Xie S, Wei L, Liu Y, Meng J, Cao W, Qiu B, Li X. Size-tunable nanogels for cascaded release of metronidazole and chemotherapeutic agents to combat Fusobacterium nucleatum-infected colorectal cancer. J Control Release 2024; 365:16-28. [PMID: 37956925 DOI: 10.1016/j.jconrel.2023.11.018] [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: 07/06/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/21/2023]
Abstract
Bacteria play important roles in tumor formation, growth and metastasis through downregulating immune response and initiating drug resistance. Herein, size-tunable nanogels (NGs) have been developed to address the existing size paradox in tumor accumulation, intratumoral penetration and intracellular release of therapeutics for the treatment of Fusobacterium nucleatum (F. nucleatum)-infected colorectal cancer. Zinc-imidazolate frameworks with doxorubicin (DOX) loading and folate grafting (f-ZIFD) were mixed with metronidazole (MET) and encapsulated in NGs through thiol-ene click crosslinking of sulfhydryl hyaluronan, sulfhydryl alginate and 4-arm poly(ethylene glycol) acrylate. Hyaluronidase-initiated matrix degradation causes NG swelling to release sufficient MET and maintains a large size for an extended time period, and the gradually discharged f-ZIFD nanoparticles (NPs) from NGs exhibit acid-responsive intracellular release of DOX after folate-mediated internalization into tumor cells. The encapsulation into NGs significantly enhances the bioavailability and increases half-lives of MET and DOX by around 20 times. In the F. nucleatum-infected tumor model, the extended retention of swollen NGs and the efficient tumor infiltration and cellular uptake of the discharged f-ZIFD NPs cause 6 times higher DOX levels in tumors than that of free DOX administration. F. nucleatum promotes tumor cell proliferation and tumor growth, and the cascaded releases of MET and f-ZIFD NPs eliminate F. nucleatum to effectively inhibit tumor growth with a significant extension of animal survival. Thus, the hyaluronidase-mediated NG expansion and dual-responsive cascaded drug release have overcome challenges in the release regimen and size paradox of drug delivery carriers to combat bacteria-infected cancer.
Collapse
Affiliation(s)
- Shuang Xie
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, PR China; Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Li Wei
- Department of Pathology, Western Theater Command Air Force Hospital, Chengdu 610021, PR China
| | - Yuan Liu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Jie Meng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, PR China; Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Wenxiong Cao
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, PR China; Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Bo Qiu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Xiaohong Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, PR China; Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China.
| |
Collapse
|
12
|
Kenchegowda M, Hani U, Al Fatease A, Haider N, Ramesh KVRNS, Talath S, Gangadharappa HV, Kiran Raj G, Padmanabha SH, Osmani RAM. Tiny titans- unravelling the potential of polysaccharides and proteins based dissolving microneedles in drug delivery and theranostics: A comprehensive review. Int J Biol Macromol 2023; 253:127172. [PMID: 37793514 DOI: 10.1016/j.ijbiomac.2023.127172] [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: 08/17/2023] [Revised: 09/22/2023] [Accepted: 09/29/2023] [Indexed: 10/06/2023]
Abstract
In recent years, microneedles (MNs) have emerged as a promising alternative to traditional drug delivery systems in transdermal drug delivery. The use of MNs has demonstrated significant potential in improving patient acceptance and convenience while avoiding the invasiveness of traditional injections. Dissolving, solid, hollow, coated, and hydrogel microneedles are among the various types studied for drug delivery. Dissolving microneedles (DMNs), in particular, have gained attention for their safety, painlessness, patient convenience, and high delivery efficiency. This comprehensive review primarily focuses on different types of microneedles, fabrication methods, and materials used in fabrication of DMNs such as hyaluronic acid, chitosan, alginate, gelatin, collagen, silk fibroin, albumin, cellulose and starch, to list a few. The review also provides an exhaustive discussion on the applications of DMNs, including the delivery of vaccines, cosmetic agents, contraceptives, hormone and genes, and other therapeutic applications like for treating cancer, skin diseases, and diabetes, among others, are covered in this review. Additionally, this review highlights some of the DMN systems that are presently undergoing clinical trials. Finally, the review discusses current advances and trends in DMNs, as well as future prospective directions for this ground-breaking technology in drug delivery.
Collapse
Affiliation(s)
- Madhuchandra Kenchegowda
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Adel Al Fatease
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Nazima Haider
- Department of Pathology, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
| | - K V R N S Ramesh
- Department of Pharmaceutics, RAK College of Pharmaceutical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Sirajunisa Talath
- Department of Pharmaceutical Chemistry, RAK College of Pharmaceutical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Hosahalli V Gangadharappa
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - G Kiran Raj
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| | - Sharath Honganoor Padmanabha
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
| | - Riyaz Ali M Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India.
| |
Collapse
|
13
|
Zhang X, Wang J, Zhang Y, Yang Z, Gao J, Gu Z. Synthesizing biomaterials in living organisms. Chem Soc Rev 2023; 52:8126-8164. [PMID: 37921625 DOI: 10.1039/d2cs00999d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Living organisms fabricate biomacromolecules such as DNA, RNA, and proteins by the self-assembly process. The research on the mechanism of biomacromolecule formation also inspires the exploration of in vivo synthesized biomaterials. By elaborate design, artificial building blocks or precursors can self-assemble or polymerize into functional biomaterials within living organisms. In recent decades, these so-called in vivo synthesized biomaterials have achieved extensive applications in cell-fate manipulation, disease theranostics, bioanalysis, cellular surface engineering, and tissue regeneration. In this review, we classify strategies for in vivo synthesis into non-covalent, covalent, and genetic types. The development of these approaches is based on the chemical principles of supramolecular chemistry and synthetic chemistry, biological cues such as enzymes and microenvironments, and the means of synthetic biology. By summarizing the design principles in detail, some insights into the challenges and opportunities in this field are provided to enlighten further research.
Collapse
Affiliation(s)
- Xiangyang Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China.
| | - Junxia Wang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China.
| | - Zhimou Yang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China.
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China.
| | - Zhen Gu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
14
|
Geng Y, Williams GR. Developing and scaling up captopril-loaded electrospun ethyl cellulose fibers for sustained-release floating drug delivery. Int J Pharm 2023; 648:123557. [PMID: 39491226 DOI: 10.1016/j.ijpharm.2023.123557] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/05/2024]
Abstract
In this work ethyl cellulose (EC) was used as the matrix polymer and loaded with captopril, with the goal to fabricate electrospun fibers as potential sustained-release floating gastro-retentive drug delivery systems. Fibers were prepared with monoaxial and coaxial electrospinning, and both bench-top and scaled-up (needle-based) methods were explored. With monoaxial electrospinning, EC-based fibers in the shape of cylinders and with smooth surfaces were obtained both at 1 and 20 mL/h. For coaxial electrospinning, the drug was encapsulated in the core end fibers generated with core/shell feeding rates of 0.5/1 and 5/10 mL/h. The fibers were cylindrical in shape with a wrinkled surface, and confocal microscopy suggested them to have a core/shell structure. X-ray diffraction and differential scanning calorimetry results showed that all the fibers were amorphous. The encapsulation efficiency of all the formulations was almost 100%. Release studies in simulated gastric fluid indicated that the monoaxial electrospun fibers gave slower release profiles compared with a physical mixture of captopril and EC, but there was still an initial "burst" of release at the start of the experiment. Fibers with low drug-loading (9.09% w/w) showed a slower release than fibers with high loading (23.08% w/w). The coaxial fibers exhibited sustained release profiles with reduced initial burst release. Both monoaxial and coaxial fibers could float on the surface of simulated gastric fluid for over 24 hours at 37 °C. After storage under ambient conditions (19-21°C, relative humidity 30-40%) for 8 weeks, all the fibers remained amorphous and the release profiles had no significant changes compared with fresh fibers. This work thus highlights the potential of coaxial electrospinning for fabricating a sustained-release floating gastro-retentive drug delivery system for captopril.
Collapse
Affiliation(s)
- Yuhao Geng
- UCL School of Pharmacy, 29 - 39 Brunswick Square, London, WC1N 1AX
| | | |
Collapse
|
15
|
Zetrini AE, Lip H, Abbasi AZ, Alradwan I, Ahmed T, He C, Henderson JT, Rauth AM, Wu XY. Remodeling Tumor Immune Microenvironment by Using Polymer-Lipid-Manganese Dioxide Nanoparticles with Radiation Therapy to Boost Immune Response of Castration-Resistant Prostate Cancer. RESEARCH (WASHINGTON, D.C.) 2023; 6:0247. [PMID: 37795337 PMCID: PMC10546607 DOI: 10.34133/research.0247] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/17/2023] [Indexed: 10/06/2023]
Abstract
Despite substantial progress in the treatment of castration-resistant prostate cancer (CRPC), including radiation therapy and immunotherapy alone or in combination, the response to treatment remains poor due to the hypoxic and immunosuppressive nature of the tumor microenvironment. Herein, we exploited the bioreactivity of novel polymer-lipid manganese dioxide nanoparticles (PLMDs) to remodel the tumor immune microenvironment (TIME) by increasing the local oxygen levels and extracellular pH and enhancing radiation-induced immunogenic cell death. This study demonstrated that PLMD treatment sensitized hypoxic human and murine CRPC cells to radiation, significantly increasing radiation-induced DNA double-strand breaks and ultimately cell death, which enhanced the secretion of damage-associated molecular patterns, attributable to the induction of autophagy and endoplasmic reticulum stress. Reoxygenation via PLMDs also polarized hypoxic murine RAW264.7 macrophages toward the M1 phenotype, enhancing tumor necrosis factor alpha release, and thus reducing the viability of murine CRPC TRAMP-C2 cells. In a syngeneic TRAMP-C2 tumor model, intravenous injection of PLMDs suppressed, while radiation alone enhanced recruitment of regulatory T cells and myeloid-derived suppressor cells. Pretreatment with PLMDs followed by radiation down-regulated programmed death-ligand 1 and promoted the infiltration of antitumor CD8+ T cells and M1 macrophages to tumor sites. Taken together, TIME modulation by PLMDs plus radiation profoundly delayed tumor growth and prolonged median survival compared with radiation alone. These results suggest that PLMDs plus radiation is a promising treatment modality for improving therapeutic efficacy in radioresistant and immunosuppressive solid tumors.
Collapse
Affiliation(s)
- Abdulmottaleb E. Zetrini
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy,
University of Toronto, M5S 3M2, Toronto, ON, Canada
| | - HoYin Lip
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy,
University of Toronto, M5S 3M2, Toronto, ON, Canada
| | - Azhar Z. Abbasi
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy,
University of Toronto, M5S 3M2, Toronto, ON, Canada
| | - Ibrahim Alradwan
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy,
University of Toronto, M5S 3M2, Toronto, ON, Canada
| | - Taksim Ahmed
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy,
University of Toronto, M5S 3M2, Toronto, ON, Canada
| | - Chunsheng He
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy,
University of Toronto, M5S 3M2, Toronto, ON, Canada
| | - Jeffrey T. Henderson
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy,
University of Toronto, M5S 3M2, Toronto, ON, Canada
| | - Andrew M. Rauth
- Departments of Medical Biophysics and Radiation Oncology,
University of Toronto, M5G 1L7, Toronto, ON, Canada
| | - Xiao Yu Wu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy,
University of Toronto, M5S 3M2, Toronto, ON, Canada
| |
Collapse
|
16
|
Yan Z, Liu Y, Zhao L, Hu J, Du Y, Peng X, Liu Z. In situ stimulus-responsive self-assembled nanomaterials for drug delivery and disease treatment. MATERIALS HORIZONS 2023; 10:3197-3217. [PMID: 37376926 DOI: 10.1039/d3mh00592e] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
The individual motifs that respond to specific stimuli for the self-assembly of nanomaterials play important roles. In situ constructed nanomaterials are formed spontaneously without human intervention and have promising applications in bioscience. However, due to the complex physiological environment of the human body, designing stimulus-responsive self-assembled nanomaterials in vivo is a challenging problem for researchers. In this article, we discuss the self-assembly principles of various nanomaterials in response to the tissue microenvironment, cell membrane, and intracellular stimuli. We propose the applications and advantages of in situ self-assembly in drug delivery and disease diagnosis and treatment, with a focus on in situ self-assembly at the lesion site, especially in cancer. Additionally, we introduce the significance of introducing exogenous stimulation to construct self-assembly in vivo. Based on this foundation, we put forward the prospects and possible challenges in the field of in situ self-assembly. This review uncovers the relationship between the structure and properties of in situ self-assembled nanomaterials and provides new ideas for innovative drug molecular design and development to solve the problems in the targeted delivery and precision medicine.
Collapse
Affiliation(s)
- Ziling Yan
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, P. R. China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, P. R. China
| | - Licheng Zhao
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, P. R. China
| | - Jiaxin Hu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, P. R. China.
| | - Yimin Du
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, P. R. China.
| | - Xingxing Peng
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, P. R. China.
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, P. R. China.
- Molecular Imaging Research Center of Central South University, Changsha 410008, Hunan Province, P. R. China
| |
Collapse
|
17
|
Santra S, Das S, Sengupta A, Molla MR. Tumor acidity-induced surface charge modulation in covalent nanonetworks for activated cellular uptake: targeted delivery of anticancer drugs and selective cancer cell death. Biomater Sci 2023; 11:5549-5559. [PMID: 37401615 DOI: 10.1039/d3bm00491k] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
A β-thioester and tertiary amine based covalently cross-linked nanoassembly coined as a nanonetwork (NN) endowed with dual pH responsive features (tumor acidity induced surface charge modulation and endosomal pH triggered controlled degradation) has been designed and synthesized for stable sequestration and sustained release of drug molecules in response to endosomal pH. An amphiphile integrated with tertiary amine and acrylate (ATA) functionalities was synthesized to fabricate the nanonetwork. This amphiphile showed entropically driven self-assembly and micellar nanostructures (nanoassemblies), which can sequester hydrophobic drug molecules at neutral pH. To further stabilize the nanoassemblies and the sequestered drug molecules even below its critical aggregation concentration (CAC), the micellar core was cross-linked via the thiol-acrylate Michael addition click reaction to generate multiple copies of acid labile β-thioester functionalities in the core, which undergo slow hydrolysis at endosomal pH (∼5.0), thus enabling sustained release of the anti-cancer drug doxorubicin at endosomal pH. The nanonetworks showed a significant decrease in drug leakage compared to the nanoassemblies (NAs), which was also justified by a low leakage coefficient calculated from the fluorescence resonance energy transfer experiment. The NN also exhibited dilution insensitivity and high serum stability, whereas the NA disassembled upon dilution and during serum treatment. The biological evaluation revealed tumor extracellular matrix pH (∼6.4-6.8) induced surface charge modulation and cancer cell (HeLa) selective activated cellular uptake of the doxorubicin loaded nanonetwork (NN-DOX). In contrast, the benign nature of NN-DOX towards normal cells (H9c2) suggests excellent cell specificity. Thus, we believe that the ease of synthesis, nanonetwork fabrication reproducibility, robust stability, smart nature of tumor microenvironment sensitive surface charge modulation, boosted tumoral-cell uptake, and triggered drug release will make this system a potential nanomedicine for chemotherapeutic treatments.
Collapse
Affiliation(s)
- Subrata Santra
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata-700009, India.
| | - Shreya Das
- Department of Life Science & Biotechnology, Jadavpur University, Kolkata-700032, India
| | - Arunima Sengupta
- Department of Life Science & Biotechnology, Jadavpur University, Kolkata-700032, India
| | - Mijanur Rahaman Molla
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata-700009, India.
| |
Collapse
|
18
|
Lin G, Zhou J, Cheng H, Liu G. Smart Nanosystems for Overcoming Multiple Biological Barriers in Cancer Nanomedicines Transport: Design Principles, Progress, and Challenges. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207973. [PMID: 36971279 DOI: 10.1002/smll.202207973] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/28/2023] [Indexed: 06/18/2023]
Abstract
The development of smart nanosystems, which could overcome diverse biological barriers of nanomedicine transport, has received intense scientific interest in improving the therapeutic efficacies of traditional nanomedicines. However, the reported nanosystems generally hold disparate structures and functions, and the knowledge of involved biological barriers is usually scattered. There is an imperative need for a summary of biological barriers and how these smart nanosystems conquer biological barriers, to guide the rational design of the new-generation nanomedicines. This review starts from the discussion of major biological barriers existing in nanomedicine transport, including blood circulation, tumoral accumulation and penetration, cellular uptake, drug release, and response. Design principles and recent progress of smart nanosystems in overcoming the biological barriers are overviewed. The designated physicochemical properties of nanosystems can dictate their functions in biological environments, such as protein absorption inhibition, tumor accumulation, penetration, cellular internalization, endosomal escape, and controlled release, as well as modulation of tumor cells and their resident tumor microenvironment. The challenges facing smart nanosystems on the road heading to clinical approval are discussed, followed by the proposals that could further advance the nanomedicine field. It is expected that this review will provide guidelines for the rational design of the new-generation nanomedicines for clinical use.
Collapse
Affiliation(s)
- Gan Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- Department of Chemistry, the University of Chicago, Chicago, IL, 60637, USA
| | - Jiajing Zhou
- College of Biomass Science and Engineering, Key Laboratory of Leather Chemistry and Engineering of Ministry of Education, National Engineering Laboratory for Clean Technology of Leather Manufacture, Sichuan University, Chengdu, 610065, China
| | - Hongwei Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
19
|
Li X, Huang Z, Liao Z, Liu A, Huo S. Transformable nanodrugs for overcoming the biological barriers in the tumor environment during drug delivery. NANOSCALE 2023; 15:8532-8547. [PMID: 37114478 DOI: 10.1039/d2nr06621a] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Drug delivery systems have been studied massively with explosive growth in the last few decades. However, challenges such as biological barriers are still obstructing the delivery efficiency of nanomedicines. Reports have shown that the physicochemical properties, such as the morphologies of nanodrugs, could highly affect their biodistribution and bioavailability. Therefore, transformable nanodrugs that take advantage of different sizes and shapes allow for overcoming multiple biological barriers, providing promising prospects for drug delivery. This review aims to present an overview of the most recent developments of transformable nanodrugs in this emerging field. First, the design principles and transformation mechanisms which serve as guidelines for smart nanodrugs are summarized. Afterward, their applications in overcoming biological barriers, including the bloodstream, intratumoral pressure, cellular membrane, endosomal wrapping, and nuclear membrane, are highlighted. Finally, discussions on the current developments and future perspectives of transformable nanodrugs are given.
Collapse
Affiliation(s)
- Xuejian Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Zhenkun Huang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Zhihuan Liao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Aijie Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Shuaidong Huo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
20
|
Wlodarczyk MT, Dragulska SA, Chen Y, Poursharifi M, Acosta Santiago M, Martignetti JA, Mieszawska AJ. Pt(II)-PLGA Hybrid in a pH-Responsive Nanoparticle System Targeting Ovarian Cancer. Pharmaceutics 2023; 15:pharmaceutics15020607. [PMID: 36839929 PMCID: PMC9961376 DOI: 10.3390/pharmaceutics15020607] [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: 01/13/2023] [Revised: 02/01/2023] [Accepted: 02/06/2023] [Indexed: 02/15/2023] Open
Abstract
Platinum-based agents are the main treatment option in ovarian cancer (OC). Herein, we report a poly(lactic-co-glycolic acid) (PLGA) nanoparticle (NP) encapsulating platinum (II), which is targeted to a cell-spanning protein overexpressed in above 90% of late-stage OC, mucin 1 (MUC1). The NP is coated with phospholipid-DNA aptamers against MUC1 and a pH-sensitive PEG derivative containing an acid-labile hydrazone linkage. The pH-sensitive PEG serves as an off-on switch that provides shielding effects at the physiological pH and is shed at lower pH, thus exposing the MUC1 ligands. The pH-MUC1-Pt NPs are stable in the serum and display pH-dependent PEG cleavage and drug release. Moreover, the NPs effectively internalize in OC cells with higher accumulation at lower pH. The Pt (II) loading into the NP was accomplished via PLGA-Pt (II) coordination chemistry and was found to be 1.62 wt.%. In vitro screening using a panel of OC cell lines revealed that pH-MUC1-Pt NP has a greater effect in reducing cellular viability than carboplatin, a clinically relevant drug analogue. Biodistribution studies have demonstrated NP accumulation at tumor sites with effective Pt (II) delivery. Together, these results demonstrate a potential for pH-MUC1-Pt NP for the enhanced Pt (II) therapy of OC and other solid tumors currently treated with platinum agents.
Collapse
Affiliation(s)
- Marek T. Wlodarczyk
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY 10016, USA
| | - Sylwia A. Dragulska
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
| | - Ying Chen
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Mina Poursharifi
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, USA
| | - Maxier Acosta Santiago
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
| | - John A. Martignetti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
- Women’s Health Research Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
- Rudy Ruggles Research Institute, Western Connecticut Health Network, 131 West St., Danbury, CT 06810, USA
| | - Aneta J. Mieszawska
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Correspondence:
| |
Collapse
|
21
|
Kolarikova M, Hosikova B, Dilenko H, Barton-Tomankova K, Valkova L, Bajgar R, Malina L, Kolarova H. Photodynamic therapy: Innovative approaches for antibacterial and anticancer treatments. Med Res Rev 2023. [PMID: 36757198 DOI: 10.1002/med.21935] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 12/07/2022] [Accepted: 01/03/2023] [Indexed: 02/10/2023]
Abstract
Photodynamic therapy is an alternative treatment mainly for cancer but also for bacterial infections. This treatment dates back to 1900 when a German medical school graduate Oscar Raab found a photodynamic effect while doing research for his doctoral dissertation with Professor Hermann von Tappeiner. Unexpectedly, Raab revealed that the toxicity of acridine on paramecium depends on the intensity of light in his laboratory. Photodynamic therapy is therefore based on the administration of a photosensitizer with subsequent light irradiation within the absorption maxima of this substance followed by reactive oxygen species formation and finally cell death. Although this treatment is not a novelty, there is an endeavor for various modifications to the therapy. For example, selectivity and efficiency of the photosensitizer, as well as irradiation with various types of light sources are still being modified to improve final results of the photodynamic therapy. The main aim of this review is to summarize anticancer and antibacterial modifications, namely various compounds, approaches, and techniques, to enhance the effectiveness of photodynamic therapy.
Collapse
Affiliation(s)
- Marketa Kolarikova
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Barbora Hosikova
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Hanna Dilenko
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Katerina Barton-Tomankova
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Lucie Valkova
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Robert Bajgar
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Lukas Malina
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Hana Kolarova
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
22
|
Sun X, Xu X, Wang J, Zhang X, Zhao Z, Liu X, Wang G, Teng L, Chen X, Wang D, Li Y. Acid-switchable nanoparticles induce self-adaptive aggregation for enhancing antitumor immunity of natural killer cells. Acta Pharm Sin B 2023. [PMID: 37521862 PMCID: PMC10373095 DOI: 10.1016/j.apsb.2023.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Deficiency of natural killer (NK) cells shows a significant impact on tumor progression and failure of immunotherapy. It is highly desirable to boost NK cell immunity by upregulating active receptors and relieving the immunosuppressive tumor microenvironment. Unfortunately, mobilization of NK cells is hampered by poor accumulation and short retention of drugs in tumors, thus declining antitumor efficiency. Herein, we develop an acid-switchable nanoparticle with self-adaptive aggregation property for co-delivering galunisertib and interleukin 15 (IL-15). The nanoparticles induce morphology switch by a decomposition-metal coordination cascade reaction, which provides a new methodology to trigger aggregation. It shows self-adaptive size-enlargement upon acidity, thus improving drug retention in tumor to over 120 h. The diameter of agglomerates is increased and drug release is effectively promoted following reduced pH values. The nanoparticles activate both NK cell and CD8+ T cell immunity in vivo. It significantly suppresses CT26 tumor in immune-deficient BALB/c mice, and the efficiency is further improved in immunocompetent mice, indicating that the nanoparticles can not only boost innate NK cell immunity but also adaptive T cell immunity. The approach reported here provides an innovative strategy to improve drug retention in tumors, which will enhance cancer immunotherapy by boosting NK cells.
Collapse
|
23
|
Ramachandran M, Ma Z, Lin K, De Souza C, Li Y. Transformable nanoparticles to bypass biological barriers in cancer treatment. NANOSCALE ADVANCES 2022; 4:4470-4480. [PMID: 36341301 PMCID: PMC9595105 DOI: 10.1039/d2na00485b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/03/2022] [Indexed: 06/02/2023]
Abstract
Nanomedicine based drug delivery platforms provide an interesting avenue to explore for the future of cancer treatment. Here we discuss the barriers for drug delivery in cancer therapeutics and how nanomaterials have been designed to bypass these blockades through stimuli responsive transformation in the most recent update. Nanomaterials that address the challenges of each step provide a promising solution for new cancer therapeutics.
Collapse
Affiliation(s)
- Mythili Ramachandran
- Department of Biochemistry and Molecular Medicine, University of California-Davis USA
| | - Zhao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University Jinan Shandong China
| | - Kai Lin
- College of Food Science and Engineering, Ocean University of China Qingdao China
| | | | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, University of California-Davis USA
| |
Collapse
|
24
|
Anboo S, Lau SY, Kansedo J, Yap P, Hadibarata T, Jeevanandam J, Kamaruddin AH. Recent advancements in enzyme-incorporated nanomaterials: Synthesis, mechanistic formation, and applications. Biotechnol Bioeng 2022; 119:2609-2638. [PMID: 35851660 PMCID: PMC9543334 DOI: 10.1002/bit.28185] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/21/2022] [Accepted: 07/15/2022] [Indexed: 11/09/2022]
Abstract
Over the past decade, nanotechnology has been developed and employed across various entities. Among the numerous nanostructured material types, enzyme-incorporated nanomaterials have shown great potential in various fields, as an alternative to biologically derived as well as synthetically developed hybrid structures. The mechanism of incorporating enzyme onto a nanostructure depends on several factors including the method of immobilization, type of nanomaterial, as well as operational and environmental conditions. The prospects of enzyme-incorporated nanomaterials have shown promising results across various applications, such as biocatalysts, biosensors, drug therapy, and wastewater treatment. This is due to their excellent ability to exhibit chemical and physical properties such as high surface-to-volume ratio, recovery and/or reusability rates, sensitivity, response scale, and stable catalytic activity across wide operating conditions. In this review, the evolution of enzyme-incorporated nanomaterials along with their impact on our society due to its state-of-the-art properties, and its significance across different industrial applications are discussed. In addition, the weakness and future prospects of enzyme-incorporated nanomaterials were also discussed to guide scientists for futuristic research and development in this field.
Collapse
Affiliation(s)
- Shamini Anboo
- Department of Chemical EngineeringFaculty of Engineering and Science, Curtin University MalaysiaMiriSarawakMalaysia
| | - Sie Yon Lau
- Department of Chemical EngineeringFaculty of Engineering and Science, Curtin University MalaysiaMiriSarawakMalaysia
| | - Jibrail Kansedo
- Department of Chemical EngineeringFaculty of Engineering and Science, Curtin University MalaysiaMiriSarawakMalaysia
| | - Pow‐Seng Yap
- Department of Civil EngineeringXi'an Jiaotong‐Liverpool UniversitySuzhouChina
| | - Tony Hadibarata
- Department of Chemical EngineeringFaculty of Engineering and Science, Curtin University MalaysiaMiriSarawakMalaysia
| | | | - Azlina H. Kamaruddin
- School of Chemical EngineeringUniversiti Sains MalaysiaSeberang Perai SelatanPenangMalaysia
| |
Collapse
|
25
|
Thankachan D, Anbazhagan R, Krishnamoorthi R, Tsai HC, Gebrie HT, Darge HF, Lu CH, Chen JK. MnO2 nanoparticle encapsulated in polyelectrolytic hybrids from alkyl functionalized carboxymethyl cellulose and azide functionalized gelatin to treat tumors by photodynamic therapy and photothermal therapy. J Taiwan Inst Chem Eng 2022. [DOI: 10.1016/j.jtice.2022.104503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
26
|
He X, Zhu H, Shang J, Li M, Zhang Y, Zhou S, Gong G, He Y, Blocki A, Guo J. Intratumoral synthesis of transformable metal-phenolic nanoaggregates with enhanced tumor penetration and retention for photothermal immunotherapy. Am J Cancer Res 2022; 12:6258-6272. [PMID: 36168635 PMCID: PMC9475467 DOI: 10.7150/thno.74808] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/19/2022] [Indexed: 11/05/2022] Open
Abstract
Rationale: Effective photothermal therapy (PTT) remains a great challenge due to the difficulties of delivering photothermal agents with both deep penetration and prolonged retention at tumor lesion spatiotemporally. Methods: Here, we report an intratumoral self-assembled nanostructured aggregate named FerH, composed of a natural polyphenol and a commercial iron supplement. FerH assemblies possess size-increasing dynamic kinetics as a pseudo-stepwise polymerization from discrete nanocomplexes to microscale aggregates. Results: The nanocomplex can penetrate deeply into solid tumors, followed by prolonged retention (> 6 days) due to the in vivo growth into nanoaggregates in the tumor microenvironment. FerH performs a targeting ablation of tumors with a high photothermal conversion efficiency (60.2%). Importantly, an enhanced immunotherapeutic effect on the distant tumor can be triggered when co-administrated with checkpoint-blockade PD-L1 antibody. Conclusions: Such a therapeutic approach by intratumoral synthesis of metal-phenolic nanoaggregates can be instructive to address the challenges associated with malignant tumors.
Collapse
Affiliation(s)
- Xianglian He
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Hongfu Zhu
- Collage of Material Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Jiaojiao Shang
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Meifeng Li
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China.,Department of Biomass Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yaoyao Zhang
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China.,Key Laboratory of Birth Defects and Related of Women and Children of Ministry of Education, Department of Pediatrics, The Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shicheng Zhou
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China.,Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Guidong Gong
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yunxiang He
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Anna Blocki
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong (CUHK), Shatin, Hong Kong SAR, China
| | - Junling Guo
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China.,State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan 610065, China.,Bioproducts Institute, Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
27
|
Taghipour YD, Zarebkohan A, Salehi R, Rahimi F, Torchilin VP, Hamblin MR, Seifalian A. An update on dual targeting strategy for cancer treatment. J Control Release 2022; 349:67-96. [PMID: 35779656 DOI: 10.1016/j.jconrel.2022.06.044] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/04/2022] [Accepted: 06/24/2022] [Indexed: 12/18/2022]
Abstract
The key issue in the treatment of solid tumors is the lack of efficient strategies for the targeted delivery and accumulation of therapeutic cargoes in the tumor microenvironment (TME). Targeting approaches are designed for more efficient delivery of therapeutic agents to cancer cells while minimizing drug toxicity to normal cells and off-targeting effects, while maximizing the eradication of cancer cells. The highly complicated interrelationship between the physicochemical properties of nanoparticles, and the physiological and pathological barriers that are required to cross, dictates the need for the success of targeting strategies. Dual targeting is an approach that uses both purely biological strategies and physicochemical responsive smart delivery strategies to increase the accumulation of nanoparticles within the TME and improve targeting efficiency towards cancer cells. In both approaches, either one single ligand is used for targeting a single receptor on different cells, or two different ligands for targeting two different receptors on the same or different cells. Smart delivery strategies are able to respond to triggers that are typical of specific disease sites, such as pH, certain specific enzymes, or redox conditions. These strategies are expected to lead to more precise targeting and better accumulation of nano-therapeutics. This review describes the classification and principles of dual targeting approaches and critically reviews the efficiency of dual targeting strategies, and the rationale behind the choice of ligands. We focus on new approaches for smart drug delivery in which synthetic and/or biological moieties are attached to nanoparticles by TME-specific responsive linkers and advanced camouflaged nanoparticles.
Collapse
Affiliation(s)
- Yasamin Davatgaran Taghipour
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zarebkohan
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Roya Salehi
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Fariborz Rahimi
- Department of Electrical Engineering, University of Bonab, Bonab, Iran
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine and Department of Chemical Engineering, Northeastern University, Boston, USA
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Laser Research Centre, Faculty of Health Science, University of Johannesburg, South Africa
| | - Alexander Seifalian
- Nanotechnology & Regenerative Medicine Commercialization Centre (NanoRegMed Ltd), London BioScience Innovation Centre, London, United Kingdom
| |
Collapse
|
28
|
Qian X, Xu X, Wu Y, Wang J, Li J, Chen S, Wen J, Li Y, Zhang Z. Strategies of engineering nanomedicines for tumor retention. J Control Release 2022; 346:193-211. [PMID: 35447297 DOI: 10.1016/j.jconrel.2022.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/03/2022] [Accepted: 04/05/2022] [Indexed: 01/29/2023]
Abstract
The retention of therapeutic agents in solid tumors at sufficient concentration and duration is crucial for their antitumor effects. Given the important contribution of nanomedicines to oncology, we herein summarized two major strategies of nanomedicines for tumor retention, such as transformation- and interactions-mediated strategies. The transformation-mediated retention strategy was achieved by enlarging particle size of nanomedicines or modulating the morphology into fibrous structures, while the interactions-mediated retention strategy was accomplished by modulating nanomedicines to promote their interactions with versatile cells or components in tumors. Moreover, we provide some considerations and perspectives of tumor-retaining nanomedicines for effective cancer therapy.
Collapse
Affiliation(s)
- Xindi Qian
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoxuan Xu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yao Wu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiaoying Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shuo Chen
- School of Pharmacy, the University of Auckland, Auckland 1142, New Zealand
| | - Jingyuan Wen
- School of Pharmacy, the University of Auckland, Auckland 1142, New Zealand
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China.; University of Chinese Academy of Sciences, Beijing 100049, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China.
| | - Zhiwen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China.; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
29
|
Jiang M, Liu Y, Dong Y, Wang K, Yuan Y. Bioorthogonal chemistry and illumination controlled programmed size-changeable nanomedicine for synergistic photodynamic and hypoxia-activated therapy. Biomaterials 2022; 284:121480. [DOI: 10.1016/j.biomaterials.2022.121480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/16/2022] [Accepted: 03/19/2022] [Indexed: 11/25/2022]
|
30
|
Cheng Y, Zhang Y, Wu H. Polymeric Fibers as Scaffolds for Spinal Cord Injury: A Systematic Review. Front Bioeng Biotechnol 2022; 9:807533. [PMID: 35223816 PMCID: PMC8864123 DOI: 10.3389/fbioe.2021.807533] [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: 11/02/2021] [Accepted: 12/16/2021] [Indexed: 11/30/2022] Open
Abstract
Spinal cord injury (SCI) is a complex neurological condition caused by trauma, inflammation, and other diseases, which often leads to permanent changes in strength and sensory function below the injured site. Changes in the microenvironment and secondary injuries continue to pose challenges for nerve repair and recovery after SCI. Recently, there has been progress in the treatment of SCI with the use of scaffolds for neural tissue engineering. Polymeric fibers fabricated by electrospinning have been increasingly used in SCI therapy owing to their biocompatibility, complex porous structure, high porosity, and large specific surface area. Polymer fibers simulate natural extracellular matrix of the nerve fiber and guide axon growth. Moreover, multiple channels of polymer fiber simulate the bundle of nerves. Polymer fibers with porous structure can be used as carriers loaded with drugs, nerve growth factors and cells. As conductive fibers, polymer fibers have electrical stimulation of nerve function. This paper reviews the fabrication, characterization, and application in SCI therapy of polymeric fibers, as well as potential challenges and future perspectives regarding their application.
Collapse
Affiliation(s)
- Yuanpei Cheng
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yanbo Zhang
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Han Wu
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
31
|
Hu Y, Gao S, Khan AR, Yang X, Ji J, Xi Y, Zhai G. Tumor microenvironment-responsive size-switchable drug delivery nanosystems. Expert Opin Drug Deliv 2022; 19:221-234. [PMID: 35164610 DOI: 10.1080/17425247.2022.2042512] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Compared with ordinary chemotherapeutic drugs, the variable-size nanoparticles (NPs) have better therapeutic effects and fewer side effects. AREAS COVERED This review mainly summarizes the strategies used to construct smart, size-tunable nanocarriers based on characteristic factors of tumor microenvironment (TME) to dramatically increase the penetration and retention of drugs within tumors. EXPERT OPINION Nanosystems with changeable sizes based on the TME have been extensively studied in the past decade, and their permeability and retention have been greatly improved, making them a very promising treatment for tumors.
Collapse
Affiliation(s)
- Yue Hu
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Shan Gao
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Abdur Rauf Khan
- Government of Punjab, Specialized HealthCare and Medical Education Department, Lahore, Pakistan
| | - Xiaoye Yang
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Jianbo Ji
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Yanwei Xi
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Guangxi Zhai
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| |
Collapse
|
32
|
Zhang J, Lin Y, Lin Z, Wei Q, Qian J, Ruan R, Jiang X, Hou L, Song J, Ding J, Yang H. Stimuli-Responsive Nanoparticles for Controlled Drug Delivery in Synergistic Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103444. [PMID: 34927373 PMCID: PMC8844476 DOI: 10.1002/advs.202103444] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/28/2021] [Indexed: 05/10/2023]
Abstract
Cancer immunotherapy has achieved promising clinical progress over the recent years for its potential to treat metastatic tumors and inhibit their recurrences effectively. However, low patient response rates and dose-limiting toxicity remain as major dilemmas for immunotherapy. Stimuli-responsive nanoparticles (srNPs) combined with immunotherapy offer the possibility to amplify anti-tumor immune responses, where the weak acidity, high concentration of glutathione, overexpressions of enzymes, and reactive oxygen species, and external stimuli in tumors act as triggers for controlled drug release. This review highlights the design of srNPs based on tumor microenvironment and/or external stimuli to combine with different anti-tumor drugs, especially the immunoregulatory agents, which eventually realize synergistic immunotherapy of malignant primary or metastatic tumors and acquire a long-term immune memory to prevent tumor recurrence. The authors hope that this review can provide theoretical guidance for the construction and clinical transformation of smart srNPs for controlled drug delivery in synergistic cancer immunotherapy.
Collapse
Affiliation(s)
- Jin Zhang
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Yandai Lin
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Zhe Lin
- Ruisi (Fujian) Biomedical Engineering Research Center Co LtdFuzhou350100P. R. China
| | - Qi Wei
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
- State Key Laboratory of Molecular Engineering of PolymersFudan University220 Handan RoadShanghai200433P. R. China
| | - Jiaqi Qian
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Renjie Ruan
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Xiancai Jiang
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Linxi Hou
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Jibin Song
- MOE Key Laboratory for Analytical Science of Food Safety and BiologyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
- State Key Laboratory of Molecular Engineering of PolymersFudan University220 Handan RoadShanghai200433P. R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and BiologyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| |
Collapse
|
33
|
Affiliation(s)
- Xiao Xu
- State Key Laboratory of Natural Medicines Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases Center of Advanced Pharmaceuticals and Biomaterials School of Life Science and Technology China Pharmaceutical University Nanjing China
| | - Shiyang Shen
- State Key Laboratory of Natural Medicines Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases Center of Advanced Pharmaceuticals and Biomaterials School of Life Science and Technology China Pharmaceutical University Nanjing China
| | - Ran Mo
- State Key Laboratory of Natural Medicines Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases Center of Advanced Pharmaceuticals and Biomaterials School of Life Science and Technology China Pharmaceutical University Nanjing China
| |
Collapse
|
34
|
Li J, Gong S, Li S, Li X, Lan S, Sun M. Tumor-penetrating iron oxide nanoclusters for T1/ T2 dual mode MR imaging-guided combination therapy. Biomater Sci 2022; 10:5254-5264. [DOI: 10.1039/d2bm00667g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A hyaluronic acid (HA)-stabilized iron oxide nanocluster (Fe2O3@PFDH NC) was developed as an intelligent tumor-penetrating theranostic nanoagent for dual-mode MRI guided chemo-photothermal therapy.
Collapse
Affiliation(s)
- Jing Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Siman Gong
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Shiyu Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Xinchong Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Siyi Lan
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Minjie Sun
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
35
|
Triple cascade nanocatalyst with laser-activatable O 2 supply and photothermal enhancement for effective catalytic therapy against hypoxic tumor. Biomaterials 2021; 280:121308. [PMID: 34896860 DOI: 10.1016/j.biomaterials.2021.121308] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 12/13/2022]
Abstract
Nanozymes have been combined with glucose oxidase (GOx) for dual-enzyme cascade catalytic therapy. However, their catalysis efficiency is restricted because of the hypoxia tumor microenvironment (TME). Although many methods are developed for O2 supply, the O2 leakage and consumption of H2O2 compromised their practical application. Herein, a biocompatible carbon nitride (C3N4)/nanozyme/GOx triple cascade nanocatalyst was designed with laser-activatable O2 self-supply via water splitting to relieve tumor hypoxia and thus improve the catalysis efficiency. To this end, polydopamine (PDA) nanosphere was prepared and attached with C3N4 nanosheet to improve water splitting efficiency and realize photothermal-enhanced catalysis, simultaneously. The PDA@C3N4 composite was then coated with MIL-100 (Fe), where GOx was loaded, to form C3N4/MIL-100/GOx triple cascade nanocatalyst. The triple cascade catalysis was realized with laser-activatable O2 supply from PDA@C3N4, H2O2 generation with GOx, and •OH production from peroxidase-like MIL-100 (Fe) for tumor therapy. Upon 808 nm irradiation, PDA, as a photothermal agent, realized photothermal therapy and enhanced the catalytic therapy. Thus, the synergy of laser-activatable O2 supply and photothermal enhancement in our triple cascade nanocatalyst improved the performance of catalytic therapy without drug resistance and toxicity to normal tissues.
Collapse
|
36
|
Wang H, Gao L, Fan T, Zhang C, Zhang B, Al-Hartomy OA, Al-Ghamdi A, Wageh S, Qiu M, Zhang H. Strategic Design of Intelligent-Responsive Nanogel Carriers for Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:54621-54647. [PMID: 34767342 DOI: 10.1021/acsami.1c13634] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Owing to the distinctive constituents of tumor tissue from those healthy organs, nanomedicine strategies show significant potentials in smart drug delivery. Nowadays, stimuli-responsive nanogels are playing increasingly important roles in the application of cancer therapy because of their sensitivity to various internal or external physicochemical stimuli, which exhibit site-specific and markedly enhanced drug release. Besides, nanogels are promising as drug carriers because of their porous structures, good biocompatibility, large surface area, and excellent capability with drugs. Taking advantage of multiresponsiveness, recent years have witnessed the rapid evolution of stimulus-responsive nanogels from monoresponsive to multiresponsive systems; however, there lacks a comprehensive review summarizing these reports. In this Review, we discuss the properties, synthesis, and characterization of nanogels. Moreover, tumor microenvironment and corresponding designing strategies for stimuli-response nanogels, both exogenous (temperature, magnetic field, light) and endogenous (pH, biomolecular, redox, ROS, pressure, hypoxia) are summarized on the basis of the recent advances in multistimuli-responsive nanogel systems. Nanogel and two-dimensional material composites show excellent performance in the field of constructing multistimulus-responsive nanoparticles and precise intelligent drug release integrated system for multimodal cancer diagnosis and therapy. Finally, potential progresses and suggestions are provided for the further design of hybrid nanogels based on emerging two-dimensional materials.
Collapse
Affiliation(s)
- Hao Wang
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, College of Optoelectronic Engineering, Institute of Microscale Optoelectronics, Shenzhen Institute of Translational Medicine, Department of Otolaryngology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen 518060, China
| | - Lingfeng Gao
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, No. 2318 Yuhangtang Rd., Cangqian, Yuhang District, Hangzhou 311121, China
| | - Taojian Fan
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, College of Optoelectronic Engineering, Institute of Microscale Optoelectronics, Shenzhen Institute of Translational Medicine, Department of Otolaryngology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen 518060, China
| | - Chen Zhang
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, College of Optoelectronic Engineering, Institute of Microscale Optoelectronics, Shenzhen Institute of Translational Medicine, Department of Otolaryngology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen 518060, China
| | - Bin Zhang
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, College of Optoelectronic Engineering, Institute of Microscale Optoelectronics, Shenzhen Institute of Translational Medicine, Department of Otolaryngology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen 518060, China
| | - Omar A Al-Hartomy
- Department of Physics, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ahmed Al-Ghamdi
- Department of Physics, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Swelm Wageh
- Department of Physics, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Meng Qiu
- Key Laboratory of Marine Chemistry Theory and Technology, Ocean University of China, Ministry of Education, Qingdao 266100, China
| | - Han Zhang
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, College of Optoelectronic Engineering, Institute of Microscale Optoelectronics, Shenzhen Institute of Translational Medicine, Department of Otolaryngology, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
37
|
Deng M, Rao JD, Guo R, Li M, He Q. Size-Adjustable Nano-Drug Delivery Systems for Enhanced Tumor Retention and Penetration. PHARMACEUTICAL FRONTS 2021. [DOI: 10.1055/s-0041-1736474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Over the past decades, nano-drug delivery systems have shown great potential in improving tumor treatment. And the controllability and design flexibility of nanoparticles endow them a broad development space. The particle size is one of the most important factors affecting the potency of nano-drug delivery systems. Large-size (100–200 nm) nanoparticles are more conducive to long circulation and tumor retention, but have poor tumor penetration; small-size (<50 nm) nanoparticles can deeply penetrate tumor but are easily cleared. Most of the current fixed-size nanoparticles are difficult to balance the retention and penetration, while the proposal of size-adjustable nano-drug delivery systems offers a solution to this paradox. Many endogenous and exogenous stimuli, such as acidic pH, upregulated enzymes, temperature, light, catalysts, redox conditions, and reactive oxygen species, can trigger the in situ transformation of nanoparticles based on protonation, hydrolysis, click reaction, phase transition, photoisomerization, redox reaction, etc. In this review, we summarize the principles and applications of stimuli-responsive size-adjustable strategies, including size-enlargement strategies and size-shrinkage strategies. We also propose the challenges faced by size-adjustable nano-drug delivery systems, hoping to promote the development of this strategy.
Collapse
Affiliation(s)
- Miao Deng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| | - Jing-Dong Rao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| | - Rong Guo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
38
|
Zhang X, Chen Y, He X, Zhang Y, Zhou M, Peng C, He Z, Gui S, Li Z. Smart Nanogatekeepers for Tumor Theranostics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2103712. [PMID: 34677898 DOI: 10.1002/smll.202103712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/05/2021] [Indexed: 06/13/2023]
Abstract
Nanoparticulate drug delivery systems (nano-DDSs) are required to reliably arrive and persistently reside at the tumor site with minimal off-target side effects for clinical theranostics. However, due to the complicated environment and high interstitial pressure in tumor tissue, they can return to the bloodstream and cause secondary side effects in normal organs. Recently, a number of nanogatekeepers have been engineered via structure-transformable/stable strategies to overcome this undesirable dilemma. The emerging structure-transformable nanogatekeepers for tumor imaging and therapy are first overviewed here, particularly for nanogatekeepers undergoing structural transformation in tumor microenvironments, cell membranes, and organelles. Thereafter, intelligent structure-stable nanogatekeepers through reversible activation and artificial individualization receptors are overviewed. Finally, the ongoing challenges and prospects of nanogatekeepers for clinical translation are briefly discussed.
Collapse
Affiliation(s)
- Xunfa Zhang
- College of Pharmacy, Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230012, China
| | - Yang Chen
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xian He
- College of Pharmacy, Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Education Office of Anhui Province, Hefei, 230012, China
| | - Yachao Zhang
- College of Pharmacy, Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Education Office of Anhui Province, Hefei, 230012, China
| | - Mei Zhou
- College of Pharmacy, Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230012, China
| | - Chengjun Peng
- College of Pharmacy, Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Education Office of Anhui Province, Hefei, 230012, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Shuangying Gui
- College of Pharmacy, Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Education Office of Anhui Province, Hefei, 230012, China
| | - Zhenbao Li
- College of Pharmacy, Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Education Office of Anhui Province, Hefei, 230012, China
| |
Collapse
|
39
|
Yu X, Su Q, Chang X, Chen K, Yuan P, Liu T, Tian R, Bai Y, Zhang Y, Chen X. Multimodal obstruction of tumorigenic energy supply via bionic nanocarriers for effective tumor therapy. Biomaterials 2021; 278:121181. [PMID: 34653932 DOI: 10.1016/j.biomaterials.2021.121181] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/05/2021] [Accepted: 10/07/2021] [Indexed: 12/11/2022]
Abstract
Sufficient energy generation based on effective transport of nutrient via abundant blood vessels in tumor tissue and subsequent oxidative metabolism in mitochondria is critical for growth, proliferation and migration of tumor. Thus the strategy to cut off this transport pathway (blood vessels) and simultaneously close the power house (mitochondria) is highly desired for tumor treatment. Herein, we fabricated a bionic nanocarrier with core-shell-corona structure to give selective and effective tumor therapy via stepwise destruction of existed tumor vessel, inhibition of tumor angiogenesis and dysfunction of tumor mitochondria. The core of this bionic nanocarrier consists of combretastatin A4 phosphate (CA4P) and vitamin K2 (VK2) co-loaded mesoporous silica nanoparticle (MSNs), which is in charge of the vasculature destruction and mitochondrial dysfunction after cargos release. The N-tert-butylacrylamide (TBAM) and tri-sulfated N-acetylglucosamine (TSAG) shell served as artificial affinity reagent against vascular endothelial growth factor (VEGF) for angiogenesis inhibition. As to guarantee that these actions only happened in tumor, the hyaluronic acid (HA) corona was introduced to endow the nanocarrier with tumor targeting property and stimuli-responsiveness for accurate therapy. Both in vitro and in vivo results indicated that the CA4P/VK2-MSNs-TBAM/TSAG-HA (CVMMGH for short) nanocarrier combined well-controllable manipulation of tumor vasculature and tumor mitochondria to effectivly cut off the tumorigenic energy supply, which performed significant inhibition of tumor growth, demonstrating the great candidate of our strategy for effective tumor therapy.
Collapse
Affiliation(s)
- Xiaoqian Yu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Qi Su
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Xiaowei Chang
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Kun Chen
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Pingyun Yuan
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Tao Liu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ran Tian
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yongkang Bai
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China.
| | - Xin Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
40
|
Cheng L, Niu MM, Yan T, Ma Z, Huang K, Yang L, Zhong X, Li C. Bioresponsive micro-to-nano albumin-based systems for targeted drug delivery against complex fungal infections. Acta Pharm Sin B 2021; 11:3220-3230. [PMID: 34729311 PMCID: PMC8546853 DOI: 10.1016/j.apsb.2021.04.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/09/2021] [Accepted: 03/25/2021] [Indexed: 12/14/2022] Open
Abstract
As a typical human pathogenic fungus, Cryptococcus neoformans is a life-threatening invasive fungal pathogen with a worldwide distribution causing ∼700,000 deaths annually. Cryptococcosis is not just an infection with multi-organ involvement, intracellular survival and extracellular multiplication of the fungus also play important roles in the pathogenesis of C. neoformans infections. Because adequate accumulation of drugs at target organs and cells is still difficult to achieve, an effective delivery strategy is desperately required to treat these infections. Here, we report a bioresponsive micro-to-nano (MTN) system that effectively clears the C. neoformans in vivo. This strategy is based on our in-depth study of the overexpression of matrix metalloproteinase 3 (MMP-3) in infectious microenvironments (IMEs) and secreted protein acidic and rich in cysteine (SPARC) in several associated target cells. In this MTN system, bovine serum albumin (BSA, a natural ligand of SPARC) was used for the preparation of nanoparticles (NPs), and then microspheres were constructed by conjugation with a special linker, which mainly consisted of a BSA-binding peptide and an MMP-3-responsive peptide. This MTN system was mechanically captured by the smallest capillaries of the lungs after intravenous injection, and then hydrolyzed into BSA NPs by MMP-3 in the IMEs. The NPs further targeted the lung tissue, brain and infected macrophages based on the overexpression of SPARC, reaching multiple targets and achieving efficient treatment. We have developed a size-tunable strategy where microspheres "shrink" to NPs in IMEs, which effectively combines active and passive targeting and may be especially powerful in the fight against complex fungal infections.
Collapse
Key Words
- Albumin
- AmB, amphotericin B
- BBB, blood‒brain barrier
- BSA, bovine serum albumin
- Complex fungal infection
- DDS, drug delivery system
- IME, infectious microenvironment
- MMP-3
- MMP-3, matrix metalloproteinase 3
- MTN, micro-to-nano
- Microenvironment responsive
- NP, nanoparticle
- PEG, polyethylene glycol
- PMVECs, pulmonary microvascular endothelial cells
- RFP, red fluorescent protein
- SPARC
- SPARC, secreted protein acidic and rich in cysteine
- Size-tunable strategy
Collapse
Affiliation(s)
- Liting Cheng
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Miao-Miao Niu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing 210009, China
| | - Tong Yan
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhongyi Ma
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Kexin Huang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Ling Yang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xin Zhong
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Chong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
41
|
Shen Y, Xu C, Chen J, Guan Z, Huang Y, Zeng Z, Xu X, Tan X, Zhao C. Phototriggered Self-Adaptive Functionalized MOC-Based Drug Delivery Platform Promises High Antitumor Efficacy. Adv Healthc Mater 2021; 10:e2100676. [PMID: 34414688 DOI: 10.1002/adhm.202100676] [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: 04/08/2021] [Revised: 07/23/2021] [Indexed: 11/10/2022]
Abstract
Due to their great stability and special cavities, metal-organic cages (MOCs) are increasingly considered as promising nanocarriers for drug delivery. However, the size and surface dilemmas restrict their further biomedical applications. The ultrasmall size of MOCs facilitates tumor penetration but suffers from quick clearance and poor accumulation at the tumor site. Hydrophobicity of MOC surfaces improves internalization into tumor cells while causing low blood circulation time as well as poor biocompatibility. Therefore, it remains challenging for the MOC-based drug delivery nanoplatform to realize high therapeutic efficacy because it requires different or even opposite dimensions and surface characteristics in different steps of circulation, penetration, accumulation, and internalization processes. In this study, an unprecedented phototriggered self-adaptive platform (ZnPc@polySCage) is developed by integrating functionalized MOCs and a photodynamic therapy based reactive oxygen species responsive strategy to realize high-efficiency tumor-specific therapy. ZnPc@polySCage remains hydrophilic and stealthy during circulation, and retains its small original size for tumor penetration, while transforming to a larger size for effective accumulation and hydrophobic for enhanced internalization under laser irradiation in tumor tissue. With these essential transitions, ZnPc@polySCage demonstrates prominent antitumor effects. Overall, the work provides an advantageous strategy for functional MOC-based platforms and biomedical applications.
Collapse
Affiliation(s)
- Yifeng Shen
- School of Pharmaceutical Sciences Sun Yat‐sen University Guangzhou 510006 P. R. China
| | - Congjun Xu
- School of Pharmaceutical Sciences Sun Yat‐sen University Guangzhou 510006 P. R. China
| | - Jie Chen
- School of Pharmaceutical Sciences Sun Yat‐sen University Guangzhou 510006 P. R. China
| | - Zilin Guan
- School of Pharmaceutical Sciences Sun Yat‐sen University Guangzhou 510006 P. R. China
| | - Yanjuan Huang
- School of Pharmaceutical Sciences Sun Yat‐sen University Guangzhou 510006 P. R. China
| | - Zishan Zeng
- School of Pharmaceutical Sciences Sun Yat‐sen University Guangzhou 510006 P. R. China
| | - Xiaoyu Xu
- School of Pharmaceutical Sciences Sun Yat‐sen University Guangzhou 510006 P. R. China
| | - Xiaomin Tan
- School of Pharmaceutical Sciences Sun Yat‐sen University Guangzhou 510006 P. R. China
| | - Chunshun Zhao
- School of Pharmaceutical Sciences Sun Yat‐sen University Guangzhou 510006 P. R. China
| |
Collapse
|
42
|
Liu Y, Jiang M, Tu Y, Wang K, Zong Q, Yuan Y. Time-programmed activation of dual polyprodrugs for synergistic cascade oxidation-chemotherapy. Biomaterials 2021; 278:121136. [PMID: 34562835 DOI: 10.1016/j.biomaterials.2021.121136] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/14/2021] [Accepted: 09/17/2021] [Indexed: 01/20/2023]
Abstract
Combination therapy using multiple drugs with time-programmed administration is promising for enhanced cancer treatment. However, it is still challenging to achieve time-programmed drug release from a single nanocarrier. Here, dual polyprodrugs of hemicyanine dye (CyNH2) and doxorubicin (DOX) are developed to achieve time-programmed prodrug activation for synergistic cascade oxidation therapy and chemotherapy. The polyprodrug NPDOX/Cy, composed of CyNH2, is modified with a glutathione (GSH)-responsive disulfate group, while DOX is modified with a reactive oxygen species (ROS)-response thioketal (TK) group. Upon uptake by cancer cells overexpressing GSH, CyNH2 can be activated quickly and accumulate in the mitochondria to induce mitochondrial damage and ROS upregulation, thus achieving subsequent burst activation of DOX through the ROS-triggered cleavage of the TK linker. The early activation of CyNH2 makes the cancer cells more sensitive to subsequent DOX treatment for a synergistic effect of from oxidation therapy and chemotherapy. Therefore, the polyprodrug with time-programmed drug activation developed in this work provides a promising strategy for synergistic cancer therapy.
Collapse
Affiliation(s)
- Ye Liu
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, PR China
| | - Maolin Jiang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China
| | - Yalan Tu
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, PR China
| | - Kewei Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China
| | - Qingyu Zong
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, PR China
| | - Youyong Yuan
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China.
| |
Collapse
|
43
|
Wang S, Zhang Z, Wei S, He F, Li Z, Wang HH, Huang Y, Nie Z. Near-infrared light-controllable MXene hydrogel for tunable on-demand release of therapeutic proteins. Acta Biomater 2021; 130:138-148. [PMID: 34082094 DOI: 10.1016/j.actbio.2021.05.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/10/2021] [Accepted: 05/26/2021] [Indexed: 12/22/2022]
Abstract
Precise delivery of therapeutic protein drugs that specifically modulate desired cellular responses is critical in clinical practice. However, the spatiotemporal regulation of protein drugs release to manipulate the target cell population in vivo remains a huge challenge. Herein, we have rationally developed an injectable and Near-infrared (NIR) light-responsive MXene-hydrogel composed of Ti3C2, agarose, and protein that enables flexibly and precisely control the release profile of protein drugs to modulate cellular behaviors with high spatiotemporal precision remotely. As a proof-of-concept study, we preloaded hepatic growth factor (HGF) into the MXene@hydrogel (MXene@agarose/HGF) to activate the c-Met-mediated signaling by NIR light. We demonstrated NIR light-instructed cell diffusion, migration, and proliferation at the user-defined localization, further promoting angiogenesis and wound healing in vivo. Our approach's versatility was validated by preloading tumor necrotic factor-α (TNF-α) into the composite hydrogel (MXene@agarose/TNF-α) to promote the pro-apoptotic signaling pathway, achieving the NIR light-induced programmed cell deaths (PCD) of tumor spheroids. Taking advantage of the deep-tissue penetrative NIR light, we could eradicate the deep-seated tumors in a xenograft model exogenously. Therefore, the proposed MXene-hydrogel provides the impetus for developing therapeutic synthetic materials for light-controlled drug release under thick tissue, which will find promising applications in regenerative medicine and tumor therapy. STATEMENT OF SIGNIFICANCE: Current stimuli-responsive hydrogels for therapeutic proteins delivery mainly depend on self-degradation, passive diffusion, or the responsiveness to cues relevant to diseases. However, it remains challenging to spatiotemporally deliver protein-based drugs to manipulate the target cell population in vivo in an "on-demand" manner. Therefore, we have rationally constructed an injectable and Near-infrared (NIR) light-responsive composite hydrogel by embedding Ti3C2 MXene and protein drugs within an agarose hydrogel to enable the remote control of protein drugs delivery with high spatiotemporal precision. The NIR light-controlled release of the growth factor or cytokine has been carried out to regulate receptor-mediated cellular behaviors under deep tissue for skin wound healing or cancer therapy. This system will provide the potential for precision medicine through the development of intelligent drug delivery systems.
Collapse
Affiliation(s)
- Song Wang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082 P.R. China
| | - Zhenhua Zhang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082 P.R. China
| | - Shaohua Wei
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082 P.R. China
| | - Fang He
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082 P.R. China
| | - Zhu Li
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082 P.R. China
| | - Hong-Hui Wang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082 P.R. China.
| | - Yan Huang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082 P.R. China
| | - Zhou Nie
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082 P.R. China.
| |
Collapse
|
44
|
Pan P, Yue Q, Li J, Gao M, Yang X, Ren Y, Cheng X, Cui P, Deng Y. Smart Cargo Delivery System based on Mesoporous Nanoparticles for Bone Disease Diagnosis and Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004586. [PMID: 34165902 PMCID: PMC8224433 DOI: 10.1002/advs.202004586] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/11/2021] [Indexed: 05/05/2023]
Abstract
Bone diseases constitute a major issue for modern societies as a consequence of progressive aging. Advantages such as open mesoporous channel, high specific surface area, ease of surface modification, and multifunctional integration are the driving forces for the application of mesoporous nanoparticles (MNs) in bone disease diagnosis and treatment. To achieve better therapeutic effects, it is necessary to understand the properties of MNs and cargo delivery mechanisms, which are the foundation and key in the design of MNs. The main types and characteristics of MNs for bone regeneration, such as mesoporous silica (mSiO2 ), mesoporous hydroxyapatite (mHAP), mesoporous calcium phosphates (mCaPs) are introduced. Additionally, the relationship between the cargo release mechanisms and bone regeneration of MNs-based nanocarriers is elucidated in detail. Particularly, MNs-based smart cargo transport strategies such as sustained cargo release, stimuli-responsive (e.g., pH, photo, ultrasound, and multi-stimuli) controllable delivery, and specific bone-targeted therapy for bone disease diagnosis and treatment are analyzed and discussed in depth. Lastly, the conclusions and outlook about the design and development of MNs-based cargo delivery systems in diagnosis and treatment for bone tissue engineering are provided to inspire new ideas and attract researchers' attention from multidisciplinary areas spanning chemistry, materials science, and biomedicine.
Collapse
Affiliation(s)
- Panpan Pan
- Department of Chemistry, Department of Gastroenterology, Zhongshan Hospital of Fudan University, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Qin Yue
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610051, China
| | - Juan Li
- Department of Chemistry, Department of Gastroenterology, Zhongshan Hospital of Fudan University, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Meiqi Gao
- Department of Chemistry, Department of Gastroenterology, Zhongshan Hospital of Fudan University, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Xuanyu Yang
- Department of Chemistry, Department of Gastroenterology, Zhongshan Hospital of Fudan University, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Yuan Ren
- Department of Chemistry, Department of Gastroenterology, Zhongshan Hospital of Fudan University, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Xiaowei Cheng
- Department of Chemistry, Department of Gastroenterology, Zhongshan Hospital of Fudan University, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Penglei Cui
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yonghui Deng
- Department of Chemistry, Department of Gastroenterology, Zhongshan Hospital of Fudan University, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
- State Key Lab of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| |
Collapse
|
45
|
Ye Q, Wang Y, Shen S, Xu C, Wang J. Biomaterials-Based Delivery of Therapeutic Antibodies for Cancer Therapy. Adv Healthc Mater 2021; 10:e2002139. [PMID: 33870637 DOI: 10.1002/adhm.202002139] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/05/2021] [Indexed: 12/19/2022]
Abstract
Considerable breakthroughs in the treatment of malignant tumors using antibody drugs, especially immunomodulating monoclonal antibodies (mAbs), have been made in the past decade. Despite technological advancements in antibody design and manufacture, multiple challenges face antibody-mediated cancer therapy, such as instability in vivo, poor tumor penetration, limited response rate, and undesirable off-target cytotoxicity. In recent years, an increasing number of biomaterials-based delivery systems have been reported to enhance the antitumor efficacy of antibody drugs. This review summarizes the advances and breakthroughs in integrating biomaterials with therapeutic antibodies for enhanced cancer therapy. A brief introduction to the principal mechanism of antibody-based cancer therapy is first established, and then various antibody immobilization strategies are provided. Finally, the current state-of-the-art in biomaterials-based antibody delivery systems and their applications in cancer treatment are summarized, highlighting how the delivery systems augment the therapeutic efficacy of antibody drugs. The outlook and perspective on biomaterials-based delivery of antitumor antibodies are also discussed.
Collapse
Affiliation(s)
- Qian‐Ni Ye
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
| | - Yue Wang
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- Shenzhen Bay Laboratory Shenzhen 518132 P. R. China
| | - Song Shen
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education South China University of Technology Guangzhou 510006 P. R. China
| | - Cong‐Fei Xu
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education South China University of Technology Guangzhou 510006 P. R. China
| | - Jun Wang
- School of Biomedical Sciences and Engineering South China University of Technology Guangzhou International Campus Guangzhou 511442 P. R. China
- National Engineering Research Center 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 Center for Tissue Restoration and Reconstruction South China University of Technology Guangzhou 510006 P. R. China
| |
Collapse
|
46
|
Wang Y, Li Z, Hu Q. Emerging self-regulated micro/nano drug delivery devices: A step forward towards intelligent diagnosis and therapy. NANO TODAY 2021; 38:101127. [DOI: 10.1016/j.nantod.2021.101127] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
47
|
Zhang Q, Zhang J, Song J, Liu Y, Ren X, Zhao Y. Protein-Based Nanomedicine for Therapeutic Benefits of Cancer. ACS NANO 2021; 15:8001-8038. [PMID: 33900074 DOI: 10.1021/acsnano.1c00476] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Proteins, a type of natural biopolymer that possess many prominent merits, have been widely utilized to engineer nanomedicine for fighting against cancer. Motivated by their ever-increasing attention in the scientific community, this review aims to provide a comprehensive showcase on the current landscape of protein-based nanomedicine for cancer therapy. On the basis of role differences of proteins in nanomedicine, protein-based nanomedicine engineered with protein therapeutics, protein carriers, enzymes, and composite proteins is introduced. The cancer therapeutic benefits of the protein-based nanomedicine are also discussed, including small-molecular therapeutics-mediated therapy, macromolecular therapeutics-mediated therapy, radiation-mediated therapy, reactive oxygen species-mediated therapy, and thermal effect-mediated therapy. Lastly, future developments and potential challenges of protein-based nanomedicine are elucidated toward clinical translation. It is believed that protein-based nanomedicine will play a vital role in the battle against cancer. We hope that this review will inspire extensive research interests from diverse disciplines to further push the developments of protein-based nanomedicine in the biomedical frontier, contributing to ever-greater medical advances.
Collapse
Affiliation(s)
- Qiuhong Zhang
- International Joint Research Center for Molecular Science, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Junmin Zhang
- International Joint Research Center for Molecular Science, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jun Song
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Yizhen Liu
- International Joint Research Center for Molecular Science, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xiangzhong Ren
- International Joint Research Center for Molecular Science, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Yanli Zhao
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore
| |
Collapse
|
48
|
Synthesis and biological evaluation of fluorescent hyaluronic acid modified amorphous calcium phosphate drug carriers for tumor-targeting. Int J Biol Macromol 2021; 182:1445-1454. [PMID: 34015404 DOI: 10.1016/j.ijbiomac.2021.05.068] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/29/2021] [Accepted: 05/10/2021] [Indexed: 12/18/2022]
Abstract
Cancer is becoming a major threat to national public health security. The integration of disease diagnosis and monitoring with treatment has become a hot spot for researchers. The amorphous calcium phosphate (ACP) nanoparticles prepared by the group in the previous stage could not precisely treat the lesion without tumor targeting and imaging characteristics. In this paper, water-soluble hyaluronic acid fluorescent carbon nanoparticles (HA-FCNs) were prepared and co-interacting with ACP nanoparticles to form hyaluronic acid fluorescent carbon/amorphous calcium phosphate (HA-FCNs/ACP) nanoparticles. The basic characteristics were characterized and the biological characteristics before and after drug loading were evaluated. HA-FCNs/ACP nanoparticles have good hemocompatibility, pH responsiveness, and enzymatic release. HA-FCNs and HA-FCNs/ACP nanoparticles are dispersed in the cytoplasm through the overexpressed CD44 receptors, which are actively targeted into A549 cells. Besides, the migration of A549 cells would be inhibited after cells were treated with drug-loaded nanomaterials. Therefore, the as-prepared nanoparticles can be used to monitor and treat focal sites through tumor-targeting bioimaging, pH-responsive, and enzymatic drug release properties, thus enabling integrated diagnosis and treatment.
Collapse
|
49
|
Huang Y, Xie D, Gou S, Canup BSB, Zhang G, Dai F, Li C, Xiao B. Quadruple-responsive nanoparticle-mediated targeted combination chemotherapy for metastatic breast cancer. NANOSCALE 2021; 13:5765-5779. [PMID: 33704300 DOI: 10.1039/d0nr08579k] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The synergism of combination chemotherapy can only be achieved under specific drug ratios. Herein, hyaluronic acid (HA)-functionalized regenerated silk fibroin-based nanoparticles (NPs) were used to concurrently deliver curcumin (CUR) and 5-fluorouracil (5-FU) at various weight ratios (3.3 : 1, 1.6 : 1, 1.1 : 1, 1 : 1, and 1 : 1.2) to breast tumor cells. The generated HA-CUR/5-FU-NPs were found to have desirable particle sizes (around 200 nm), narrow size distributions, and negative zeta potentials (about -26.0 mV). Interestingly, these NPs showed accelerated drug release rates when they were exposed to buffers that mimicked the multi-hallmarks in the tumor microenvironment (pH/hydrogen peroxide/glutathione/hyaluronidase). The surface functionalization of NPs with HA endowed them with in vitro and in vivo breast tumor-targeting properties. Furthermore, we found that the co-loading of CUR and 5-FU in HA-functionalized NPs exhibited obvious synergistic anti-cancer, pro-apoptotic, and anti-migration effects, and the strongest synergism was found at the CUR/5-FU weight ratio of 1 : 1.2. Most importantly, mice experiments revealed that HA-CUR/5-FU-NPs (1 : 1.2) showed a superior anti-cancer activity against metastatic breast cancer compared to the single drug-loaded NPs and non-functionalized CUR/5-FU-NPs (1 : 1.2). Collectively, these results demonstrate that HA-CUR/5-FU-NPs (1 : 1.2) can be exploited as a robust nanococktail for the treatment of breast cancer and its lung metastasis.
Collapse
Affiliation(s)
- Yamei Huang
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Beibei, Chongqing 400715, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Wang Y, Li S, Wang X, Chen Q, He Z, Luo C, Sun J. Smart transformable nanomedicines for cancer therapy. Biomaterials 2021; 271:120737. [PMID: 33690103 DOI: 10.1016/j.biomaterials.2021.120737] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023]
Abstract
Despite that great progression has been made in nanoparticulate drug delivery systems (nano-DDS), multiple drug delivery dilemmas still impair the delivery efficiency of nanomedicines. Rational design of smart transformable nano-DDS based on the in vivo drug delivery process represents a promising strategy for overcoming delivery obstacle of nano-DDS. In recent years, tremendous efforts have been devoted to developing smart transformable anticancer nanomedicines. Herein, we provide a review to outline the advances in this emerging field. First, smart size-reducible nanoparticles (NPs) for deep tumor penetration are summarized, including carrier degradation-induced, protonation-triggered and photobleaching-induced size reduction. Second, emerging transformable nanostructures for various therapeutic applications are discussed, including prolonging tumor retention, reversing drug-resistance, inhibiting tumor metastasis, preventing tumor recurrence and non-pharmaceutical therapy. Third, shell-detachable nanocarriers are introduced, focusing on chemical bonds breaking-initiated, charge repulsion-mediated and exogenous stimuli-triggered shell detachment approaches. Finally, the future perspectives and challenges of transformable nanomedicines in clinical cancer therapy are highlighted.
Collapse
Affiliation(s)
- Yuequan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Shumeng Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Xinhui Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Qin Chen
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, PR China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| |
Collapse
|