1
|
He Y, Sun H, Bao H, Hou J, Zhou Q, Wu F, Wang X, Sun M, Shi J, Tang G, Bai H. A natural adhesive-based nanomedicine initiates photothermal-directed in situ immunotherapy with durability and maintenance. Biomaterials 2025; 312:122751. [PMID: 39121726 DOI: 10.1016/j.biomaterials.2024.122751] [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: 05/09/2024] [Revised: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Tumor immunotherapies have emerged as a promising frontier in the realm of cancer treatment. However, challenges persist in achieving localized, durable immunostimulation while counteracting the tumor's immunosuppressive environment. Here, we develop a natural mussel foot protein-based nanomedicine with spatiotemporal control for tumor immunotherapy. In this nanomedicine, an immunoadjuvant prodrug and a photosensitizer are integrated, which is driven by their dynamic bonding and non-covalent assembling with the protein carrier. Harnessing the protein carrier's bioadhesion, this nanomedicine achieves a drug co-delivery with spatiotemporal precision, by which it not only promotes tumor photothermal ablation but also broadens tumor antigen repertoire, facilitating in situ immunotherapy with durability and maintenance. This nanomedicine also modulates the tumor microenvironment to overcome immunosuppression, thereby amplifying antitumor responses against tumor progression. Our strategy underscores a mussel foot protein-derived design philosophy of drug delivery aimed at refining combinatorial immunotherapy, offering insights into leveraging natural proteins for cancer treatment.
Collapse
Affiliation(s)
- Yunhong He
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, 310028 Hangzhou, PR China
| | - Hong Sun
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, 310028 Hangzhou, PR China
| | - Hanxiao Bao
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, 310028 Hangzhou, PR China
| | - Jue Hou
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, 310028 Hangzhou, PR China
| | - Qiaomei Zhou
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, 310028 Hangzhou, PR China; Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 310028 Hangzhou, PR China
| | - Fan Wu
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, 310028 Hangzhou, PR China; Department of Neurosurgery, First Affiliated Hospital, School of Medicine, Zhejiang University, 310028 Hangzhou, PR China
| | | | - Mingli Sun
- Zhejiang Laboratory, 311100 Hangzhou, PR China
| | - Junhui Shi
- Zhejiang Laboratory, 311100 Hangzhou, PR China
| | - Guping Tang
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, 310028 Hangzhou, PR China; Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 310028 Hangzhou, PR China
| | - Hongzhen Bai
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, 310028 Hangzhou, PR China.
| |
Collapse
|
2
|
Nasir A, Rehman MU, Khan T, Husn M, Khan M, Khan A, Nuh AM, Jiang W, Farooqi HMU, Bai Q. Advances in nanotechnology-assisted photodynamic therapy for neurological disorders: a comprehensive review. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:84-103. [PMID: 38235991 DOI: 10.1080/21691401.2024.2304814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/03/2024] [Indexed: 01/19/2024]
Abstract
Neurological disorders such as neurodegenerative diseases and nervous system tumours affect more than one billion people throughout the globe. The physiological sensitivity of the nervous tissue limits the application of invasive therapies and leads to poor treatment and prognosis. One promising solution that has generated attention is Photodynamic therapy (PDT), which can potentially revolutionise the treatment landscape for neurological disorders. PDT attracted substantial recognition for anticancer efficacy and drug conjugation for targeted drug delivery. This review thoroughly explained the basic principles of PDT, scientific interventions and advances in PDT, and their complicated mechanism in treating brain-related pathologies. Furthermore, the merits and demerits of PDT in the context of neurological disorders offer a well-rounded perspective on its feasibility and challenges. In conclusion, this review encapsulates the significant potential of PDT in transforming the treatment landscape for neurological disorders, emphasising its role as a non-invasive, targeted therapeutic approach with multifaceted applications.
Collapse
Affiliation(s)
- Abdul Nasir
- Medical Research Center, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mujeeb Ur Rehman
- Department of Zoology, Islamia College University, Peshawar, Pakistan
| | - Tamreez Khan
- Department of Zoology, Abdul Wali Khan University, Mardan, Pakistan
| | - Mansoor Husn
- Department of Biochemistry, Abdul Wali Khan University, Mardan, Pakistan
| | - Manzar Khan
- Department of Zoology, Hazara University Mansehra, Mansehra, Pakistan
| | - Ahmad Khan
- Department of Psychology, University of Karachi, Karachi, Pakistan
| | - Abdifatah Mohamed Nuh
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Jiang
- Medical Research Center, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | - Qain Bai
- Medical Research Center, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Pathania H, Chauhan P, Chaudhary V, Khosla A, Neetika, Kumar S, Gaurav, Sharma M. Engineering core-shell mesoporous silica and Fe 3O 4@Au nanosystems for targeted cancer therapeutics: a review. Biotechnol Genet Eng Rev 2024; 40:3653-3681. [PMID: 36444150 DOI: 10.1080/02648725.2022.2147685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/08/2022] [Indexed: 11/30/2022]
Abstract
The extensive utilization of nanoparticles in cancer therapies has inspired a new field of study called cancer nanomedicine. In contrast to traditional anticancer medications, nanomedicines offer a targeted strategy that eliminates side effects and has high efficacy. With its vast surface area, variable pore size, high pore volume, abundant surface chemistry and specific binding affinity, mesoporous silica nanoparticles (MPSNPs) are a potential candidate for cancer diagnosis and treatment. However, there are several bottlenecks associated with nanoparticles, including specific toxicity or affinity towards particular body fluid, which can cater by architecting core-shell nanosystems. The core-shell chemistries, synergistic effects, and interfacial heterojunctions in core-shell nanosystems enhance their stability, catalytic and physicochemical attributes, which possess high performance in cancer therapeutics. This review article summarizes research and development dedicated to engineering mesoporous core-shell nanosystems, especially silica nanoparticles and Fe3O4@Au nanoparticles, owing to their unique physicochemical characteristics. Moreover, it highlights state-of-the-art magnetic and optical attributes of Fe3O4@Au and MPSNP-based cancer therapy strategies. It details the designing of Fe3O4@Au and MPSN to bind with drugs, receptors, ligands, and destroy tumour cells and targeted drug delivery. This review serves as a fundamental comprehensive structure to guide future research towards prospects of core-shell nanosystems based on Fe3O4@Au and MPSNP for cancer theranostics.
Collapse
Affiliation(s)
- Himani Pathania
- Department of Botany, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Priyanka Chauhan
- Department of Botany, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Vishal Chaudhary
- Research Cell and Physics Department, Bhagini Nivedita College, University of Delhi, Delhi, India
| | - Ajit Khosla
- Department of Applied Chemistry, School of Advanced Materials and Nanotechnology, Xidian University, PR China
| | - Neetika
- Department of Botany, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Sunil Kumar
- Department of Animal Sciences, Central University of Himachal Pradesh, Shahpur, India
| | - Gaurav
- Department of Botany, Ramjas College, University of Delhi, Delhi, India
| | - Mamta Sharma
- Department of Botany, Shoolini University of Biotechnology and Management Sciences, Solan, India
- Department of Botany, Vivekananda Bhawan, Sardar Patel University, Mandi, India
| |
Collapse
|
4
|
Liu Y, Li J, Guo H, Fang C, Yang Q, Qin W, Wang H, Xian Y, Yan X, Yin B, Zhang K. Nanomaterials for stroke diagnosis and treatment. iScience 2024; 27:111112. [PMID: 39502285 PMCID: PMC11536039 DOI: 10.1016/j.isci.2024.111112] [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] [Indexed: 11/08/2024] Open
Abstract
Nanomaterials and nanotechnology innovations possess unique physicochemical properties that present new opportunities in the realm of stroke detection, diagnosis, and treatment. This comprehensive review explores the utilization of nanomaterials in the diagnosis and treatment of strokes, encompassing recent advancements in computed tomography (CT), magnetic resonance imaging (MRI) and magnetic particle imaging (MPI), as well as groundbreaking applications of nanomaterials and bionanomaterials in drug delivery systems and brain tissue repair. Additionally, this review meticulously examines significant challenges such as biocompatibility toxicity and long-term safety, proposing potential strategies to surmount these obstacles. Moreover, this review delves into the application of nanomaterials to improve the clinical diagnosis of stroke patients, elucidates the potential of nanotechnology in post-stroke therapy, and identifies future research directions and potential clinical applications.
Collapse
Affiliation(s)
- Yang Liu
- Department of Oncology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of Pharmacy and Central Laboratory, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu 610072, China
| | - Junying Li
- Instrumentation and Service Center for Science and Technology, Beijing Normal University, No. 18 Jinfeng Road, Zhuhai 519087, Guangdong Province, China
| | - Huaijuan Guo
- Department of Pharmacy and Central Laboratory, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu 610072, China
| | - Chao Fang
- Department of Pharmacy and Central Laboratory, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu 610072, China
| | - Qiaoling Yang
- Department of Pharmacy and Central Laboratory, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu 610072, China
| | - Wen Qin
- Department of Pharmacy and Central Laboratory, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu 610072, China
| | - Hai Wang
- Department of Pharmacy and Central Laboratory, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu 610072, China
| | - Yong Xian
- Department of Pharmacy and Central Laboratory, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu 610072, China
| | - Xuebing Yan
- Department of Oncology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Provincial Innovation and Practice Base for Postdoctors, Suining People’s Hospital, Affiliated Hospital of Xuzhou Medical University, Suining 221200, China
| | - Binxu Yin
- Department of Pharmacy and Central Laboratory, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu 610072, China
| | - Kun Zhang
- Department of Pharmacy and Central Laboratory, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu 610072, China
| |
Collapse
|
5
|
Wu K, Wang JP, Natekar NA, Ciannella S, González-Fernández C, Gomez-Pastora J, Bao Y, Liu J, Liang S, Wu X, Nguyen T Tran L, Mercedes Paz González K, Choe H, Strayer J, Iyer PR, Chalmers J, Chugh VK, Rezaei B, Mostufa S, Tay ZW, Saayujya C, Huynh Q, Bryan J, Kuo R, Yu E, Chandrasekharan P, Fellows B, Conolly S, Hadimani RL, El-Gendy AA, Saha R, Broomhall TJ, Wright AL, Rotherham M, El Haj AJ, Wang Z, Liang J, Abad-Díaz-de-Cerio A, Gandarias L, Gubieda AG, García-Prieto A, Fdez-Gubieda ML. Roadmap on magnetic nanoparticles in nanomedicine. NANOTECHNOLOGY 2024; 36:042003. [PMID: 39395441 PMCID: PMC11539342 DOI: 10.1088/1361-6528/ad8626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 07/11/2024] [Accepted: 10/12/2024] [Indexed: 10/14/2024]
Abstract
Magnetic nanoparticles (MNPs) represent a class of small particles typically with diameters ranging from 1 to 100 nanometers. These nanoparticles are composed of magnetic materials such as iron, cobalt, nickel, or their alloys. The nanoscale size of MNPs gives them unique physicochemical (physical and chemical) properties not found in their bulk counterparts. Their versatile nature and unique magnetic behavior make them valuable in a wide range of scientific, medical, and technological fields. Over the past decade, there has been a significant surge in MNP-based applications spanning biomedical uses, environmental remediation, data storage, energy storage, and catalysis. Given their magnetic nature and small size, MNPs can be manipulated and guided using external magnetic fields. This characteristic is harnessed in biomedical applications, where these nanoparticles can be directed to specific targets in the body for imaging, drug delivery, or hyperthermia treatment. Herein, this roadmap offers an overview of the current status, challenges, and advancements in various facets of MNPs. It covers magnetic properties, synthesis, functionalization, characterization, and biomedical applications such as sample enrichment, bioassays, imaging, hyperthermia, neuromodulation, tissue engineering, and drug/gene delivery. However, as MNPs are increasingly explored forin vivoapplications, concerns have emerged regarding their cytotoxicity, cellular uptake, and degradation, prompting attention from both researchers and clinicians. This roadmap aims to provide a comprehensive perspective on the evolving landscape of MNP research.
Collapse
Affiliation(s)
- Kai Wu
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX, United States of America
| | - Jian-Ping Wang
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN, United States of America
| | | | - Stefano Ciannella
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX, United States of America
| | - Cristina González-Fernández
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX, United States of America
- Department of Chemical and Biomolecular Engineering, University of Cantabria, Santander, Spain
| | - Jenifer Gomez-Pastora
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX, United States of America
| | - Yuping Bao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, United States of America
| | - Jinming Liu
- Western Digital Corporation, San Jose, CA, United States of America
| | - Shuang Liang
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, United States of America
| | - Xian Wu
- William G Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States of America
| | - Linh Nguyen T Tran
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX, United States of America
| | | | - Hyeon Choe
- William G Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States of America
| | - Jacob Strayer
- William G Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States of America
| | - Poornima Ramesh Iyer
- William G Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States of America
| | - Jeffrey Chalmers
- William G Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States of America
| | - Vinit Kumar Chugh
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN, United States of America
| | - Bahareh Rezaei
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX, United States of America
| | - Shahriar Mostufa
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX, United States of America
| | - Zhi Wei Tay
- National Institute of Advanced Industrial Science and Technology (AIST), Health and Medical Research Institute, Tsukuba, Ibaraki 305-8564, Japan
| | - Chinmoy Saayujya
- Department of Electrical Engineering and Computer Sciences, University of California Berkeley, Berkeley, CA, United States of America
| | - Quincy Huynh
- Department of Electrical Engineering and Computer Sciences, University of California Berkeley, Berkeley, CA, United States of America
| | - Jacob Bryan
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, United States of America
| | - Renesmee Kuo
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, United States of America
| | - Elaine Yu
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, United States of America
| | - Prashant Chandrasekharan
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, United States of America
| | | | - Steven Conolly
- Department of Electrical Engineering and Computer Sciences, University of California Berkeley, Berkeley, CA, United States of America
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, United States of America
| | - Ravi L Hadimani
- Department of Mechanical and Nuclear Engineering, Virginia Commonwealth University, Richmond, VA, United States of America
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, United States of America
- Department of Psychiatry, Harvard Medical School, Harvard University, Boston, MA, United States of America
| | - Ahmed A El-Gendy
- Department of Physics, University of Texas at El Paso, El Paso, TX, United States of America
| | - Renata Saha
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN, United States of America
| | - Thomas J Broomhall
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Abigail L Wright
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Michael Rotherham
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, Institute of Translational Medicine, Birmingham, United Kingdom
| | - Alicia J El Haj
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, United Kingdom
- National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, Institute of Translational Medicine, Birmingham, United Kingdom
| | - Zhiyi Wang
- Spin-X Institute, School of Chemistry and Chemical Engineering, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou, Guangdong Province, People’s Republic of China
| | - Jiarong Liang
- Spin-X Institute, School of Chemistry and Chemical Engineering, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou, Guangdong Province, People’s Republic of China
| | - Ana Abad-Díaz-de-Cerio
- Dpto. Inmunología, Microbiología y Parasitología, Universidad del País Vasco–UPV/EHU, Leioa, Spain
| | - Lucía Gandarias
- Bioscience and Biotechnology Institute of Aix-Marseille (BIAM), Aix-Marseille Université, CNRS, CEA—UMR 7265, Saint-Paul-lez-Durance, France
- Dpto. Electricidad y Electrónica, Universidad del País Vasco—UPV/EHU, Leioa, Spain
| | - Alicia G Gubieda
- Dpto. Inmunología, Microbiología y Parasitología, Universidad del País Vasco–UPV/EHU, Leioa, Spain
| | - Ana García-Prieto
- Dpto. Física Aplicada, Universidad del País Vasco–UPV/EHU, Bilbao, Spain
| | | |
Collapse
|
6
|
Dutta D, Pajaniradje S, Nair AS, Chandramohan S, Bhat SA, Manikandan E, Rajagopalan R. An in-vitro study of active targeting & anti-cancer effect of folic acid conjugated chitosan encapsulated indole curcumin analogue nanoparticles. Int J Biol Macromol 2024; 282:136990. [PMID: 39505180 DOI: 10.1016/j.ijbiomac.2024.136990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 10/18/2024] [Accepted: 10/26/2024] [Indexed: 11/08/2024]
Abstract
Natural compounds like Curcumin with anti-cancer, anti-inflammatory and anti-bacterial properties are good target for drug development but its poor aqueous solubility, bioavailability, and low retention properties makes it a poor drug candidate in clinical settings. Here in this study, we have used an indole curcumin analogue (ICA) that has better bioavailability and enhanced permeability and retention (EPR) effect than curcumin. To make an active targeting drug we have designed folic acid conjugated chitosan-based nanoparticles encapsulating Indole curcumin analogue (CS-FA-ICA-np). The physical characteristics of CS-FA-ICA-np were evaluated by DLS, SEM, FTIR, XPS, XRD and TGA. Anti-cancer activity was analyzed using MTT, Fluorescence staining, Flow cytometry, comet assay, DNA fragmentation assay, wound healing, gelatin zymography, chick chorioallantoic membrane (CAM) assay and hemolysis assay. The size of CS-FA-ICA-nps were found to be 111 nm, and spherical in shape as observed in SEM. In-vitro assays show that CS-FA-ICA np has IC50 of 90 μg/mL in MDA-MB-231, increases ROS concentration, arrests cell cycle in G2-M phase, reduces matrix metalloproteinase-9 (MMP-9) activity and initiates apoptosis in cancer cells. Our results indicate that encapsulation of ICA increases its anti-cancer effect, drug stability, enhanced drug delivery to cancer microenvironment.
Collapse
Affiliation(s)
- Dipranil Dutta
- Department of Biochemistry and Molecular Biology, School of Life Science, Pondicherry University, Puducherry 605014, India
| | - Sankar Pajaniradje
- Department of Biochemistry and Molecular Biology, School of Life Science, Pondicherry University, Puducherry 605014, India
| | - Anjali Suresh Nair
- Department of Biochemistry and Molecular Biology, School of Life Science, Pondicherry University, Puducherry 605014, India
| | - Sathyapriya Chandramohan
- Department of Biochemistry and Molecular Biology, School of Life Science, Pondicherry University, Puducherry 605014, India
| | - Suhail Ahmad Bhat
- Department of Biochemistry and Molecular Biology, School of Life Science, Pondicherry University, Puducherry 605014, India
| | - E Manikandan
- Centre for Nano Sciences and Technology, Madanjeet School of Green Energy Technologies, Pondicherry University, Puducherry 605014, India
| | - Rukkumani Rajagopalan
- Department of Biochemistry and Molecular Biology, School of Life Science, Pondicherry University, Puducherry 605014, India.
| |
Collapse
|
7
|
Raveendran A, Ser J, Park SH, Jang P, Choi HS, Cho H. Lysosome-Targeted Bifunctional Therapeutics Induce Autodynamic Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401424. [PMID: 39231370 PMCID: PMC11538690 DOI: 10.1002/advs.202401424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 08/01/2024] [Indexed: 09/06/2024]
Abstract
Autodynamic cancer therapy possesses tremendous potential for enhancing therapeutic efficacy by initiating the treatment process autonomously within targeted cells. However, challenges related to biocompatibility and targeted delivery have hindered its clinical translation owing to the induction of adverse effects and cytotoxicity in healthy cells. In this study, a novel approach for auto-initiated dynamic therapy by conjugating zwitterionic near-infrared fluorophores to a cell-penetrating peptide is proposed. This enables efficient cellular uptake and specific targeting of therapy to desired cells while avoiding off-target uptake. The zwitterionic bioconjugate causes cancer-specific toxicity following its internalization into the targeted cells, triggered by specific intracellular conditions in lysosomes. This innovative approach enables selective targeting of lysosomes in malignant cells while minimizing cytotoxic effects on normal cells. By targeting lysosomes, the method overcomes inherent risks and side effects associated with conventional cancer treatments, offering a selective and effective approach to cancer therapy.
Collapse
Affiliation(s)
- Athira Raveendran
- Department of Materials Science and EngineeringChonnam National UniversityGwangju61186Republic of Korea
| | - Jinhui Ser
- Department of Materials Science and EngineeringChonnam National UniversityGwangju61186Republic of Korea
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Seung Hun Park
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Paul Jang
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Hak Soo Choi
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Hoonsung Cho
- Department of Materials Science and EngineeringChonnam National UniversityGwangju61186Republic of Korea
| |
Collapse
|
8
|
Zhou Y, Jin Y, Wu T, Wang Y, Dong Y, Chen P, Hu C, Pan N, Ye C, Shen L, Lin M, Fang T, Wu R. New insights on mitochondrial heteroplasmy observed in ovarian diseases. J Adv Res 2024; 65:211-226. [PMID: 38061426 PMCID: PMC11519015 DOI: 10.1016/j.jare.2023.11.033] [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: 09/14/2023] [Revised: 10/26/2023] [Accepted: 11/29/2023] [Indexed: 01/01/2024] Open
Abstract
BACKGROUND The reportedly high mutation rate of mitochondrial DNA (mtDNA) may be attributed to the absence of histone protection and complete repair mechanisms. Mitochondrial heteroplasmy refers to the coexistence of wild-type and mutant mtDNA. Most healthy individuals carry a low point mutation load (<1 %) in their mtDNA, typically without any discernible phenotypic effects. However, as it exceeds a certain threshold, it may cause the onset of various diseases. Since the ovary is a highly energy-intensive organ, it relies heavily on mitochondrial function. Mitochondrial heteroplasmy can potentially contribute to a variety of significant ovarian disorders. AIM OF REVIEW In this review, we have elucidated the close relationship between mtDNA heteroplasmy and ovarian diseases, and summarized novel avenues and strategies for the potential treatment of these ovarian diseases. KEY SCIENTIFIC CONCEPTS OF REVIEW Mitochondrial heteroplasmy can potentially contribute to a variety of significant ovarian disorders, including polycystic ovary syndrome, premature ovarian insufficiency, and endometriosis. Current strategies related to mitochondrial heteroplasmy are untargeted and have low bioavailability. Nanoparticle delivery systems loaded with mitochondrial modulators, mitochondrial replacement/transplantation therapy, and mitochondria-targeted gene editing therapy may offer promising paths towards potentially more effective treatments for these diseases, despite ongoing challenges.
Collapse
Affiliation(s)
- Yong Zhou
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China; Women's Reproductive Health Key Laboratory of Zhejiang Province, People's Republic of China
| | - Yang Jin
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Tianyu Wu
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Yinfeng Wang
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Yuanhang Dong
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Pei Chen
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Changchang Hu
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Ningping Pan
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Chaoshuang Ye
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Li Shen
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Mengyan Lin
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Tao Fang
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Ruijin Wu
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China; Women's Reproductive Health Key Laboratory of Zhejiang Province, People's Republic of China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, People's Republic of China.
| |
Collapse
|
9
|
Meng F, Fu Y, Xie H, Wang H. Nanoparticle-assisted Targeting Delivery Technologies for Preventing Organ Rejection. Transplantation 2024; 108:2174-2185. [PMID: 38597913 DOI: 10.1097/tp.0000000000005025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Although organ transplantation is a life-saving medical procedure, the challenge of posttransplant rejection necessitates safe and effective immune modulation strategies. Nanodelivery approaches may have the potential to overcome the limitations of small-molecule immunosuppressive drugs, achieving efficacious treatment options for transplant tolerance without compromising overall host immunity. This review highlights recent advances in biomaterial-assisted formulations and technologies for targeted nanodrug delivery with transplant organ- or immune cell-level precision for treating graft rejection after transplantation. We provide an overview of the mechanism of transplantation rejection, current clinically approved immunosuppressive drugs, and their relevant limitations. Finally, we discuss the targeting principles and advantages of organ- and immune cell-specific delivery technologies. The development of biomaterial-assisted novel therapeutic strategies holds considerable promise for treating organ rejection and clinical translation.
Collapse
Affiliation(s)
- Fanchao Meng
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong Province, People's Republic of China
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Yang Fu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Haiyang Xie
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Hangxiang Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong Province, People's Republic of China
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| |
Collapse
|
10
|
Abtahi MS, Fotouhi A, Rezaei N, Akalin H, Ozkul Y, Hossein-Khannazer N, Vosough M. Nano-based drug delivery systems in hepatocellular carcinoma. J Drug Target 2024; 32:977-995. [PMID: 38847573 DOI: 10.1080/1061186x.2024.2365937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/22/2024] [Accepted: 06/02/2024] [Indexed: 06/19/2024]
Abstract
The high recurrence rate of hepatocellular carcinoma (HCC) and poor prognosis after medical treatment reflects the necessity to improve the current chemotherapy protocols, particularly drug delivery methods. Development of targeted and efficient drug delivery systems (DDSs), in all active, passive and stimuli-responsive forms for selective delivery of therapeutic drugs to the tumour site has been extended to improve efficacy and reduce the severe side effects. Recent advances in nanotechnology offer promising breakthroughs in the diagnosis, treatment and monitoring of cancer cells. In this review, the specific design of DDSs based on the different nano-particles and their surface engineering is discussed. In addition, the innovative clinical studies in which nano-based DDS was used in the treatment of HCC were highlighted.
Collapse
Affiliation(s)
- Maryam Sadat Abtahi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Alireza Fotouhi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Niloufar Rezaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hilal Akalin
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Yusuf Ozkul
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Nikoo Hossein-Khannazer
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
11
|
Bhamidipati K, Malleswara Rao Nakka N, Ahmed M, Javvaji K, Banerjee R, Puvvada N, Sesha Sainath AV, Chakravarty S. Enhancing cancer immunotherapy with mannose mimicking glycopolymer nanoparticles induced activation of Dendritic cells. Bioorg Chem 2024; 152:107711. [PMID: 39178706 DOI: 10.1016/j.bioorg.2024.107711] [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: 07/25/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024]
Abstract
Cancer immunotherapy leverages the immune system's inherent capacity to combat malignancies. However, effective stimulation of Dendritic cells (DCs) is challenging due to their limited distribution and the immune-suppressive tumor microenvironment. Thus, targeting mannose receptors, which are highly expressed on DCs, represents a promising strategy. This study investigates the development of mannose-based glycopolymer nanoparticles to induce activation of DCs through enhanced antigen presentation. A novel ABA-type triblock bioconjugated glycopolymer (PMn-b-PCL-b-PMn), which mimics mannose was synthesized. This polymer was further modified with Dihexadecyldimethylammonium bromide (DHDAB) to prepare cationic nanoparticles (CMNP) for gene delivery of pCMV-TRP2, an antigenic marker for both melanoma and glioblastoma. The immune response generated by CMNP and the CMNP-TRP2 polyplex was compared to an untreated control following subcutaneous injection in mice. Post-injection cytometric analysis revealed robust DC activation and increased T-cell populations in secondary lymphoid organs, including the spleen and lymph nodes. These findings suggest that CMNP can serve as a potent biomimicking vaccination vehicle against cancer, enhancing the immune response through targeted DCs activation.
Collapse
Affiliation(s)
- Keerti Bhamidipati
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Naga Malleswara Rao Nakka
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India; Polymers and Functional Materials and Fluoro-Agrochemicals Department, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Mariam Ahmed
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Kalpana Javvaji
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Rajkumar Banerjee
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Nagaprasad Puvvada
- Department of Oils, Lipids Sciences & Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India.
| | - Annadanam V Sesha Sainath
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India; Polymers and Functional Materials and Fluoro-Agrochemicals Department, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India.
| | - Sumana Chakravarty
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India.
| |
Collapse
|
12
|
Jing L, Zhang J, Li L, Luo S, Tang Z, Liu X, Zhong Y, Yuan M. Preparation of double-loaded bitter ginseng derivative B21-DOX liposomes co-modified with SP94 and BR2 ligand and its in vitro anti-hepatocarcinogenic effect. J Microencapsul 2024; 41:535-546. [PMID: 39150022 DOI: 10.1080/02652048.2024.2390955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/07/2024] [Indexed: 08/17/2024]
Abstract
AIM To construct a novel liposomal drug delivery system co-modified with SP94 and BR2 ligands, encapsulating both the bitter ginseng derivative B21 and doxorubicin (DOX), to achieve superior anti-tumour efficacy and reduced toxic side effects. METHODS Liposomes were prepared using an organic phase reaction method, with B21 encapsulated in the lipid phase and DOX in the aqueous phase. The liposomes were further modified with SP94 and BR2 peptides. The characterisations, cytotoxicity, and in vitro targeting effects were assessed through various methods including ultraviolet spectrophotometry, high-performance liquid chromatography, nano-size analysis, ultrafiltration centrifugation, dialysis, transmission electron microscopy, flow cytometry, Methylthiazolyldiphenyl-tetrazolium bromide assay, confocal laser scanning microscopy, transwell assay, and tumorsphere assay. RESULTS SP94/BR2-B21/DOX-LP liposomes were spherical with an average diameter of 120.87 ± 1.00 nm, a polydispersity index (PDI) of 0.223 ± 0.006, and a surface charge of -23.1 ± 1.27 mV. The encapsulation efficiencies for B21 and DOX were greater than 85% and 97% (mg/mg), respectively. The results indicated that SP94/BR2-B21/DOX-LP exhibited enhanced targeting and cytotoxicity compared to single-ligand modified and unmodified liposomes, with the combined encapsulation of B21 and DOX showing synergistic anti-hepatocarcinogenic effects. CONCLUSION SP94/BR2-B21/DOX-LP liposomes represent a promising targeted drug delivery system for hepatocellular carcinoma, offering improved membrane penetration, enhanced therapeutic efficacy, and reduced systemic toxicity.
Collapse
Affiliation(s)
- Lin Jing
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China
| | - Jiajia Zhang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China
- United Laboratories Pharmaceutical Company Ltd, Zhongshan, China
| | - Lili Li
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China
| | - Simei Luo
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China
| | - Zijun Tang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China
| | - Xu Liu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China
| | - Yonglong Zhong
- Department of Thoracic Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Mingqing Yuan
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China
| |
Collapse
|
13
|
Bazzazan MA, Fattollazadeh P, Keshavarz Shahbaz S, Rezaei N. Polymeric nanoparticles as a promising platform for treating triple-negative breast cancer: Current status and future perspectives. Int J Pharm 2024; 664:124639. [PMID: 39187034 DOI: 10.1016/j.ijpharm.2024.124639] [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: 05/31/2024] [Revised: 08/04/2024] [Accepted: 08/23/2024] [Indexed: 08/28/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer that lacks expression of estrogen, progesterone, and HER2 receptor targets for therapy. Polymeric nanoparticles help address the challenges in treating TNBC by enabling tailored and targeted drug delivery. Biocompatible polymeric nanoparticles leverage enhanced tumor permeability for site-specific accumulation and ligand-mediated active targeting to boost specificity. Controlled, sustained intratumorally release of encapsulated chemotherapies, such as paclitaxel and curcumin, improves antitumor efficacy as demonstrated through preclinical TNBC models. However, the practical application of these nanomedicines still has room for improvement. Advancing personalized nanoparticle platforms that align treatments to TNBC's expanding molecular subtypes shows promise. Expanding the polymer range through novel copolymers or drug conjugates may improve tumor penetration, stability, and drug encapsulation. Incorporating gene therapies, imaging agents, or triggering stimuli responsiveness into polymeric nanoparticles can also overcome innate and acquired drug resistance in TNBC while monitoring outcomes. This article reviews the different types of nanoparticles used to treat TNBC and the different mechanisms of nanoparticles that can deliver drugs to tumor cells. Collaboration across different disciplines aimed at developing combination therapies, immuno-oncology, tumor-targeting ligands, and translating preclinical safety/efficacy via scalable manufacturing practices is essential. Well-designed polymeric nanoparticles offer immense potential for patient-centric TNBC treatment, but continued optimization across bench to bedside efforts is critical for clinical realization and transforming patient outcomes.
Collapse
Affiliation(s)
- Mohammad Amin Bazzazan
- Student Research Committee, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Science, Qazvin, Iran
| | - Pouriya Fattollazadeh
- Student Research Committee, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Science, Qazvin, Iran
| | - Sanaz Keshavarz Shahbaz
- USERN Office, Qazvin University of Medical Science, Qazvin, Iran; Cellular and Molecular Research Center, Research Institute for Prevention of Noncommunicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran; Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
14
|
Laghrissi A, Juodėnas M, Tamulevičius T, Kunstmann C, Rubahn HG, Fiutowski J. Magnetic-assisted sequential templated self-assembly of hybrid colloid nanoparticle systems. NANOSCALE 2024. [PMID: 39431523 DOI: 10.1039/d4nr03665d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
The assembly of hybrid nanoparticles is a pioneering route for developing nanoscale functional devices, enabling breakthroughs in various fields, including electronics, photonics, energy, sensing, and biomedical applications. Here, we focus on the templated assembly of nano-sized colloidal systems using a combination of silica-coated superparamagnetic beads (MBs) and polymer-coated gold nanoparticles (AuNPs) or silver nanoparticles (AgNPs). These hybrid nanoparticles introduce new functionalities that allow them to be used as nanomachines with numerous possible applications. Using sequential capillarity-assisted particle assembly (sCAPA), we deposit MBs with activating nano-transducers made of PNIPAm@AuNPs, which respond to specific external stimuli such as temperature variations. We deposit MBs with surface-enhanced Raman scattering (SERS) AgNPs, which have the ability to detect molecules at low concentrations. A key achievement of our study is demonstrating the successful CAPA assembly of nanoparticles with ingenious surface chemistry and retrieving these assembled structures from nanotraps in a liquid medium. This ability highlights the potential of hybrid colloids in precisely targeted drug delivery systems and highly effective mobile sensors. It represents a significant leap forward in using nanotechnology to create more complex and responsive nanoscale assemblies.
Collapse
Affiliation(s)
- Ayoub Laghrissi
- Mads Clausen Institute, University of Southern Denmark, Sønderborg, Denmark.
| | - Mindaugas Juodėnas
- Institute of Materials Science, Kaunas University of Technology, K. Baršausko St. 59, Kaunas, Lietuva, Lithuania
| | - Tomas Tamulevičius
- Institute of Materials Science, Kaunas University of Technology, K. Baršausko St. 59, Kaunas, Lietuva, Lithuania
| | - Casper Kunstmann
- Mads Clausen Institute, University of Southern Denmark, Sønderborg, Denmark.
| | - Horst-Günter Rubahn
- Mads Clausen Institute, University of Southern Denmark, Sønderborg, Denmark.
| | - Jacek Fiutowski
- Mads Clausen Institute, University of Southern Denmark, Sønderborg, Denmark.
| |
Collapse
|
15
|
Li Y, Li XM, Wei LS, Ye JF. Advancements in mitochondrial-targeted nanotherapeutics: overcoming biological obstacles and optimizing drug delivery. Front Immunol 2024; 15:1451989. [PMID: 39483479 PMCID: PMC11524880 DOI: 10.3389/fimmu.2024.1451989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/19/2024] [Indexed: 11/03/2024] Open
Abstract
In recent decades, nanotechnology has significantly advanced drug delivery systems, particularly in targeting subcellular organelles, thus opening new avenues for disease treatment. Mitochondria, critical for cellular energy and health, when dysfunctional, contribute to cancer, neurodegenerative diseases, and metabolic disorders. This has propelled the development of nanomedicines aimed at precise mitochondrial targeting to modulate their function, marking a research hotspot. This review delves into the recent advancements in mitochondrial-targeted nanotherapeutics, with a comprehensive focus on targeting strategies, nanocarrier designs, and their therapeutic applications. It emphasizes nanotechnology's role in enhancing drug delivery by overcoming biological barriers and optimizing drug design for specific mitochondrial targeting. Strategies exploiting mitochondrial membrane potential differences and specific targeting ligands improve the delivery and mitochondrial accumulation of nanomedicines. The use of diverse nanocarriers, including liposomes, polymer nanoparticles, and inorganic nanoparticles, tailored for effective mitochondrial targeting, shows promise in anti-tumor and neurodegenerative treatments. The review addresses the challenges and future directions in mitochondrial targeting nanotherapy, highlighting the need for precision, reduced toxicity, and clinical validation. Mitochondrial targeting nanotherapy stands at the forefront of therapeutic strategies, offering innovative treatment perspectives. Ongoing innovation and research are crucial for developing more precise and effective treatment modalities.
Collapse
Affiliation(s)
- Yang Li
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Xiao-meng Li
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Li-si Wei
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Jun-feng Ye
- General Surgery Center, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
Desai S, Thorat P, Majumdar A. A promise of nose to brain delivery of bevacizumab intranasal sol-gel formulation substantiated in rat C6 glioma model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03536-3. [PMID: 39417842 DOI: 10.1007/s00210-024-03536-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Glioblastoma is one of the rapidly spreading cancers, with its potent malignancy often linked to pronounced angiogenesis within tumors. To mitigate this vascularization profile, bevacizumab (Avastin®), a monoclonal antibody, has been utilized for its antiangiogenic activity. However, its effectiveness is hindered by challenges in crossing the blood-brain barrier and the risk of off-target organ toxicity. Delivering drugs directly from the nose to the brain through the olfactory or trigeminal nerves bypassing the blood-brain barrier offers enhanced bioavailability and a more precise targeting strategy. To overcome these challenges, we aimed to develop bevacizumab in situ gel loaded mesoporous silica nanoparticles for intranasal delivery and further examine their pharmacokinetic and pharmacodynamic characteristics. The intranasal gel of mesoporous silica nanoparticles loaded with bevacizumab was optimized and formulated using a factorial and quality by design approach. In the case of bevacizumab mesoporous silica nanoparticles, lower particle size and most negative zeta potential were selected as quality target product profiles which is important for drug loading on the mesoporous silica nanoparticles and also transport of these nanoparticles across the nasal mucosa to the brain. A design space with a multidimensional combination of input variables and process parameters has been demonstrated to assure quality. To optimize the design space and achieve the desired quality standards, the base catalyst and surfactant concentration were chosen as the critical process parameters, while particle size and zeta potential were identified as the critical quality attributes. The novel formulation was assessed for physicochemical parameters such as particle size, zeta potential, entrapment efficiency, appearance, color, consistency, and pH. Additionally, studies on in vitro release, ex vivo permeation, stability, nasal toxicity, organ safety, and bioavailability were conducted. The efficacy study was conducted in an orthotopic murine glioblastoma rat model in which C6 Luc cells were instilled in the striatum of the rat's brain. In vivo, bioluminescence imaging of brain tumors was carried out to observe the tumor regression after treatment with the intranasal and intravenous bevacizumab formulation. Biochemical parameters and histopathology were performed for organ safety studies. The optimized intranasal formulation exhibited an average particle size of 318.8 nm and a zeta potential of - 14.7 mV for the mesoporous silica nanoparticles. The formulation also demonstrated an entrapment efficiency of 91.34% and a loading capacity of 45.67%. Further pharmacokinetic studies revealed that the optimized intranasal bevacizumab formulation achieved a significantly higher brain concentration Cmax = 147.9 ng/ml, indicating improved bioavailability compared to rats administered with intravenous bevacizumab formulation (BEVATAS®), which had a Cmax of 127.2 ng/ml. Moreover, this nanoparticle formulation entirely mitigated systemic exposure to bevacizumab. Organ safety evaluation of different biochemical parameters and histopathological analyses revealed that the intranasal bevacizumab-treated group was showing less off-target organ toxicity compared to the group treated with intravenous bevacizumab formulation. Subsequently, the efficacy of this nanosystem was evaluated in an orthotopic glioblastoma rat model, monitoring tumor growth over time through in vivo bioluminescence imaging and assessing anti-angiogenic effects. Twenty-one days post-induction, mesoporous silica nanoparticles loaded with bevacizumab in situ gel exhibited a marked reduction in average bioluminescence radiance (4.39 × 103) from day 7 (1.35 × 107) emphasizing an enhanced anti-angiogenic effect compared to the group treated with intravenous bevacizumab formulation which showed a gradual decrease in average bioluminescence radiance (4.82 × 104) from day 7 (1.42 × 107). These results suggest that the proposed novel formulation of mesoporous silica nanoparticles loaded bevacizumab in situ gel could serve as a promising avenue to enhance glioblastoma treatment efficacy, thereby potentially improving patient quality of life and survival rates significantly. Furthermore, the success of this delivery method could open new avenues for treating other neurological disorders, such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, and stroke. By providing effective brain-targeted therapies, this approach has the potential to revolutionize treatment options and improve outcomes for a broad spectrum of neurological conditions.
Collapse
Affiliation(s)
- Siddhesh Desai
- Department of Pharmacology, Bombay College of Pharmacy, Santacruz East, Mumbai, 400098, India
| | - Prajakta Thorat
- Department of Pharmacology, Bombay College of Pharmacy, Santacruz East, Mumbai, 400098, India
| | - Anuradha Majumdar
- Department of Pharmacology, Bombay College of Pharmacy, Santacruz East, Mumbai, 400098, India.
| |
Collapse
|
17
|
Tarab-Ravski D, Stotsky-Oterin L, Elisha A, Kundoor GR, Ramishetti S, Hazan-Halevy I, Haas H, Peer D. The future of genetic medicines delivered via targeted lipid nanoparticles to leukocytes. J Control Release 2024; 376:286-302. [PMID: 39401676 DOI: 10.1016/j.jconrel.2024.10.014] [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: 07/05/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Genetic medicines hold vast therapeutic potential, offering the ability to silence or induce gene expression, knock out genes, and even edit DNA fragments. Applying these therapeutic modalities to leukocytes offers a promising path for treating various conditions yet overcoming the obstacles of specific and efficient delivery to leukocytes remains a major bottleneck in their clinical translation. Lipid nanoparticles (LNPs) have emerged as the leading delivery system for nucleic acids due to their remarkable versatility and ability to improve their in vivo stability, pharmacokinetics, and therapeutic benefits. Equipping LNPs with targeting moieties can promote their specific cellular uptake and internalization to leukocytes, making targeted LNPs (tLNPs) an inseparable part of developing leukocyte-targeted gene therapy. However, despite the significant advancements in research, genetic medicines for leukocytes using targeted delivery approaches have not been translated into the clinic yet. Herein, we discuss the important aspects of designing tLNPs and highlight the considerations for choosing an appropriate bioconjugation strategy and targeting moiety. Furthermore, we provide our insights on limiting challenges and identify key areas for further research to advance these exciting therapies for patient care.
Collapse
Affiliation(s)
- Dana Tarab-Ravski
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Lior Stotsky-Oterin
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Aviad Elisha
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Govinda Reddy Kundoor
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | | | - Inbal Hazan-Halevy
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Heinrich Haas
- NeoVac Ltd. 127 Olympic Ave., OX14 4SA, Milton Park, Oxfordshire, UK; Department of Biopharmaceutics and Pharmaceutical Technology, Johannes Gutenberg-University, Mainz, Germany
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
18
|
Arshad M, Atochina-Vasserman EN, Chenna SS, Maurya DS, Shalihin MI, Sahoo D, Lewis AC, Lewis JJ, Ona N, Vasserman JA, Ni H, Park WJ, Weissman D, Percec V. Accelerated Ten-Gram-Scale Synthesis of One-Component Multifunctional Sequence-Defined Ionizable Amphiphilic Janus Dendrimer 97. Biomacromolecules 2024; 25:6871-6882. [PMID: 39361876 DOI: 10.1021/acs.biomac.4c01107] [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: 10/05/2024]
Abstract
One-component multifunctional sequence-defined ionizable amphiphilic Janus dendrimers (IAJDs) were discovered in our laboratories in 2021 to represent a new class of synthetic vectors for the targeted delivery of messenger RNA (mRNA). They coassemble with mRNA by simple injection of their ethanol solution into a pH 4 acetate buffer containing the nucleic acid into monodisperse dendrimersome nanoparticles (DNPs) with predictable dimensions. DNPs are competitive with 4-component lipid nanoparticles (LNPs), which are used in commercial COVID-19 vaccines, except that IAJDs are prepared in fewer reaction steps than each individual component of the LNPs. This simple methodology for the synthesis of IAJDs and their coassembly with mRNA into DNPs, together with the precise placement of their individual components and indefinite stability at room temperature in air, make them attractive candidates for the development of nanomedicine-based targeted mRNA delivery. Access to the large-scale synthesis of IAJDs without the need for sophisticated technologies, instrumentation, and synthetic skills is expected to open numerous new opportunities worldwide in nanomedicine. The goal of this publication is to report an accelerated ten-gram-scale synthesis of IAJD97 from inexpensive food additives obtained from renewable plant phenolic acid starting materials by methodologies accessible to any laboratory. This accelerated synthesis can be accomplished in 4 days. We expect that the work reported here will impact the field of nanomedicine in both developed and less developed countries.
Collapse
Affiliation(s)
- Mahwish Arshad
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Elena N Atochina-Vasserman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Srijay S Chenna
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Devendra S Maurya
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
| | - Muhammad Irhash Shalihin
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
| | - Dipankar Sahoo
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
| | - Alec C Lewis
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
| | - Jordan J Lewis
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
| | - Nathan Ona
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jessica A Vasserman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Houping Ni
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Wook-Jin Park
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Virgil Percec
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6323, United States
| |
Collapse
|
19
|
Mohaghegh N, Ahari A, Buttles C, Davani S, Hoang H, Huang Q, Huang Y, Hosseinpour B, Abbasgholizadeh R, Cottingham AL, Farhadi N, Akbari M, Kang H, Khademhosseini A, Jucaud V, Pearson RM, Hassani Najafabadi A. Simvastatin-Loaded Polymeric Nanoparticles: Targeting Inflammatory Macrophages for Local Adipose Tissue Browning in Obesity Treatment. ACS NANO 2024; 18:27764-27781. [PMID: 39342648 DOI: 10.1021/acsnano.4c10742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Obesity is defined as chronic, low-grade inflammation within specific tissues. Given the escalating prevalence of obesity among individuals of all ages, obesity has reached epidemic proportions, posing an important public health challenge. Despite significant advancements in treating obesity, conventional approaches remain largely ineffective or involve severe side effects, thus underscoring the pressing need to explore and develop treatment approaches. Targeted and local immunomodulation using nanoparticles (NPs) can influence fat production and utilization processes. Statins, known for their anti-inflammatory properties, show the potential for mitigating obesity-related inflammation. A localized delivery option offers several advantages over oral and parenteral delivery methods. Here, we developed simvastatin (Sim) encapsulated within PLGA NPs (Sim-NP) for localized delivery of Sim to adipose tissues (ATs) for immunomodulation to treat obesity. In vitro experiments revealed the strong anti-inflammatory effects of Sim-NPs, which resulted in enhanced modulation of macrophage (MΦ) polarization and induction of AT browning. We then extended our investigation to an in vivo mouse model of high-fat-diet (HFD)-induced obesity. Sim-NP administration led to the controlled release of Sim within AT, directly impacting MΦ activity and inducing AT browning while inducing weight loss. Our findings demonstrated that Sim-NP administration effectively inhibited the progression of obesity-related inflammation, controlled white fat production, and enhanced AT modulation. These results highlight the potential of Sim-NP as a potent nanotherapy for treating obesity by modulating the immune system.
Collapse
Affiliation(s)
- Neda Mohaghegh
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Amir Ahari
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Surgery, University of California-Los Angeles, Los Angeles, California 90095, United States
| | - Claire Buttles
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Biology, Indiana University Bloomington, Bloomington, Indiana 47405, United States
| | - Saya Davani
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Briggs Hall, Davis, California 95616, United States
| | - Hanna Hoang
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California 90024, United States
| | - Qiang Huang
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Yixuan Huang
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Bahareh Hosseinpour
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Chemistry, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| | - Reza Abbasgholizadeh
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Andrea L Cottingham
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Neda Farhadi
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Mohsen Akbari
- Department of Mechanical Engineering, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
- Biotechnology Center, Silesian University of Technology, Gliwice 44-100, Poland
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Ryan M Pearson
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | | |
Collapse
|
20
|
Ashkarran AA, Lin Z, Rana J, Bumpers H, Sempere L, Mahmoudi M. Impact of Nanomedicine in Women's Metastatic Breast Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2301385. [PMID: 37269217 PMCID: PMC10693652 DOI: 10.1002/smll.202301385] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/16/2023] [Indexed: 06/04/2023]
Abstract
Metastatic breast cancer is responsible for 90% of mortalities among women suffering from various types of breast cancers. Traditional cancer treatments such as chemotherapy and radiation therapy can cause significant side effects and may not be effective in many cases. However, recent advances in nanomedicine have shown great promise in the treatment of metastatic breast cancer. For example, nanomedicine demonstrated robust capacity in detection of metastatic cancers at early stages (i.e., before the metastatic cells leave the initial tumor site), which gives clinicians a timely option to change their treatment process (for example, instead of endocrine therapy they may use chemotherapy). Here recent advances in nanomedicine technology in the identification and treatment of metastatic breast cancers are reviewed.
Collapse
Affiliation(s)
- Ali Akbar Ashkarran
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Zijin Lin
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Jatin Rana
- Division of Hematology and Oncology, Michigan State University, East Lansing, MI, 48824, USA
| | - Harvey Bumpers
- Department of Surgery, Michigan State University, East Lansing, MI, 48824, USA
| | - Lorenzo Sempere
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, 48824, USA
- Connors Center for Women's Health & Gender Biology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
21
|
Xu Y, Dang H, Teng C, Yin D, Yan L. ATP Inhibition for Starvation/Mild Photothermal/Photodynamic Synergy Therapy Using Polypeptide Nanoparticles Conjugating 2-Deoxy-D-Glucose and Dye under NIR Phototheranostic Strategy. Adv Healthc Mater 2024; 13:e2401219. [PMID: 38758576 DOI: 10.1002/adhm.202401219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/27/2024] [Indexed: 05/18/2024]
Abstract
Rapid propagation of tumor cells requires plenty of energy, which is adenosine triphosphate (ATP) dependent. ATP inhibition in tumors not only results in the starvation of tumor cells but also down-regulation of the level of heat shock proteins (HSPs), which usually increase during traditional photothermal therapy (PTT), especially when the temperature is up 50 °C. 2-deoxy-D-glucose (2DG) is an anti-glycolytic reagent and can be used as an efficient agent for ATP inhibition in tumors. Compared with typical PTT, low-temperature mild photothermal therapy (MPTT) is receiving more and more attention because it avoids the high temperatures causing damage to the normal tissue, and the increase of HSPs which decrease PTT. Here, multifunctional polypeptide nanoparticles pDG@Ahx conjugating both a NIR probe Ahx-BDP and 2DG into the side chain of the amphiphilic polypeptide have been prepared. In vitro and in vivo studies reveal that the as-prepared nanoparticles achieve a synergistic effect of starvation/MPTT/PDT (photodynamic therapy), and it provides a new strategy to NIR-I/II fluorescence imaging-guided starvation/MPTT/PDT synergy therapy for tumors.
Collapse
Affiliation(s)
- Yixuan Xu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China. Hefei, Jinzai Road 96, Anhui, 230026, P. R. China
- Key Laboratory of Precision and Intelligent Chemistry, and Department of Chemical Physics, University of Science and Technology of China. Hefei, Jinzai road 96, Anhui, 230026, P. R. China
| | - Huiping Dang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China. Hefei, Jinzai Road 96, Anhui, 230026, P. R. China
- Key Laboratory of Precision and Intelligent Chemistry, and Department of Chemical Physics, University of Science and Technology of China. Hefei, Jinzai road 96, Anhui, 230026, P. R. China
| | - Changchang Teng
- Key Laboratory of Precision and Intelligent Chemistry, and Department of Chemical Physics, University of Science and Technology of China. Hefei, Jinzai road 96, Anhui, 230026, P. R. China
| | - Dalong Yin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China. Hefei, Jinzai Road 96, Anhui, 230026, P. R. China
| | - Lifeng Yan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China. Hefei, Jinzai Road 96, Anhui, 230026, P. R. China
- Key Laboratory of Precision and Intelligent Chemistry, and Department of Chemical Physics, University of Science and Technology of China. Hefei, Jinzai road 96, Anhui, 230026, P. R. China
| |
Collapse
|
22
|
Al-Samydai A, Abu Hajleh MN, Al-Sahlawi F, Nsairat H, Khatib AA, Alqaraleh M, Ibrahim AK. Advancements of metallic nanoparticles: A promising frontier in cancer treatment. Sci Prog 2024; 107:368504241274967. [PMID: 39370817 PMCID: PMC11459474 DOI: 10.1177/00368504241274967] [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: 10/08/2024]
Abstract
The incidence of cancer is increasing and evolving as a major source of mortality. Nanotechnology has garnered considerable scientific interest in recent decades and can offer a promising solution to the challenges encountered with traditional chemotherapy. Nanoparticle utilization holds promise in combating cancer and other diseases, offering exciting prospects for drug delivery systems and medicinal applications. Metallic nanoparticles exhibit remarkable physical and chemical properties, such as their minute size, chemical composition, structure, and extensive surface area, rendering them versatile and cost-effective. Research has demonstrated their significant and beneficial impact on cancer treatment, characterized by enhanced targeting abilities, gene activity suppression, and improved drug delivery efficiency. By incorporating targeting ligands, functionalized metal nanoparticles ensure precise energy deposition within tumors, thereby augmenting treatment accuracy. Moreover, beyond their therapeutic efficacy, metal nanoparticles serve as valuable tools for cancer cell visualization, contributing to diagnostic techniques. Utilizing metal nanoparticles in therapeutic systems allows for simultaneous cancer diagnosis and treatment, while also facilitating controlled drug release, thus revolutionizing cancer care. This narrative review investigates the advancements of metal nanoparticles in cancer treatment, types and mechanisms in targeting cancer cells, application in clinical scenarios, and potential toxicity in medicine.
Collapse
Affiliation(s)
- Ali Al-Samydai
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Maha N. Abu Hajleh
- Department of Cosmetic Science, Pharmacological and Diagnostic Research Centre, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | - Farah Al-Sahlawi
- Department of Pharmaceutics at the College of Pharmacy, University of Alkafeel, AlNajaf, Iraq
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Arwa Al Khatib
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Moath Alqaraleh
- Department of Medical Laboratory Sciences, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Alia K. Ibrahim
- Faculty of Medicine, Al-Balqa Applied University, Al-Salt, Jordan
| |
Collapse
|
23
|
Zhang X, Liang L, Wang F, Jose PA, Chen K, Zeng C. Irisin-Encapsulated Mitochondria-Targeted Biomimetic Nanotherapeutics for Alleviating Acute Kidney Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402805. [PMID: 39119832 PMCID: PMC11481180 DOI: 10.1002/advs.202402805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/30/2024] [Indexed: 08/10/2024]
Abstract
Acute kidney injury (AKI) is the sudden decrease in renal function that can be attributed to dysregulated reactive oxygen species (ROS) production and impaired mitochondrial function. Irisin, a type I membrane protein secreted by skeletal muscles in response to physical activity, has been reported to alleviate kidney damage through regulation of mitochondrial biogenesis and oxidative metabolism. In this study, a macrophage membrane-coated metal-organic framework (MCM@MOF) is developed as a nanocarrier for encapsulating irisin to overcome the inherent characteristics of irisin, including a short circulation time, limited kidney-targeting ability, and low membrane permeability. The engineered irisin-mediated biomimetic nanotherapeutics have extended circulation time and enhanced targeting capability toward injured kidneys due to the preservation of macrophage membrane proteins. The irisin-encapsulated biomimetic nanotherapeutics effectively mitigate acute ischemia-reperfusion injury by protecting mitochondrial function and modulating SOD2 levels in renal tubular epithelial cells. The present study provides novel insights to advance the development of irisin as a potential therapeutic approach for AKI.
Collapse
Affiliation(s)
- Xia Zhang
- Department of CardiologyDaping HospitalThird Military Medical University (Army Medical University)Chongqing400042P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease ResearchMinistry of Education of ChinaChongqing400042P. R. China
- Chongqing Key Laboratory for Hypertension ResearchCardiovascular Clinical Research CenterChongqing Institute of CardiologyChongqing400042P. R. China
| | - Lijia Liang
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease ResearchMinistry of Education of ChinaChongqing400042P. R. China
- Chongqing Institute of Green and Intelligent TechnologyChinese Academy of SciencesChongqing400714P. R. China
- Chongqing General HospitalChongqing401147P. R. China
| | - Fengxian Wang
- Department of CardiologyDaping HospitalThird Military Medical University (Army Medical University)Chongqing400042P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease ResearchMinistry of Education of ChinaChongqing400042P. R. China
- Chongqing Institute of Green and Intelligent TechnologyChinese Academy of SciencesChongqing400714P. R. China
| | - Pedro A. Jose
- Division of Renal Diseases & HypertensionDepartment of Medicine and Pharmacology‐PhysiologyThe George Washington University School of Medicine & Health SciencesWashington DC20037USA
| | - Ken Chen
- Department of CardiologyDaping HospitalThird Military Medical University (Army Medical University)Chongqing400042P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease ResearchMinistry of Education of ChinaChongqing400042P. R. China
- Chongqing Key Laboratory for Hypertension ResearchCardiovascular Clinical Research CenterChongqing Institute of CardiologyChongqing400042P. R. China
| | - Chunyu Zeng
- Department of CardiologyDaping HospitalThird Military Medical University (Army Medical University)Chongqing400042P. R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease ResearchMinistry of Education of ChinaChongqing400042P. R. China
- Chongqing Key Laboratory for Hypertension ResearchCardiovascular Clinical Research CenterChongqing Institute of CardiologyChongqing400042P. R. China
- Chongqing Institute of Green and Intelligent TechnologyChinese Academy of SciencesChongqing400714P. R. China
| |
Collapse
|
24
|
Geisler HC, Safford HC, Mitchell MJ. Rational Design of Nanomedicine for Placental Disorders: Birthing a New Era in Women's Reproductive Health. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2300852. [PMID: 37191231 PMCID: PMC10651803 DOI: 10.1002/smll.202300852] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/16/2023] [Indexed: 05/17/2023]
Abstract
The placenta is a transient organ that forms during pregnancy and acts as a biological barrier, mediating exchange between maternal and fetal circulation. Placental disorders, such as preeclampsia, fetal growth restriction, placenta accreta spectrum, and gestational trophoblastic disease, originate in dysfunctional placental development during pregnancy and can lead to severe complications for both the mother and fetus. Unfortunately, treatment options for these disorders are severely lacking. Challenges in designing therapeutics for use during pregnancy involve selectively delivering payloads to the placenta while protecting the fetus from potential toxic side effects. Nanomedicine holds great promise in overcoming these barriers; the versatile and modular nature of nanocarriers, including prolonged circulation times, intracellular delivery, and organ-specific targeting, can control how therapeutics interact with the placenta. In this review, nanomedicine strategies are discussed to treat and diagnose placental disorders with an emphasis on understanding the unique pathophysiology behind each of these diseases. Finally, prior study of the pathophysiologic mechanisms underlying these placental disorders has revealed novel disease targets. These targets are highlighted here to motivate the rational design of precision nanocarriers to improve therapeutic options for placental disorders.
Collapse
Affiliation(s)
- Hannah C. Geisler
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Hannah C. Safford
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19014, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| |
Collapse
|
25
|
Hameed H, Sarwar HS, Younas K, Zaman M, Jamshaid M, Irfan A, Khalid M, Sohail MF. Exploring the potential of nanomedicine for gene therapy across the physicochemical and cellular barriers. Funct Integr Genomics 2024; 24:177. [PMID: 39340586 DOI: 10.1007/s10142-024-01459-z] [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: 08/06/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024]
Abstract
After COVID-19, a turning point in the way of pharmaceutical technology is gene therapy with beneficial potential to start a new medical era. However, commercialization of such pharmaceuticals would never be possible without the help of nanotechnology. Nanomedicine can fulfill the growing needs linked to safety, efficiency, and site-specific targeted delivery of Gene therapy-based pharmaceuticals. This review's goal is to investigate how nanomedicine may be used to transfer nucleic acids by getting beyond cellular and physicochemical barriers. Firstly, we provide a full description of types of gene therapy, their mechanism, translation, transcription, expression, type, and details of diseases with possible mechanisms that can only be treated with genes-based pharmaceuticals. Additionally, we also reviewed different types of physicochemical barriers, physiological and cellular barriers in nucleic acids (DNA/RNA) based drug delivery. Finally, we highlight the need and importance of cationic lipid-based nanomedicine/nanocarriers in gene-linked drug delivery and how nanotechnology can help to overcome the above-discussed barrier in gene therapy and their biomedical applications.
Collapse
Affiliation(s)
- Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan.
| | - Hafiz Shoaib Sarwar
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Komel Younas
- Faculty of Pharmacy, University Paris Saclay, 17 Avenue des sciences, 91190, Orsay, France
| | - Muhammad Zaman
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Muhammad Jamshaid
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Ali Irfan
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Maha Khalid
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Muhammad Farhan Sohail
- Department of Chemistry, SBASSE, Lahore University of Management Sciences (LUMS), Lahore, 54000, Pakistan
- Alliant College of Pharmacy and Allied Health Sciences, Lahore, 54000, Pakistan
| |
Collapse
|
26
|
Khabirova S, Menshikov-Tonyan M, Aleshin G, Prikhodko A, Kozlov D, Anokhin E, Babeshkin K, Titchenko N, Zubenko A, Shchukina A, Fedorov Y, Kalmykov S. Assessing the biocompatibility and stability of CeO 2 nanoparticle conjugates with azacrowns for use as radiopharmaceuticals. RSC Med Chem 2024:d4md00515e. [PMID: 39345713 PMCID: PMC11428044 DOI: 10.1039/d4md00515e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/31/2024] [Indexed: 10/01/2024] Open
Abstract
The application of nanoparticles is promising for the purposes of nuclear medicine due to the possibilities of using them as vectors and transporters of radionuclides. In this study, we have successfully synthesised conjugates of CeO2 nanoparticles and azacrown ligands. Then, the radiolabelling conditions with radionuclides 65Zn, 44Sc and 207Bi were selected and the kinetic stability of the complexes in biologically significant media was evaluated. Optimum conditions for CeO2-APTES-L and CeO2-APTES-DOTA labelling were found: 0.1 g l-1 conjugate and 10-9 M metal cations at 90 °C for complexes with [65Zn]Zn2+, [44Sc]Sc3+ and [207Bi]Bi3+. CeO2-APTES-L-44Sc (radiochemical purity more than 90%) was stable in fetal bovine serum. The obtained results enabled us to choose the most promising complex for biomedical applications for carrying out in vitro and in vivo biodistribution research. Nanoceria and its derivative showed no obvious toxicity to human endothelial cells EA.hy926. Then, the in vivo stability of the studied scandium complex was demonstrated. Taken together, our studies show that functionalised cerium oxide nanoparticles lead to stable radiolabelled nanosystems that may be used for targeted drug delivery, diagnosis and treatment of oncological diseases.
Collapse
Affiliation(s)
- Sofia Khabirova
- Faculty of Chemistry, Lomonosov Moscow State University 119991 Leninskie Gory, 1/3 Moscow Russia
| | - Mikhail Menshikov-Tonyan
- A.N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences 119991, Vavilova, 28 Moscow Russia
| | - Gleb Aleshin
- Faculty of Chemistry, Lomonosov Moscow State University 119991 Leninskie Gory, 1/3 Moscow Russia
| | - Anastasia Prikhodko
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University 119991 Leninskie Gory, 1/40 Moscow Russia
- Faculty of Bioengineering and Bioinformatics 119991, Leninskie Gory, 1/73 Moscow Russia
| | - Daniil Kozlov
- Kurnakov Institute of General and Inorganic Chemistry of the Russian Academy of Sciences 119991, Leninsky Prosp. 31 Moscow Russia
| | - Evgeny Anokhin
- Faculty of Chemistry, Lomonosov Moscow State University 119991 Leninskie Gory, 1/3 Moscow Russia
- The Advanced Educational Scientific Center (AESC), Lomonosov Moscow State University 121352, Kremenchugskaya, 11 Moscow Russia
| | - Konstantin Babeshkin
- Kurnakov Institute of General and Inorganic Chemistry of the Russian Academy of Sciences 119991, Leninsky Prosp. 31 Moscow Russia
| | - Nikolay Titchenko
- Faculty of Chemistry, Lomonosov Moscow State University 119991 Leninskie Gory, 1/3 Moscow Russia
| | - Anastasia Zubenko
- A.N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences 119991, Vavilova, 28 Moscow Russia
| | - Anna Shchukina
- A.N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences 119991, Vavilova, 28 Moscow Russia
| | - Yuri Fedorov
- A.N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences 119991, Vavilova, 28 Moscow Russia
| | - Stepan Kalmykov
- Faculty of Chemistry, Lomonosov Moscow State University 119991 Leninskie Gory, 1/3 Moscow Russia
| |
Collapse
|
27
|
Duan M, Cao R, Yang Y, Chen X, Liu L, Ren B, Wang L, Goh BC. Blood-Brain Barrier Conquest in Glioblastoma Nanomedicine: Strategies, Clinical Advances, and Emerging Challenges. Cancers (Basel) 2024; 16:3300. [PMID: 39409919 PMCID: PMC11475686 DOI: 10.3390/cancers16193300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma (GBM) is a prevalent type of malignancy within the central nervous system (CNS) that is associated with a poor prognosis. The standard treatment for GBM includes the surgical resection of the tumor, followed by radiotherapy and chemotherapy; yet, despite these interventions, overall treatment outcomes remain suboptimal. The blood-brain barrier (BBB), which plays a crucial role in maintaining the stability of brain tissue under normal physiological conditions of the CNS, also poses a significant obstacle to the effective delivery of therapeutic agents to GBMs. Recent preclinical studies have demonstrated that nanomedicine delivery systems (NDDSs) offer promising results, demonstrating both effective GBM targeting and safety, thereby presenting a potential solution for targeted drug delivery. In this review, we first explore the various strategies employed in preclinical studies to overcome the BBB for drug delivery. Subsequently, the results of the clinical translation of NDDSs are summarized, highlighting the progress made. Finally, we discuss potential strategies for advancing the development of NDDSs and accelerating their translational research through well-designed clinical trials in GBM therapy.
Collapse
Affiliation(s)
- Mengyun Duan
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.D.); (X.C.)
| | - Ruina Cao
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Wuhan 430070, China;
| | - Yuan Yang
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| | - Xiaoguang Chen
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.D.); (X.C.)
| | - Lian Liu
- Department of Pharmacology, Health Science Center, Yangtze University, Jingzhou 434023, China;
| | - Boxu Ren
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.D.); (X.C.)
| | - Lingzhi Wang
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore;
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Boon-Cher Goh
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore;
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore 119228, Singapore
| |
Collapse
|
28
|
Lee JR, Kim YM, Kim EJ, Jang MK, Park SC. Advancing Breast Cancer Therapeutics: Targeted Gene Delivery Systems Unveiling the Potential of Estrogen Receptor-Targeting Ligands. Biomater Res 2024; 28:0087. [PMID: 39319107 PMCID: PMC11420687 DOI: 10.34133/bmr.0087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024] Open
Abstract
Although curcumin has been well known as a phytochemical drug that inhibits tumor promotion by modulating multiple molecular targets, its potential was not reported as a targeting ligand in the field of drug delivery system. Here, we aimed to assess the tumor-targeting efficiency of curcumin and its derivatives such as phenylalanine, cinnamic acid, coumaric acid, and ferulic acid. Curcumin exhibited a high affinity for estrogen receptors through a pull-down assay using the membrane proteins of MCF-7, a breast cancer cell line, followed by designation of a polymer-based gene therapy system. As a basic backbone for gene binding, dextran grafted with branched polyethylenimine was synthesized, and curcumin and its derivatives were linked to lysine dendrimers. In vitro and in vivo antitumor effects were evaluated using plasmid DNA expressing anti-bcl-2 short hairpin RNA. All synthesized gene carriers showed excellent DNA binding, protective effects against nuclease, and gene transfection efficiency in MCF-7 and SKBr3 breast cancer cells. Preincubation with curcumin or 17α-estradiol resulted in a marked dose-dependent decrease in gene transfer efficiency and suggested targeting specificity of curcumin. Our study indicates the potential of curcumin and its derivatives as novel targeting ligands for tumor cells and tissues.
Collapse
Affiliation(s)
- Jung Ro Lee
- National Institute of Ecology (NIE), Seocheon 33657, Korea
| | - Young-Min Kim
- Department of Chemical Engineering, College of Engineering, Sunchon National University, Suncheon, Jeonnam 57922, Korea
| | - Eun-Ji Kim
- Department of Chemical Engineering, College of Engineering, Sunchon National University, Suncheon, Jeonnam 57922, Korea
| | - Mi-Kyeong Jang
- Department of Chemical Engineering, College of Engineering, Sunchon National University, Suncheon, Jeonnam 57922, Korea
| | - Seong-Cheol Park
- Department of Chemical Engineering, College of Engineering, Sunchon National University, Suncheon, Jeonnam 57922, Korea
| |
Collapse
|
29
|
Guo L, Zhao Q, Wang M. Core-Shell Microspheres with Encapsulated Gold Nanoparticle Carriers for Controlled Release of Anti-Cancer Drugs. J Funct Biomater 2024; 15:277. [PMID: 39452576 PMCID: PMC11509066 DOI: 10.3390/jfb15100277] [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: 07/25/2024] [Revised: 08/27/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Cancer is one of the major threats to human health and lives. However, effective cancer treatments remain a great challenge in clinical medicine. As a common approach for cancer treatment, chemotherapy has saved the life of millions of people; however, patients who have gone through chemotherapy often suffer from severe side effects owing to the inherent cytotoxicity of anti-cancer drugs. Stabilizing the blood concentration of an anti-cancer drug will reduce the occurrence or severity of side effects, and relies on using an appropriate drug delivery system (DDS) for achieving sustained or even on-demand drug delivery. However, this is still an unmet clinical challenge since the mainstay of anti-cancer drugs is small molecules, which tend to be diffused rapidly in the body, and conventional DDSs exhibit the burst release phenomenon. Here, we establish a class of DDSs based on biodegradable core-shell microspheres with encapsulated doxorubicin hydrochloride-loaded gold nanoparticles (DOX@Au@MSs), with the core-shell microspheres being made of poly(lactic-co-glycolic acid) in the current study. By harnessing the physical barrier of the biodegradable shell of core-shell microspheres, DOX@Au@MSs can provide a sustained release of the anti-cancer drug in the test duration (which is 21 days in the current study). Thanks to the photothermal properties of the encapsulated gold nanoparticle carriers, the core-shell biodegradable microspheres can be ruptured through remotely controlled near-infrared (NIR) light, thereby achieving an NIR-controlled triggered release of the anti-cancer drug. Furthermore, the route of the DOX-Au@MS-enabled controlled release of the anti-cancer drug can provide durable cancer cell ablation for the long period of 72 h.
Collapse
Affiliation(s)
- Lin Guo
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China;
| | - Qilong Zhao
- Institute of Biomedical & Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518055, China
| | - Min Wang
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China;
| |
Collapse
|
30
|
Lin L, Liu Y, Tang R, Ding S, Lin H, Li H. Evodiamine: A Extremely Potential Drug Development Candidate of Alkaloids from Evodia rutaecarpa. Int J Nanomedicine 2024; 19:9843-9870. [PMID: 39345907 PMCID: PMC11430234 DOI: 10.2147/ijn.s459510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 07/23/2024] [Indexed: 10/01/2024] Open
Abstract
Evodiamine (EVO) is a tryptamine indole alkaloid and the main active ingredient in Evodia rutaecarpa. In recent years, the antitumor, cardioprotective, anti-inflammatory, and anti-Alzheimer's disease effects of EVO have been reported. EVO exerts antitumor effects by inhibiting tumor cell activity and proliferation, blocking the cell cycle, promoting apoptosis and autophagy, and inhibiting the formation of the tumor microvasculature. However, EVO has poor solubility and low bioavailability. Several derivatives with high antitumor activity have been discovered through the structural optimization of EVO, and new drug delivery systems have been developed to improve the solubility and bioavailability of EVO. Current research found that EVO could have toxic effects, such as hepatotoxicity, nephrotoxicity, and cardiac toxicity. This article reviews the pharmacological activity, derivatives, drug delivery systems, toxicity, and pharmacokinetics of EVO and provides research ideas and references for its further in-depth development and clinical applications.
Collapse
Affiliation(s)
- Longfei Lin
- Institute Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Yuling Liu
- Institute Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Ruying Tang
- Institute Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Shilan Ding
- Institute Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Hongmei Lin
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People's Republic of China
- National Medical Products Administration Key Laboratory for Research Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Hui Li
- Institute Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
- Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, People's Republic of China
| |
Collapse
|
31
|
Zheng X, Gong T, Luo W, Hu B, Gao J, Li Y, Liu R, Xie N, Yang W, Xu X, Cheng L, Zhou C, Yuan Q, Huang C, Peng X, Zhou X. Fusobacterium nucleatum extracellular vesicles are enriched in colorectal cancer and facilitate bacterial adhesion. SCIENCE ADVANCES 2024; 10:eado0016. [PMID: 39303027 DOI: 10.1126/sciadv.ado0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 08/15/2024] [Indexed: 09/22/2024]
Abstract
Fusobacterium nucleatum in colorectal cancer (CRC) tissue is implicated at multiple stages of the disease, while the mechanisms underlying bacterial translocation and colonization remain incompletely understood. Herein, we investigated whether extracellular vesicles derived from F. nucleatum (FnEVs) have impacts on bacterial colonization. In mice with colitis-related CRC, a notable enrichment of FnEVs was observed, leading to a significant increase in intratumor colonization by F. nucleatum and accelerated progression of CRC. The enrichment of FnEVs in clinical CRC tissues was demonstrated. Subsequently, we revealed that FnEVs undergo membrane fusion with CRC cells, leading to the transfer and retention of FomA on recipient cell surfaces. Given its ability to facilitate F. nucleatum autoaggregation through interaction with FN1441, the presence of FomA on CRC cell surfaces presents a target for bacterial adhesion. Collectively, the findings unveil a mechanism used by EVs to prepare a niche conducive for bacterial colonization in distal organs.
Collapse
Affiliation(s)
- Xin Zheng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
- Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Tao Gong
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Wanyi Luo
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
- Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Bing Hu
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - Jinhang Gao
- Laboratory of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - Yuqing Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Rui Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Na Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, P.R. China
| | - Wenming Yang
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - Xin Xu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
- Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
- Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, P.R. China
| | - Xian Peng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
- Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| |
Collapse
|
32
|
Li X, Hu Y, Zhang X, Shi X, Parak WJ, Pich A. Transvascular transport of nanocarriers for tumor delivery. Nat Commun 2024; 15:8172. [PMID: 39289401 PMCID: PMC11408679 DOI: 10.1038/s41467-024-52416-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
Nanocarriers (NCs) play a crucial role in delivering theranostic agents to tumors, making them a pivotal focus of research. However, the persistently low delivery efficiency of engineered NCs has been a significant challenge in the advancement of nanomedicine, stirring considerable debate. Transvascular transport is a critical pathway for NC delivery from vessels to tumors, yet a comprehensive understanding of the interactions between NCs and vascular systems remains elusive. In recent years, considerable efforts have been invested in elucidating the transvascular transport mechanisms of NCs, leading to promising advancements in tumor delivery and theranostics. In this context, we highlight various delivery mechanisms, including the enhanced permeability and retention effect, cooperative immune-driven effect, active transcytosis, and cell/bacteria-mediated delivery. Furthermore, we explore corresponding strategies aimed at enhancing transvascular transport of NCs for efficient tumor delivery. These approaches offer intriguing solutions spanning physicochemical, biological, and pharmacological domains to improve delivery and therapeutic outcomes. Additionally, we propose a forward-looking delivery framework that relies on advanced tumor/vessel models, high-throughput NC libraries, nano-bio interaction datasets, and artificial intelligence, which aims to guide the design of next-generation carriers and implementation strategies for optimized delivery.
Collapse
Affiliation(s)
- Xin Li
- DWI-Leibniz-Institute for Interactive Materials, Aachen, 52056, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen, 52074, Germany
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
| | - Yong Hu
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
| | - Xingcai Zhang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA.
| | - Xiangyang Shi
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Wolfgang J Parak
- Center for Hybrid Nanostructures (CHyN), University of Hamburg, Hamburg, 20607, Germany.
| | - Andrij Pich
- DWI-Leibniz-Institute for Interactive Materials, Aachen, 52056, Germany.
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen, 52074, Germany.
- Aachen Maastricht Institute for Biobased Materials, Maastricht University, RD Geleen, 6167, The Netherlands.
| |
Collapse
|
33
|
You J, Guo Y, Dong Z. Polypeptides-Based Nanocarriers in Tumor Therapy. Pharmaceutics 2024; 16:1192. [PMID: 39339228 PMCID: PMC11435007 DOI: 10.3390/pharmaceutics16091192] [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: 07/16/2024] [Revised: 09/07/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
Cancer remains a worldwide problem, and new treatment strategies are being actively developed. Peptides have the characteristics of good biocompatibility, strong targeting, functional diversity, modifiability, membrane permeable ability, and low immunogenicity, and they have been widely used to construct targeted drug delivery systems (DDSs). In addition, peptides, as endogenous substances, have a high affinity, which can not only regulate immune cells but also work synergistically with drugs to kill tumor cells, demonstrating significant potential for application. In this review, the latest progress of polypeptides-based nanocarriers in tumor therapy has been outlined, focusing on their applications in killing tumor cells and regulating immune cells. Additionally, peptides as carriers were found to primarily provide a transport function, which was also a subject of interest to us. At the end of the paper, the shortcomings in the construction of peptide nano-delivery system have been summarized, and possible solutions are proposed therein. The application of peptides provides a promising outlook for cancer treatment, and we hope this article can provide in-depth insights into possible future avenues of exploration.
Collapse
Affiliation(s)
- Juhua You
- School of Pharmacy, Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Xiangfang District, Harbin 150040, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Yifei Guo
- School of Pharmacy, Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Xiangfang District, Harbin 150040, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| | - Zhengqi Dong
- School of Pharmacy, Heilongjiang University of Chinese Medicine, No. 24, Heping Road, Xiangfang District, Harbin 150040, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, China
| |
Collapse
|
34
|
Yang Z, Xu C, Lee JX, Lum GZ. Magnetic Miniature Soft Robot with Reprogrammable Drug-Dispensing Functionalities: Toward Advanced Targeted Combination Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2408750. [PMID: 39246210 DOI: 10.1002/adma.202408750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/26/2024] [Indexed: 09/10/2024]
Abstract
Miniature robots are untethered actuators, which have great prospects to transform targeted drug delivery because they can potentially deliver high concentrations of medicine to the disease site(s) with minimal complications. However, existing miniature robots cannot perform advanced targeted combination therapy; majority of them can at most transport one type of drug, while those that can carry multiple drugs are unable to change their drug-dispensing sequence and dosage. Furthermore, the latter robots cannot transport more than three types of drugs, selectively dispense their drugs, maintain their mobility, or release their drugs at multiple sites. Here, a millimeter-scale soft robot is proposed, which can be actuated by alternating magnetic fields to dispense four types of drugs with reprogrammable drug-dispensing sequence and dosage (dispensing rates: 0.0992-0.231 µL h-1). This robot has six degrees-of-freedom motions, and it can deliver its drugs to multiple desired sites by rolling and two-anchor crawling across unstructured environments with negligible drug leakage. Such dexterity is highly desirable and unprecedented for miniature robots with drug-dispensing capabilities. The soft robot therefore has great potential to enable advanced targeted combination therapy, where four types of drugs must be delivered to various disease sites, each with a specific sequence and dosage of drugs.
Collapse
Affiliation(s)
- Zilin Yang
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Changyu Xu
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Jia Xin Lee
- Rehabilitation Research Institute of Singapore, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Guo Zhan Lum
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
- Rehabilitation Research Institute of Singapore, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore
| |
Collapse
|
35
|
Guo Y, Ashrafizadeh M, Tambuwala MM, Ren J, Orive G, Yu G. P-glycoprotein (P-gp)-driven cancer drug resistance: biological profile, non-coding RNAs, drugs and nanomodulators. Drug Discov Today 2024; 29:104161. [PMID: 39245345 DOI: 10.1016/j.drudis.2024.104161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/07/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Drug resistance has compromised the efficacy of chemotherapy. The dysregulation of drug transporters including P-glycoprotein (P-gp) can mediate drug resistance through drug efflux. In this review, we highlight the role of P-gp in cancer drug resistance and the related molecular pathways, including phosphoinositide 3-kinase (PI3K)-Akt, phosphatase and tensin homolog (PTEN) and nuclear factor-κB (NF-κB), along with non-coding RNAs (ncRNAs). Extracellular vesicles secreted by the cells can transport ncRNAs and other proteins to change P-gp activity in cancer drug resistance. P-gp requires ATP to function, and the induction of mitochondrial dysfunction or inhibition of glutamine metabolism can impair P-gp function, thus increasing chemosensitivity. Phytochemicals, small molecules and nanoparticles have been introduced as P-gp inhibitors to increase drug sensitivity in human cancers.
Collapse
Affiliation(s)
- Yang Guo
- Department of Respiratory and Critical Care Medicine, Shenyang Tenth People's Hospital (Shenyang Chest Hospital), No. 11 Beihai Street, Dadong District, Shenyang 110044, Liaoning, China
| | - Milad Ashrafizadeh
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China; Department of Radiation Oncology, Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, UK
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology-UIRMI (UPV/EHU-Fundación Eduardo Anitua), 01007 Vitoria-Gasteiz, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore 169856, Singapore.
| | - Guiping Yu
- Department of Cardiothoracic Surgery, The Affiliated Jiangyin Hospital of Nantong University, No. 163 Shoushan Road, Jiangyin, China.
| |
Collapse
|
36
|
Ahmed U, Gew LT, Siddiqui R, Khan NA, Alharbi AM, Alhazmi A, Anwar A. Metal Oxide Nanoparticles Exhibit Anti-Acanthamoeba castellanii Properties by Inducing Necrotic Cell Death. Acta Parasitol 2024; 69:1717-1723. [PMID: 39153011 DOI: 10.1007/s11686-024-00891-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 07/30/2024] [Indexed: 08/19/2024]
Abstract
PURPOSE The treatment of amoebic infections is often problematic, largely due to delayed diagnosis, amoebae transformation into resistant cyst form, and lack of availability of effective chemotherapeutic agents. Herein, we determined anti-Acanthamoeba castellanii properties of three metal oxide nanoparticles (TiO2, ZrO2, and Al2O3). METHODS Amoebicidal assays were performed to determine whether metal oxide nanoparticles inhibit amoebae viability. Encystation assays were performed to test whether metal oxide nanoparticles inhibit cyst formation. By measuring lactate dehydrogenase release, cytotoxicity assays were performed to determine human cell damage. Hoechst 33342/PI staining was performed to determine programmed cell death (apoptosis) and necrosis in A. castellanii. RESULTS TiO2-NPs significantly inhibited amoebae viability as observed through amoebicidal assays, as well as inhibited their phenotypic transformation as evident using encystation assays, and showed limited human cell damage as observed by measuring lactate dehydrogenase assays. Furthermore, TiO2-NPs altered parasite membranes and resulted in necrotic cell death as determined using double staining cell death assays with Hoechst33342/Propidium iodide (PI) observed through chromatin condensation. These findings suggest that TiO2-NPs offers a potential viable avenue in the rationale development of therapeutic interventions against Acanthamoeba infections.
Collapse
Affiliation(s)
- Usman Ahmed
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Lai Ti Gew
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Ruqaiyyah Siddiqui
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University Edinburgh, Edinburgh, EH14 4AS, UK
- Microbiota Research Center, Istinye University, 34010, Istanbul, Turkey
| | - Naveed Ahmed Khan
- Microbiota Research Center, Istinye University, 34010, Istanbul, Turkey.
| | - Ahmad M Alharbi
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Ayman Alhazmi
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Ayaz Anwar
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
37
|
Duan X, Wang P, He L, He Z, Wang S, Yang F, Gao C, Ren W, Lin J, Chen T, Xu C, Li J, Wu A. Peptide-Functionalized Inorganic Oxide Nanomaterials for Solid Cancer Imaging and Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311548. [PMID: 38333964 DOI: 10.1002/adma.202311548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/15/2024] [Indexed: 02/10/2024]
Abstract
The diagnosis and treatment of solid tumors have undergone significant advancements marked by a trend toward increased specificity and integration of imaging and therapeutic functions. The multifaceted nature of inorganic oxide nanomaterials (IONs), which boast optical, magnetic, ultrasonic, and biochemical modulatory properties, makes them ideal building blocks for developing multifunctional nanoplatforms. A promising class of materials that have emerged in this context are peptide-functionalized inorganic oxide nanomaterials (PFIONs), which have demonstrated excellent performance in multifunctional imaging and therapy, making them potential candidates for advancing solid tumor diagnosis and treatment. Owing to the functionalities of peptides in tumor targeting, penetration, responsiveness, and therapy, well-designed PFIONs can specifically accumulate and release therapeutic or imaging agents at the solid tumor sites, enabling precise imaging and effective treatment. This review provides an overview of the recent advances in the use of PFIONs for the imaging and treatment of solid tumors, highlighting the superiority of imaging and therapeutic integration as well as synergistic treatment. Moreover, the review discusses the challenges and prospects of PFIONs in depth, aiming to promote the intersection of the interdisciplinary to facilitate their clinical translation and the development of personalized diagnostic and therapeutic systems by optimizing the material systems.
Collapse
Affiliation(s)
- Xiaolin Duan
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Pin Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lulu He
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Zhen He
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shiwei Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fang Yang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Changyong Gao
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Wenzhi Ren
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Jie Lin
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Tianxiang Chen
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Chen Xu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Juan Li
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Aiguo Wu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| |
Collapse
|
38
|
Zhang B, Yan J, Jin Y, Yang Y, Zhao X. Curcumin-shellac nanoparticle-loaded GelMA/SilMA hydrogel for colorectal cancer therapy. Eur J Pharm Biopharm 2024; 202:114409. [PMID: 38996942 DOI: 10.1016/j.ejpb.2024.114409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
In this study, a novel approach was employed to develop a therapeutic system for colorectal cancer treatment. Specifically, a GelMA/SilMA hydrogel loaded with curcumin-shellac nanoparticles (Cur@Lac NPs) was created. A microfluidic swirl mixer was utilized to formulate stable Cur@Lac NPs, ensuring their consistent and effective encapsulation. The pH-specific release of curcumin from the NPs demonstrated their potential for colon cancer treatment. By carefully regulating the ratio of GelMA (gelatin methacrylate) and SilMA (silk fibroin methacrylate), a GelMA/SilMA dual network hydrogel was generated, offering controlled release and degradation capabilities. The incorporation of SilMA notably enhanced the mechanical properties of the dual network matrix, improving compression resistance and mitigating deformation. This mechanical improvement is crucial for maintaining the structural integrity of the hydrogel during in vivo applications. In comparison to the direct incubation of curcumin, the strategy of encapsulating curcumin into NPs and embedding them within the GelMA/SilMA hydrogel resulted in more controlled release mechanisms. This controlled release was achieved through the disintegration of the NPs and the swelling and degradation of the hydrogel matrix. The encapsulating strategy also demonstrated enhanced cellular uptake of curcumin, leveraging the advantages of both NPs and in-situ hydrogel injection. This combination ensures a more efficient and sustained delivery of the therapeutic agent directly to the tumor site. Overall, this approach holds significant promise as a smart drug delivery system, potentially improving the efficacy of colorectal cancer treatments by providing targeted, controlled, and sustained drug release with enhanced mechanical stability and biocompatibility.
Collapse
Affiliation(s)
- Bo Zhang
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Jiaxuan Yan
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Yi Jin
- Department of Pharmacy, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213000, China; The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Yushun Yang
- Jinhua Advanced Research Institute, Jinhua 321019, China
| | - Xiubo Zhao
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| |
Collapse
|
39
|
Lowry GV, Giraldo JP, Steinmetz NF, Avellan A, Demirer GS, Ristroph KD, Wang GJ, Hendren CO, Alabi CA, Caparco A, da Silva W, González-Gamboa I, Grieger KD, Jeon SJ, Khodakovskaya MV, Kohay H, Kumar V, Muthuramalingam R, Poffenbarger H, Santra S, Tilton RD, White JC. Towards realizing nano-enabled precision delivery in plants. NATURE NANOTECHNOLOGY 2024; 19:1255-1269. [PMID: 38844663 DOI: 10.1038/s41565-024-01667-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 03/27/2024] [Indexed: 09/18/2024]
Abstract
Nanocarriers (NCs) that can precisely deliver active agents, nutrients and genetic materials into plants will make crop agriculture more resilient to climate change and sustainable. As a research field, nano-agriculture is still developing, with significant scientific and societal barriers to overcome. In this Review, we argue that lessons can be learned from mammalian nanomedicine. In particular, it may be possible to enhance efficiency and efficacy by improving our understanding of how NC properties affect their interactions with plant surfaces and biomolecules, and their ability to carry and deliver cargo to specific locations. New tools are required to rapidly assess NC-plant interactions and to explore and verify the range of viable targeting approaches in plants. Elucidating these interactions can lead to the creation of computer-generated in silico models (digital twins) to predict the impact of different NC and plant properties, biological responses, and environmental conditions on the efficiency and efficacy of nanotechnology approaches. Finally, we highlight the need for nano-agriculture researchers and social scientists to converge in order to develop sustainable, safe and socially acceptable NCs.
Collapse
Affiliation(s)
- Gregory V Lowry
- Civil and Environmental Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| | - Juan Pablo Giraldo
- Botany and Plant Sciences, University of California, Riverside, Riverside, CA, USA.
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California San Diego, San Diego, CA, USA
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
- Department of Radiology, University of California San Diego, San Diego, CA, USA
- Center for Nano-ImmunoEngineering, University of California San Diego, San Diego, CA, USA
- Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, San Diego, CA, USA
- Center for Engineering in Cancer, Institute of Engineering in Medicine, University of California San Diego, San Diego, CA, USA
- Moores Cancer Center, University of California, University of California San Diego, San Diego, CA, USA
- Institute for Materials Discovery and Design, University of California San Diego, San Diego, CA, USA
| | | | - Gozde S Demirer
- Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Kurt D Ristroph
- Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, USA
| | - Gerald J Wang
- Civil and Environmental Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Christine O Hendren
- Geological and Environmental Sciences, Appalachian State University, Boone, NC, USA
| | | | - Adam Caparco
- Department of NanoEngineering, University of California San Diego, San Diego, CA, USA
| | | | | | - Khara D Grieger
- Applied Ecology, North Carolina State University, Raleigh, NC, USA
| | - Su-Ji Jeon
- Botany and Plant Sciences, University of California, Riverside, Riverside, CA, USA
| | | | - Hagay Kohay
- Civil and Environmental Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Vivek Kumar
- Civil and Environmental Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | | | - Swadeshmukul Santra
- Department of Chemistry and Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Robert D Tilton
- Chemical Engineering and Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Jason C White
- The Connecticut Agricultural Research Station, New Haven, CT, USA
| |
Collapse
|
40
|
ten Hove M, Smyris A, Booijink R, Wachsmuth L, Hansen U, Alic L, Faber C, Hӧltke C, Bansal R. Engineered SPIONs functionalized with endothelin a receptor antagonist ameliorate liver fibrosis by inhibiting hepatic stellate cell activation. Bioact Mater 2024; 39:406-426. [PMID: 38855059 PMCID: PMC11157122 DOI: 10.1016/j.bioactmat.2024.05.034] [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: 11/16/2023] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/11/2024] Open
Abstract
Endothelin-1/endothelin A receptor (ET-1/ETAR) pathway plays an important role in the progression of liver fibrosis by activating hepatic stellate cells (HSCs) - a key cell type involved in the pathogenesis of liver fibrosis. Inactivating HSCs by blocking the ET-1/ETAR pathway using a selective ETAR antagonist (ERA) represents a promising therapeutic approach for liver fibrosis. Unfortunately, small-molecule ERAs possess limited clinical potential due to poor bioavailability, short half-life, and rapid renal clearance. To improve the clinical applicability, we conjugated ERA to superparamagnetic iron-oxide nanoparticles (SPIONs) and investigated the therapeutic efficacy of ERA and ERA-SPIONs in vitro and in vivo and analyzed liver uptake by in vivo and ex vivo magnetic resonance imaging (MRI), HSCs-specific localization, and ET-1/ETAR-pathway antagonism in vivo. In murine and human liver fibrosis/cirrhosis, we observed overexpression of ET-1 and ETAR that correlated with HSC activation, and HSC-specific localization of ETAR. ERA and successfully synthesized ERA-SPIONs demonstrated significant attenuation in TGFβ-induced HSC activation, ECM production, migration, and contractility. In an acute CCl4-induced liver fibrosis mouse model, ERA-SPIONs exhibited higher liver uptake, HSC-specific localization, and ET-1/ETAR pathway antagonism. This resulted in significantly reduced liver-to-body weight ratio, plasma ALT levels, and α-SMA and collagen-I expression, indicating attenuation of liver fibrosis. In conclusion, our study demonstrates that the delivery of ERA using SPIONs enhances the therapeutic efficacy of ERA in vivo. This approach holds promise as a theranostic strategy for the MRI-based diagnosis and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Marit ten Hove
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| | - Andreas Smyris
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| | - Richell Booijink
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| | - Lydia Wachsmuth
- Clinic of Radiology, University Hospital Muenster, Muenster, Germany
| | - Uwe Hansen
- Institute for Musculoskeletal Medicine, University Hospital Muenster, Muenster, Germany
| | - Lejla Alic
- Department of Magnetic Detection and Imaging, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| | - Cornelius Faber
- Clinic of Radiology, University Hospital Muenster, Muenster, Germany
| | - Carsten Hӧltke
- Clinic of Radiology, University Hospital Muenster, Muenster, Germany
| | - Ruchi Bansal
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, the Netherlands
| |
Collapse
|
41
|
Longobardi G, Moore TL, Conte C, Ungaro F, Satchi-Fainaro R, Quaglia F. Polyester nanoparticles delivering chemotherapeutics: Learning from the past and looking to the future to enhance their clinical impact in tumor therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1990. [PMID: 39217459 DOI: 10.1002/wnan.1990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Polymeric nanoparticles (NPs), specifically those comprised of biodegradable and biocompatible polyesters, have been heralded as a game-changing drug delivery platform. In fact, poly(α-hydroxy acids) such as polylactide (PLA), poly(lactide-co-glycolide) (PLGA), and poly(ε-caprolactone) (PCL) have been heavily researched in the past three decades as the material basis of polymeric NPs for drug delivery applications. As materials, these polymers have found success in resorbable sutures, biodegradable implants, and even monolithic, biodegradable platforms for sustained release of therapeutics (e.g., proteins and small molecules) and diagnostics. Few fields have gained more attention in drug delivery through polymeric NPs than cancer therapy. However, the clinical translational of polymeric nanomedicines for treating solid tumors has not been congruent with the fervor or funding in this particular field of research. Here, we attempt to provide a comprehensive snapshot of polyester NPs in the context of chemotherapeutic delivery. This includes a preliminary exploration of the polymeric nanomedicine in the cancer research space. We examine the various processes for producing polyester NPs, including methods for surface-functionalization, and related challenges. After a detailed overview of the multiple factors involved with the delivery of NPs to solid tumors, the crosstalk between particle design and interactions with biological systems is discussed. Finally, we report state-of-the-art approaches toward effective delivery of NPs to tumors, aiming at identifying new research areas and re-evaluating the reasons why some research avenues have underdelivered. We hope our effort will contribute to a better understanding of the gap to fill and delineate the future research work needed to bring polyester-based NPs closer to clinical application. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
| | - Thomas Lee Moore
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Claudia Conte
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Francesca Ungaro
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, Israel
| | - Fabiana Quaglia
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
42
|
Almeida DRS, Gil JF, Guillot AJ, Li J, Pinto RJB, Santos HA, Gonçalves G. Advances in Microfluidic-Based Core@Shell Nanoparticles Fabrication for Cancer Applications. Adv Healthc Mater 2024; 13:e2400946. [PMID: 38736024 DOI: 10.1002/adhm.202400946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/09/2024] [Indexed: 05/14/2024]
Abstract
Current research in cancer therapy focuses on personalized therapies, through nanotechnology-based targeted drug delivery systems. Particularly, controlled drug release with nanoparticles (NPs) can be designed to safely transport various active agents, optimizing delivery to specific organs and tumors, minimizing side effects. The use of microfluidics (MFs) in this field has stood out against conventional methods by allowing precise control over parameters like size, structure, composition, and mechanical/biological properties of nanoscale carriers. This review compiles applications of microfluidics in the production of core-shell NPs (CSNPs) for cancer therapy, discussing the versatility inherent in various microchannel and/or micromixer setups and showcasing how these setups can be utilized individually or in combination, as well as how this technology allows the development of new advances in more efficient and controlled fabrication of core-shell nanoformulations. Recent biological studies have achieved an effective, safe, and controlled delivery of otherwise unreliable encapsulants such as small interfering RNA (siRNA), plasmid DNA (pDNA), and cisplatin as a result of precisely tuned fabrication of nanocarriers, showing that this technology is paving the way for innovative strategies in cancer therapy nanofabrication, characterized by continuous production and high reproducibility. Finally, this review analyzes the technical, biological, and technological limitations that currently prevent this technology from becoming the standard.
Collapse
Affiliation(s)
- Duarte R S Almeida
- Centre for Mechanical Technology and Automation (TEMA), Mechanical Engineering Department, University of Aveiro, Aveiro, 3810-193, Portugal
- Intelligent Systems Associate Laboratory (LASI), Guimarães, 4800-058, Portugal
| | - João Ferreira Gil
- Centre for Mechanical Technology and Automation (TEMA), Mechanical Engineering Department, University of Aveiro, Aveiro, 3810-193, Portugal
- Intelligent Systems Associate Laboratory (LASI), Guimarães, 4800-058, Portugal
| | - Antonio José Guillot
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Ave. Vicent Andrés Estellés s/n, Burjassot, Valencia, 46100, Spain
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 AV, The Netherlands
| | - Jiachen Li
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 AV, The Netherlands
| | - Ricardo J B Pinto
- CICECO-Aveiro Institute of Materials, Chemistry Department, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 AV, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
| | - Gil Gonçalves
- Centre for Mechanical Technology and Automation (TEMA), Mechanical Engineering Department, University of Aveiro, Aveiro, 3810-193, Portugal
- Intelligent Systems Associate Laboratory (LASI), Guimarães, 4800-058, Portugal
| |
Collapse
|
43
|
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
|
44
|
Huang Y, Chen C, Yu Z, Cao W, Peng S, Zhang G, Zhang Q, Zhang G, Jiang J, Yuan Y. A Simple Binary Supramolecular Co-Assembly Platform for Enhanced Tumor Imaging and Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2402763. [PMID: 39183531 DOI: 10.1002/smll.202402763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 08/10/2024] [Indexed: 08/27/2024]
Abstract
The primary challenges in tumor imaging and therapy revolve around improving targeting efficiency, enhancing probe/drug delivery efficacy, and minimizing off-target signals and toxicity. Although various carriers have been developed, many are difficult to synthesize, costly, and not universally applicable. Furthermore, numerous carriers exhibit limited delivery rates in solid tumors, particularly larger nanocarriers. To address these challenges, a simple binary co-assembly drug delivery platform has been designed using the readily synthesized small molecule Cys(SEt)-Lys-CBT (CKCBT) as the self-assembly building block. CKCBT can effectively penetrate tumor cells due to its positively charged Lys side chain and small size. Upon glutathione reduction, CKCBT co-assembles with Nile red or Chlorin e6 to form nanofibers inside tumor cells. This enables their specific accumulation in tumor cells rather than normal cells and extends their exposure time, resulting in precise and enhanced tumor imaging and treatment. Hence, this uncomplicated and highly efficient binary co-assembly drug delivery platform can be easily adapted to a broad spectrum of probes and drugs, presenting a novel approach for advancing clinical diagnosis and therapy.
Collapse
Affiliation(s)
- Yifan Huang
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
| | - Cheng Chen
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Zian Yu
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Wei Cao
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230088, China
| | - Shengjie Peng
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230088, China
| | - Guangtao Zhang
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
| | - Qianzijing Zhang
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Guozhen Zhang
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Jun Jiang
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yue Yuan
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
45
|
Wu X, Ma L, Zhang Y, Liu S, Cheng L, You C, Dong Z. Application progress of nanomaterials in the treatment of prostate cancer. ANNALES PHARMACEUTIQUES FRANÇAISES 2024:S0003-4509(24)00131-7. [PMID: 39187009 DOI: 10.1016/j.pharma.2024.08.009] [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: 07/12/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024]
Abstract
Prostate cancer is one of the most common malignant tumors in men, which seriously threatens the survival and quality of life of patients. At present, there are serious limitations in the treatment of prostate cancer, such as drug tolerance, drug resistance and easy recurrence. Sonodynamic therapy and chemodynamic therapy are two emerging tumor treatment methods, which activate specific drugs or sonosensitizers through sound waves or chemicals to produce reactive oxygen species and kill tumor cells. Nanomaterials are a kind of nanoscale materials with many excellent physical properties such as high targeting, drug release regulation and therapeutic monitoring. Sonodynamic therapy and chemodynamic therapy combined with the application of nanomaterials can improve the therapeutic effect of prostate cancer, reduce side effects and enhance tumor immune response. This article reviews the application progress of nanomaterials in the treatment of prostate cancer, especially the mechanism, advantages and challenges of nanomaterials in sonodynamic therapy and chemodynamic therapy, which provides new ideas and prospects for research in this field.
Collapse
Affiliation(s)
- Xuewu Wu
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China
| | - Longtu Ma
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China
| | - Yang Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, China
| | - Shuai Liu
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China
| | - Long Cheng
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China
| | - Chengyu You
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China
| | - Zhilong Dong
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China.
| |
Collapse
|
46
|
Lee LCC, Lo KKW. Shining New Light on Biological Systems: Luminescent Transition Metal Complexes for Bioimaging and Biosensing Applications. Chem Rev 2024; 124:8825-9014. [PMID: 39052606 PMCID: PMC11328004 DOI: 10.1021/acs.chemrev.3c00629] [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: 07/27/2024]
Abstract
Luminescence imaging is a powerful and versatile technique for investigating cell physiology and pathology in living systems, making significant contributions to life science research and clinical diagnosis. In recent years, luminescent transition metal complexes have gained significant attention for diagnostic and therapeutic applications due to their unique photophysical and photochemical properties. In this Review, we provide a comprehensive overview of the recent development of luminescent transition metal complexes for bioimaging and biosensing applications, with a focus on transition metal centers with a d6, d8, and d10 electronic configuration. We elucidate the structure-property relationships of luminescent transition metal complexes, exploring how their structural characteristics can be manipulated to control their biological behavior such as cellular uptake, localization, biocompatibility, pharmacokinetics, and biodistribution. Furthermore, we introduce the various design strategies that leverage the interesting photophysical properties of luminescent transition metal complexes for a wide variety of biological applications, including autofluorescence-free imaging, multimodal imaging, organelle imaging, biological sensing, microenvironment monitoring, bioorthogonal labeling, bacterial imaging, and cell viability assessment. Finally, we provide insights into the challenges and perspectives of luminescent transition metal complexes for bioimaging and biosensing applications, as well as their use in disease diagnosis and treatment evaluation.
Collapse
Affiliation(s)
- Lawrence Cho-Cheung Lee
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Units 1503-1511, 15/F, Building 17W, Hong Kong Science Park, New Territories, Hong Kong, P. R. China
| | - Kenneth Kam-Wing Lo
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
- State Key Laboratory of Terahertz and Millimeter Waves, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, P. R. China
| |
Collapse
|
47
|
Garnaik UC, Chandra A, Goel VK, Gulyás B, Padmanabhan P, Agarwal S. Development of SERS Active Nanoprobe for Selective Adsorption and Detection of Alzheimer's Disease Biomarkers Based on Molecular Docking. Int J Nanomedicine 2024; 19:8271-8284. [PMID: 39161360 PMCID: PMC11330857 DOI: 10.2147/ijn.s446212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/08/2024] [Indexed: 08/21/2024] Open
Abstract
Purpose Development of SERS-based Raman nanoprobes can detect the misfolding of Amyloid beta (Aβ) 42 peptides, making them a viable diagnostic technique for Alzheimer's disease (AD). The detection and imaging of amyloid peptides and fibrils are expected to help in the early identification of AD. Methods Here, we propose a fast, easy-to-use, and simple scheme based on the selective adsorption of Aβ42 molecules on SERS active gold nanoprobe (RB-AuNPs) of diameter 29 ± 3 nm for Detection of Alzheimer's Disease Biomarkers. Binding with the peptides results in a spectrum shift, which correlates with the target peptide. We also demonstrated the possibility of using silver nanoparticles (AgNPs) as precursors for the preparation of a SERS active nanoprobe with carbocyanine (CC) dye and AgNPs known as silver nanoprobe (CC-AgNPs) of diameter 25 ± 4 nm. Results RB-AuNPs probe binding with the peptides results in a spectrum shift, which correlates with the target peptide. Arginine peak appears after the conjugation confirms the binding of Aβ 42 with the nanoprobe. Tyrosine peaks appear after conjugated Aβ42 with CC-AgNPs providing binding of the peptide with the probe. The nanoprobe produced a strong, stable SERS signal. Further molecular docking was utilized to analyse the interaction and propose a structural hypothesis for the process of binding the nanoprobe to Aβ42 and Tau protein. Conclusion This peptide-probe interaction provides a general enhancement factor and the molecular structure of the misfolded peptides. Secondary structural information may be obtained at the molecular level for specific residues owing to isotope shifts in the Raman spectra. Conjugation of the nanoprobe with Aβ42 selectively detected AD in bodily fluids. The proposed nanoprobes can be easily applied to the detection of Aβ plaques in blood, saliva, and sweat samples.
Collapse
Affiliation(s)
| | - Anshuman Chandra
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Vijay Kumar Goel
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Balázs Gulyás
- Cognitive Neuroimaging Centre, Nanyang Technological University (NTU), Singapore, Singapore
| | | | - Shilpi Agarwal
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
48
|
Wang B, Hu S, Teng Y, Chen J, Wang H, Xu Y, Wang K, Xu J, Cheng Y, Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct Target Ther 2024; 9:200. [PMID: 39128942 PMCID: PMC11323968 DOI: 10.1038/s41392-024-01889-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/04/2024] [Accepted: 06/02/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a significant risk to human health. Nanomedicine is a new multidisciplinary field that is garnering a lot of interest and investigation. Nanomedicine shows great potential for cancer diagnosis and treatment. Specifically engineered nanoparticles can be employed as contrast agents in cancer diagnostics to enable high sensitivity and high-resolution tumor detection by imaging examinations. Novel approaches for tumor labeling and detection are also made possible by the use of nanoprobes and nanobiosensors. The achievement of targeted medication delivery in cancer therapy can be accomplished through the rational design and manufacture of nanodrug carriers. Nanoparticles have the capability to effectively transport medications or gene fragments to tumor tissues via passive or active targeting processes, thus enhancing treatment outcomes while minimizing harm to healthy tissues. Simultaneously, nanoparticles can be employed in the context of radiation sensitization and photothermal therapy to enhance the therapeutic efficacy of malignant tumors. This review presents a literature overview and summary of how nanotechnology is used in the diagnosis and treatment of malignant tumors. According to oncological diseases originating from different systems of the body and combining the pathophysiological features of cancers at different sites, we review the most recent developments in nanotechnology applications. Finally, we briefly discuss the prospects and challenges of nanotechnology in cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, Evidence-based Pharmacy Center, Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Shiqi Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yan Teng
- Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Junli Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haoyuan Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yezhen Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kaiyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
49
|
Yin X, Ke Y, Liang Y, Zhang S, Chen Z, Yu L, Jiang M, Liu Q, Gu X. An Immune-Enhancing Injectable Hydrogel Loaded with Esketamine and DDP Promotes Painless Immunochemotherapy to Inhibit Breast Cancer Growth. Adv Healthc Mater 2024:e2401373. [PMID: 39118566 DOI: 10.1002/adhm.202401373] [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: 04/15/2024] [Revised: 07/07/2024] [Indexed: 08/10/2024]
Abstract
Chemotherapy is the cornerstone of triple-negative breast cancer. The poor effectiveness and severe neuropathic pain caused by it have a significant impact on the immune system. Studies confirmed that immune cells in the tumor microenvironment (TME), have critical roles in tumor immune regulation and prognosis. In this study, it is revealed that the painless administration of Esketamine, combined with Cisplatin (DDP), can exert an anti-tumor effect, which is further boosted by the hydrogel delivery system. It is also discovered that Esketamine combined with DDP co-loaded in Poloxamer Hydrogel (PDEH) induces local immunity by increasing mature Dendritic Cells (mDCs) and activated T cells in PDEH group while the regulatory T cells (Tregs) known as CD4+CD25+FoxP3+decreased significantly. Finally, , CD8+ and CD4+ T cells in the spleen exhibited a significant increase, suggesting a lasting immune impact of PDEH. This study proposes that Esketamine can serve as a painless immune modulator, enhancing an anti-tumor effect while co-loaded in poloxamer hydrogel with DDP. Along with improving immune cells in the microenvironment, it can potentially alleviate anxiety and depression. With its outstanding bio-safety profile, it offers promising new possibilities for painless clinical therapy.
Collapse
Affiliation(s)
- Xiali Yin
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical School, Nanjing, 210008, China
| | - Yaohua Ke
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Ying Liang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Shuxian Zhang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Ziqi Chen
- The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing, 210008, China
| | - Lixia Yu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Ming Jiang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Qin Liu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Xiaoping Gu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical School, Nanjing, 210008, China
| |
Collapse
|
50
|
Hu M, Yingyu Z, Zhang M, Wang Q, Cheng W, Hou L, Yuan J, Yu Z, Li L, Zhang X, Zhang W. Functionalizing tetrahedral framework nucleic acids-based nanostructures for tumor in situ imaging and treatment. Colloids Surf B Biointerfaces 2024; 240:113982. [PMID: 38788473 DOI: 10.1016/j.colsurfb.2024.113982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
Timely in situ imaging and effective treatment are efficient strategies in improving the therapeutic effect and survival rate of tumor patients. In recent years, there has been rapid progress in the development of DNA nanomaterials for tumor in situ imaging and treatment, due to their unsurpassed structural stability, excellent material editability, excellent biocompatibility and individual endocytic pathway. Tetrahedral framework nucleic acids (tFNAs), are a typical example of DNA nanostructures demonstrating superior stability, biocompatibility, cell-entry performance, and flexible drug-loading ability. tFNAs have been shown to be effective in achieving timely tumor in situ imaging and precise treatment. Therefore, the progress in the fabrication, characterization, modification and cellular internalization pathway of tFNAs-based functional systems and their potential in tumor in situ imaging and treatment applications were systematically reviewed in this article. In addition, challenges and future prospects of tFNAs in tumor in situ imaging and treatment as well as potential clinical applications were discussed.
Collapse
Affiliation(s)
- Minghui Hu
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China
| | - Zhang Yingyu
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Mengxin Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China
| | - Qionglin Wang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China
| | - Weyland Cheng
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China
| | - Ligong Hou
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China
| | - Jingya Yuan
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Zhidan Yu
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China
| | - Lifeng Li
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China
| | - Xianwei Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China.
| | - Wancun Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China; Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China; Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China.
| |
Collapse
|