1
|
Martorana A, Puleo G, Miceli GC, Cancilla F, Licciardi M, Pitarresi G, Tranchina L, Marrale M, Palumbo FS. Redox/NIR dual-responsive glutathione extended polyurethane urea electrospun membranes for synergistic chemo-photothermal therapy. Int J Pharm 2025; 669:125108. [PMID: 39708849 DOI: 10.1016/j.ijpharm.2024.125108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Despite advancements in cancer treatments, therapies frequently exhibit high cytotoxicity, and surgery remains the predominant method for treating most solid tumors, often with limited success in preventing post-surgical recurrence. Implantable biomaterials, designed to release drugs at a localised site in response to specific stimuli, represent a promising approach for enhancing tumour therapy. In this study, a redox-responsive glutathione extended polyurethane urea (PolyCEGS) was used to produce paclitaxel (PTX) and gold nanorods (AuNRs) loaded electrospun membranes for combined redox/near-infrared (NIR) light-responsive release chemotherapy and hyperthermic effect. Electrospinning conditions were optimized to fabricate AuNR-loaded scaffolds, at three different AuNRs concentrations. The obtained membranes were characterized by scanning electron microscopy (SEM) analyses and photothermal profiles were evaluated by a thermocamera, showing a temperature increase, up to 42.5 °C, when exposed to NIR light (810 nm) at 3 W/cm2. The AuNRs/PTX loaded scaffolds exhibited sustained PTX release, with 15 % released over 30 days and almost 1.8 times more in a simulated reductive environment. Moreover, their excellent photothermal effects and NIR light-triggered release led to significant synergic cytotoxicity in human colon cancer (HCT-116) and human breast cancer (MCF-7) cell lines. This system potentially enables controllable locoregional PTX release at the tumour site post-surgery, preventing recurrence and enhancing cytotoxicity through combined drug and PTT effects, highlighting its potential for future anticancer treatments.
Collapse
Affiliation(s)
- Annalisa Martorana
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, Italy; Fondazione Ri.MED, c/o IRCCS ISMETT, via E. Tricomi 5, 90127, Palermo, Italy(2)
| | - Giorgia Puleo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Edificio 18, Palermo, Italy; Department of Pharmacy, University of Copenhagen, Universitetsparken 2, Copenhagen, 2100, Denmark
| | - Giovanni Carlo Miceli
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, Italy; Department of Bioengineering, Imperial College London, London, SW7 2BX, UK(2)
| | - Francesco Cancilla
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, Italy
| | - Mariano Licciardi
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, Italy
| | - Giovanna Pitarresi
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, Italy
| | - Luigi Tranchina
- Advanced Technologies Network (ATeN) Center, University of Palermo, Viale delle Scienze, edificio 18a, Palermo, 90128, Italy
| | - Maurizio Marrale
- Department of Physics and Chemistry "Emilio Segrè", University of Palermo, Viale delle Scienze, edificio 18, Palermo, 90128, Italy; National Institute for Nuclear Physics (INFN), Catania Division, Via Santa Sofia,64, Catania, 95123, Italy
| | - Fabio Salvatore Palumbo
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, Italy; Istituto per la Ricerca e Innovazione Biomedica (IRIB), CNR, Via Ugo La Malfa, 153, 90146, Palermo, Italy.
| |
Collapse
|
2
|
Liu Z, Jia J, Lei Q, Wei Y, Hu Y, Lian X, Zhao L, Xie X, Bai H, He X, Si L, Livermore C, Kuang R, Zhang Y, Wang J, Yu Z, Ma X, Huang D. Electrohydrodynamic Direct-Writing Micro/Nanofibrous Architectures: Principle, Materials, and Biomedical Applications. Adv Healthc Mater 2024; 13:e2400930. [PMID: 38847291 DOI: 10.1002/adhm.202400930] [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: 03/12/2024] [Revised: 05/21/2024] [Indexed: 07/05/2024]
Abstract
Electrohydrodynamic (EHD) direct-writing has recently gained attention as a highly promising additive manufacturing strategy for fabricating intricate micro/nanoscale architectures. This technique is particularly well-suited for mimicking the extracellular matrix (ECM) present in biological tissue, which serves a vital function in facilitating cell colonization, migration, and growth. The integration of EHD direct-writing with other techniques has been employed to enhance the biological performance of scaffolds, and significant advancements have been made in the development of tailored scaffold architectures and constituents to meet the specific requirements of various biomedical applications. Here, a comprehensive overview of EHD direct-writing is provided, including its underlying principles, demonstrated materials systems, and biomedical applications. A brief chronology of EHD direct-writing is provided, along with an examination of the observed phenomena that occur during the printing process. The impact of biomaterial selection and architectural topographic cues on biological performance is also highlighted. Finally, the major limitations associated with EHD direct-writing are discussed.
Collapse
Affiliation(s)
- Zhengjiang Liu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
| | - Jinqiao Jia
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
| | - Qi Lei
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Yan Wei
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Yinchun Hu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Xiaojie Lian
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Liqin Zhao
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Xin Xie
- Xellar Biosystems, Cambridge, MA, 02458, USA
| | - Haiqing Bai
- Xellar Biosystems, Cambridge, MA, 02458, USA
| | - Xiaomin He
- Xellar Biosystems, Cambridge, MA, 02458, USA
| | - Longlong Si
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Carol Livermore
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Rong Kuang
- Zhejiang Institute for Food and Drug Control, Hangzhou, 310000, P. R. China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, P. R. China
| | - Jiucun Wang
- Human Phenome Institute, Fudan University, Shanghai, 200433, P. R. China
| | - Zhaoyan Yu
- Shandong Public Health Clinical Center, Shandong University, Jinan, 250000, P. R. China
| | - Xudong Ma
- Cytori Therapeutics LLC., Shanghai, 201802, P. R. China
| | - Di Huang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| |
Collapse
|
3
|
Zhang X, Zheng G, Zhou Z, Zhu M, Tang S. Co-delivery of siRNA and cisplatin via electrospun Nanofibrous membranes for synergistic treatment of malignant melanoma. Heliyon 2024; 10:e37517. [PMID: 39290263 PMCID: PMC11407083 DOI: 10.1016/j.heliyon.2024.e37517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 08/13/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
Tumor recurrence and metastasis remain formidable challenges in clinical oncology. Although surgery is an effective treatment for early-stage solid tumors, residual cancer cells can lead to subsequent recurrence or metastasis. Conventional treatments for melanoma, such as anti-tumor medications and gene therapy, have distinct limitations. The rapid systemic distribution of anti-tumor drugs poses a significant challenge, often resulting in notable side effects and inadequate drug concentrations at the tumor site. Melanoma (MM), a deadly form of skin cancer, is known for its high mortality rate. In this study, we propose a novel strategy for treating MM by combining the controlled release of chemotherapeutic drugs encapsulated within Metal-Organic Frameworks (MOFs) and liposomes with gene therapy targeting Minichromosome Maintenance Proteins 4 (MCM4) using electrospinning and surface modification techniques. In vitro and in vivo results confirmed that this hierarchical membrane system can effectively deliver therapeutic MCM4 siRNA and release cisplatin to inhibit tumor growth. Furthermore, we demonstrated that MCM4 silencing promoted the sensitivity of melanoma cells to ferroptosis both in vitro and in vivo. The proposed strategy, by allowing for a controlled and sustained release of medication, could alleviate the challenges in drug delivery and aid in prevent tumor recurrence.
Collapse
Affiliation(s)
- Xuewei Zhang
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515000, China
| | - Guoxing Zheng
- Department of Spine Surgery, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515000, China
| | - Zibin Zhou
- Department of Spine Surgery, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515000, China
| | - Mingyu Zhu
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Shijie Tang
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515000, China
| |
Collapse
|
4
|
Liu Z, Fan Y, Cui M, Wang X, Zhao P. Investigation of tumour environments through advancements in microtechnology and nanotechnology. Biomed Pharmacother 2024; 178:117230. [PMID: 39116787 DOI: 10.1016/j.biopha.2024.117230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer has a significant negative social and economic impact on both developed and developing countries. As a result, understanding the onset and progression of cancer is critical for developing therapies that can improve the well-being and health of individuals with cancer. With time, study has revealed, the tumor microenvironment has great influence on this process. Micro and nanoscale engineering techniques can be used to study the tumor microenvironment. Nanoscale and Microscale engineering use Novel technologies and designs with small dimensions to recreate the TME. Knowing how cancer cells interact with one another can help researchers develop therapeutic approaches that anticipate and counteract cancer cells' techniques for evading detection and fighting anti-cancer treatments, such as microfabrication techniques, microfluidic devices, nanosensors, and nanodevices used to study or recreate the tumor microenvironment. Nevertheless, a complicated action just like the growth and in cancer advancement, and their intensive association along the environment around it that has to be studied in more detail.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Radiology, Shengjing Hospital of China Medical University, China
| | - Yan Fan
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mengyao Cui
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xu Wang
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Pengfei Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
5
|
Zou R, Wang Y, Cai Y, Xing Z, Shao Y, Li D, Qi C. Nanofiber-based delivery of evodiamine impedes malignant properties of intrahepatic cholangiocarcinoma cells by targeting HDAC4 and restoring TPM1 transcription. Hum Cell 2024; 37:1505-1521. [PMID: 39073525 DOI: 10.1007/s13577-024-01105-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 07/11/2024] [Indexed: 07/30/2024]
Abstract
The electrospun nanofiber system is correlated with high efficacy of drug delivery. This study aims to investigate the effect of nanofiber-based delivery of evodiamine, an indole alkaloid derived from Rutaceae plants Evodia rutaecarpa (Juss.) Benth, on intrahepatic cholangiocarcinoma (ICC), as well as to explore the molecular mechanisms. An electrospun nanofiber system carrying evodiamine was generated. Compared to evodiamine treatment alone, the nano-evodiamine exhibited more pronounced effects on suppressing proliferation, colony formation, invasiveness, migration, apoptosis resistance, cell cycle progression, and in vivo tumorigenesis of two ICC cell lines (HUCC-T1 and RBE). ICC cells exhibited increased expression of histone deacetylase 4 (HDAC4) while decreased tropomyosin 1 (TPM1). HDAC4 suppressed TPM1 expression by removing H3K9ac modifications from its promoter. Nano-evodiamine reduced HDAC4 protein levels in ICC cells, thus promoting transcription and expression of TPM1. Either overexpression of HDAC4 or downregulation of TPM1 negated the tumor-suppressive effects of nano-evodiamine. Collectively, this study demonstrates that the electrospun nanofiber system enhances the efficiency of evodiamine. Additionally, evodiamine suppresses the malignant properties of ICC cells. The findings may provide fresh insights into the application of electrospun nanofiber system for drug delivery and the effects of evodiamine on tumor suppression.
Collapse
Affiliation(s)
- Rui Zou
- Hepatobiliary Pancreatic Surgery Division 1, Hainan Cancer Hospital, No. 9, Changbin West Fourth Street, Xiuying District, Haikou, 570100, Hainan, People's Republic of China
| | - Yiyao Wang
- Department of Integrated Traditional Chinese and Western Medicine, Hainan Cancer Hospital, Haikou, 570100, Hainan, People's Republic of China
| | - Yaoqing Cai
- Department of Gastrointestinal Surgery, Hainan Cancer Hospital, Haikou, 570100, Hainan, People's Republic of China
| | - Zhenming Xing
- Hepatobiliary Pancreatic Surgery Division 1, Hainan Cancer Hospital, No. 9, Changbin West Fourth Street, Xiuying District, Haikou, 570100, Hainan, People's Republic of China
| | - Yongfu Shao
- Hepatobiliary Pancreatic Surgery Division 1, Hainan Cancer Hospital, No. 9, Changbin West Fourth Street, Xiuying District, Haikou, 570100, Hainan, People's Republic of China
| | - Duo Li
- Hepatobiliary Pancreatic Surgery Division 1, Hainan Cancer Hospital, No. 9, Changbin West Fourth Street, Xiuying District, Haikou, 570100, Hainan, People's Republic of China.
| | - Chunchun Qi
- Medical College of Nankai University, Tianjin, 300071, People's Republic of China.
| |
Collapse
|
6
|
Agiba AM, Elsayyad N, ElShagea HN, Metwalli MA, Mahmoudsalehi AO, Beigi-Boroujeni S, Lozano O, Aguirre-Soto A, Arreola-Ramirez JL, Segura-Medina P, Hamed RR. Advances in Light-Responsive Smart Multifunctional Nanofibers: Implications for Targeted Drug Delivery and Cancer Therapy. Pharmaceutics 2024; 16:1017. [PMID: 39204362 PMCID: PMC11359459 DOI: 10.3390/pharmaceutics16081017] [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: 06/27/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Over the last decade, scientists have shifted their focus to the development of smart carriers for the delivery of chemotherapeutics in order to overcome the problems associated with traditional chemotherapy, such as poor aqueous solubility and bioavailability, low selectivity and targeting specificity, off-target drug side effects, and damage to surrounding healthy tissues. Nanofiber-based drug delivery systems have recently emerged as a promising drug delivery system in cancer therapy owing to their unique structural and functional properties, including tunable interconnected porosity, a high surface-to-volume ratio associated with high entrapment efficiency and drug loading capacity, and high mass transport properties, which allow for controlled and targeted drug delivery. In addition, they are biocompatible, biodegradable, and capable of surface functionalization, allowing for target-specific delivery and drug release. One of the most common fiber production methods is electrospinning, even though the relatively two-dimensional (2D) tightly packed fiber structures and low production rates have limited its performance. Forcespinning is an alternative spinning technology that generates high-throughput, continuous polymeric nanofibers with 3D structures. Unlike electrospinning, forcespinning generates fibers by centrifugal forces rather than electrostatic forces, resulting in significantly higher fiber production. The functionalization of nanocarriers on nanofibers can result in smart nanofibers with anticancer capabilities that can be activated by external stimuli, such as light. This review addresses current trends and potential applications of light-responsive and dual-stimuli-responsive electro- and forcespun smart nanofibers in cancer therapy, with a particular emphasis on functionalizing nanofiber surfaces and developing nano-in-nanofiber emerging delivery systems for dual-controlled drug release and high-precision tumor targeting. In addition, the progress and prospective diagnostic and therapeutic applications of light-responsive and dual-stimuli-responsive smart nanofibers are discussed in the context of combination cancer therapy.
Collapse
Affiliation(s)
- Ahmed M. Agiba
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey 64849, Mexico; (A.M.A.); (A.O.M.); (A.A.-S.)
| | - Nihal Elsayyad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, October for Modern Sciences and Arts University, Cairo 12451, Egypt;
| | - Hala N. ElShagea
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ahram Canadian University, Cairo 12451, Egypt;
| | - Mahmoud A. Metwalli
- El Demerdash Hospital, Faculty of Medicine, Ain Shams University, Cairo 11591, Egypt;
| | - Amin Orash Mahmoudsalehi
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey 64849, Mexico; (A.M.A.); (A.O.M.); (A.A.-S.)
| | - Saeed Beigi-Boroujeni
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey 64849, Mexico; (A.M.A.); (A.O.M.); (A.A.-S.)
| | - Omar Lozano
- School of Medicine and Health Sciences, Tecnológico de Monterrey, Monterrey 64849, Mexico;
- Institute for Obesity Research, Tecnológico de Monterrey, Monterrey 64849, Mexico
| | - Alan Aguirre-Soto
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey 64849, Mexico; (A.M.A.); (A.O.M.); (A.A.-S.)
| | - Jose Luis Arreola-Ramirez
- Department of Bronchial Hyperresponsiveness, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City 14080, Mexico;
| | - Patricia Segura-Medina
- Department of Bronchial Hyperresponsiveness, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City 14080, Mexico;
- School of Medicine and Health Sciences, Tecnológico de Monterrey, Mexico City 14380, Mexico
| | - Raghda Rabe Hamed
- Department of Industrial Pharmacy, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Cairo 12566, Egypt;
| |
Collapse
|
7
|
Shetty K, Yadav KS. Temozolomide nano-in-nanofiber delivery system with sustained release and enhanced cellular uptake by U87MG cells. Drug Dev Ind Pharm 2024; 50:420-431. [PMID: 38502031 DOI: 10.1080/03639045.2024.2332906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
OBJECTIVE The study was aimed at formulating temozolomide (TMZ) loaded gelatin nanoparticles (GNPs) encapsulated into polyvinyl alcohol (PVA) nanofibers (TMZ-GNPs-PVA NFs) as the nano-in-nanofiber delivery system. The secondary objective was to explore the sustained releasing ability of this system and to assess its enhanced cellular uptake against U87MG glioma cells in vitro. SIGNIFICANCE Nano-in-nanofibers are the emerging drug delivery systems for treating a wide range of diseases including cancers as they overcome the challenges experienced by nanoparticles and nanofibers alone. METHODS The drug-loaded GNPs were formulated by one-step desolvation method. The Design of Experiments (DoE) was used to optimize nanoparticle size and entrapment efficiency. The optimized drug-loaded nanoparticles were then encapsulated within nanofibers using blend electrospinning technique. The U87MG glioma cells were used to investigate the uptake of the formulation. RESULTS A 32 factorial design was used to optimize the mean particle size (145.7 nm) and entrapment efficiency (87.6%) of the TMZ-loaded GNPs which were subsequently ingrained into PVA nanofibers by electrospinning technique. The delivery system achieved a sustained drug release for up to seven days (in vitro). The SEM results ensured that the expected nano-in-nanofiber delivery system was achieved. The uptake of TMZ-GNPs-PVA NFs by cells was increased by a factor of 1.964 compared to that of the pure drug. CONCLUSION The nano-in-nanofiber drug delivery system is a potentially useful therapeutic strategy for the management of glioblastoma multiforme.
Collapse
Affiliation(s)
- Karishma Shetty
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS (Deemed to be University), Mumbai, India
| | - Khushwant S Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS (Deemed to be University), Mumbai, India
| |
Collapse
|
8
|
Kamaraj M, Moghimi N, Chen J, Morales R, Chen S, Khademhosseini A, John JV. New dimensions of electrospun nanofiber material designs for biotechnological uses. Trends Biotechnol 2024; 42:631-647. [PMID: 38158307 PMCID: PMC11065627 DOI: 10.1016/j.tibtech.2023.11.008] [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: 09/30/2023] [Revised: 11/15/2023] [Accepted: 11/15/2023] [Indexed: 01/03/2024]
Abstract
Electrospinning technology has garnered wide attention over the past few decades in various biomedical applications including drug delivery, cell therapy, and tissue engineering. This technology can create nanofibers with tunable fiber diameters and functionalities. However, the 2D membrane nature of the nanofibers, as well as the rigidity and low porosity of electrospun fibers, lower their efficacy in tissue repair and regeneration. Recently, new avenues have been explored to resolve the challenges associated with 2D electrospun nanofiber membranes. This review discusses recent trends in creating different electrospun nanofiber microstructures from 2D nanofiber membranes by using various post-processing methods, as well as their biotechnological applications.
Collapse
Affiliation(s)
- Meenakshi Kamaraj
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Nafiseh Moghimi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Junjie Chen
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Ramon Morales
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Shixuan Chen
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| | - Johnson V John
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| |
Collapse
|
9
|
Liu YC, Chen P, Chang R, Liu X, Jhang JW, Enkhbat M, Chen S, Wang H, Deng C, Wang PY. Artificial tumor matrices and bioengineered tools for tumoroid generation. Biofabrication 2024; 16:022004. [PMID: 38306665 DOI: 10.1088/1758-5090/ad2534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 02/01/2024] [Indexed: 02/04/2024]
Abstract
The tumor microenvironment (TME) is critical for tumor growth and metastasis. The TME contains cancer-associated cells, tumor matrix, and tumor secretory factors. The fabrication of artificial tumors, so-called tumoroids, is of great significance for the understanding of tumorigenesis and clinical cancer therapy. The assembly of multiple tumor cells and matrix components through interdisciplinary techniques is necessary for the preparation of various tumoroids. This article discusses current methods for constructing tumoroids (tumor tissue slices and tumor cell co-culture) for pre-clinical use. This article focuses on the artificial matrix materials (natural and synthetic materials) and biofabrication techniques (cell assembly, bioengineered tools, bioprinting, and microfluidic devices) used in tumoroids. This article also points out the shortcomings of current tumoroids and potential solutions. This article aims to promotes the next-generation tumoroids and the potential of them in basic research and clinical application.
Collapse
Affiliation(s)
- Yung-Chiang Liu
- Oujiang Laboratory; Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang 325024, People's Republic of China
| | - Ping Chen
- Cancer Centre, Faculty of Health Sciences, MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR 999078, People's Republic of China
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Ray Chang
- Oujiang Laboratory; Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang 325024, People's Republic of China
| | - Xingjian Liu
- Oujiang Laboratory; Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang 325024, People's Republic of China
| | - Jhe-Wei Jhang
- Oujiang Laboratory; Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang 325024, People's Republic of China
| | - Myagmartsend Enkhbat
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Shan Chen
- Oujiang Laboratory; Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang 325024, People's Republic of China
| | - Hongxia Wang
- State Key Laboratory of Oncogenes and Related Genes, Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Chuxia Deng
- Cancer Centre, Faculty of Health Sciences, MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR 999078, People's Republic of China
| | - Peng-Yuan Wang
- Oujiang Laboratory; Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang 325024, People's Republic of China
| |
Collapse
|
10
|
Aazmi A, Zhang D, Mazzaglia C, Yu M, Wang Z, Yang H, Huang YYS, Ma L. Biofabrication methods for reconstructing extracellular matrix mimetics. Bioact Mater 2024; 31:475-496. [PMID: 37719085 PMCID: PMC10500422 DOI: 10.1016/j.bioactmat.2023.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/19/2023] Open
Abstract
In the human body, almost all cells interact with extracellular matrices (ECMs), which have tissue and organ-specific compositions and architectures. These ECMs not only function as cellular scaffolds, providing structural support, but also play a crucial role in dynamically regulating various cellular functions. This comprehensive review delves into the examination of biofabrication strategies used to develop bioactive materials that accurately mimic one or more biophysical and biochemical properties of ECMs. We discuss the potential integration of these ECM-mimics into a range of physiological and pathological in vitro models, enhancing our understanding of cellular behavior and tissue organization. Lastly, we propose future research directions for ECM-mimics in the context of tissue engineering and organ-on-a-chip applications, offering potential advancements in therapeutic approaches and improved patient outcomes.
Collapse
Affiliation(s)
- Abdellah Aazmi
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Duo Zhang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 51817, China
| | - Corrado Mazzaglia
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Mengfei Yu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zhen Wang
- Center for Laboratory Medicine, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Huayong Yang
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yan Yan Shery Huang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Liang Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
11
|
O’Meara CH, Nguyen TV, Jafri Z, Boyer M, Shonka DC, Khachigian LM. Personalised Medicine and the Potential Role of Electrospinning for Targeted Immunotherapeutics in Head and Neck Cancer. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 14:6. [PMID: 38202461 PMCID: PMC10780990 DOI: 10.3390/nano14010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/03/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024]
Abstract
Advanced head and neck cancer (HNC) is functionally and aesthetically destructive, and despite significant advances in therapy, overall survival is poor, financial toxicity is high, and treatment commonly exacerbates tissue damage. Although response and durability concerns remain, antibody-based immunotherapies have heralded a paradigm shift in systemic treatment. To overcome limitations associated with antibody-based immunotherapies, exploration into de novo and repurposed small molecule immunotherapies is expanding at a rapid rate. Small molecule immunotherapies also have the capacity for chelation to biodegradable, bioadherent, electrospun scaffolds. This article focuses on the novel concept of targeted, sustained release immunotherapies and their potential to improve outcomes in poorly accessible and risk for positive margin HNC cases.
Collapse
Affiliation(s)
- Connor H. O’Meara
- Department of Otorhinolaryngology, Head & Neck Surgery, The Canberra Hospital, Garran, ACT 2605, Australia
- ANU School of Medicine, Australian National University, Canberra, ACT 0200, Australia
| | - Thanh Vinh Nguyen
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia;
| | - Zuhayr Jafri
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia; (Z.J.)
| | - Michael Boyer
- Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia;
| | - David C. Shonka
- Department of Otolaryngology, Head & Neck Surgery, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Levon M. Khachigian
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia; (Z.J.)
| |
Collapse
|
12
|
Li Q, Wang Z, Shi N, Qi Y, Yao W, Yu J, Lu Y. Application and prospect of the therapeutic strategy of inhibiting cellular senescence combined with pro-regenerative biomaterials in regenerative medicine. SMART MEDICINE 2023; 2:e20230030. [PMID: 39188301 PMCID: PMC11235619 DOI: 10.1002/smmd.20230030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/20/2023] [Indexed: 08/28/2024]
Abstract
Complete regeneration of damaged tissues/organs has always been the ultimate challenge in regenerative medicine. Aging has long been considered the basis of age-related diseases, as senescent cells gradually accumulate in tissues with increasing age, tissues exhibit aging and normal physiological functions are inhibited. In recent years, in damaged tissues, scholars have found that the number of cells with features of cellular senescence continues to increase over time. The accumulation of senescent cells severely hinders the healing of damaged tissues. Furthermore, by clearing senescent cells or inhibiting the aging microenvironment, damaged tissues regained their original regenerative and repair capabilities. On the other hand, various biomaterials have been proved to have good biocompatibility and can effectively support cell regeneration after injury. Combining the two solutions, inhibiting the cellular senescence in damaged tissues and establishing a pro-regenerative environment through biomaterial technology gradually reveals a new, unexpected treatment strategy applied to the field of regenerative medicine. In this review, we first elucidate the main characteristics of senescent cells from morphological, functional and molecular levels, and discuss in detail the process of accumulation of senescent cells in tissues. Then, we will explore in depth how the accumulation of senescent cells after damage affects tissue repair and regeneration at different stages. Finally, we will turn to how to promote tissue regeneration and repair in the field of regenerative medicine by inhibiting cellular senescence combined with biomaterial technology. Our goal is to understand the relationship between cellular senescence and tissue regeneration through this new perspective, and provide valuable references for the development of new therapeutic strategies in the future.
Collapse
Affiliation(s)
- Qianyi Li
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Pôle Sino‐Français de Recherches en Sciences du Vivant et G´enomiqueShanghaiChina
- International Laboratory in Cancer, Aging and HematologyShanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d'Azur UniversityShanghaiChina
| | | | | | - Yang Qi
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wenfei Yao
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jie Yu
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yiming Lu
- Department of EmergencyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Pôle Sino‐Français de Recherches en Sciences du Vivant et G´enomiqueShanghaiChina
- International Laboratory in Cancer, Aging and HematologyShanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d'Azur UniversityShanghaiChina
- Division of Critical CareNanxiang Hospital of Jiading DistrictShanghaiChina
| |
Collapse
|
13
|
Kim M, Panagiotakopoulou M, Chen C, Ruiz SB, Ganesh K, Tammela T, Heller DA. Micro-engineering and nano-engineering approaches to investigate tumour ecosystems. Nat Rev Cancer 2023; 23:581-599. [PMID: 37353679 PMCID: PMC10528361 DOI: 10.1038/s41568-023-00593-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/25/2023] [Indexed: 06/25/2023]
Abstract
The interactions among tumour cells, the tumour microenvironment (TME) and non-tumour tissues are of interest to many cancer researchers. Micro-engineering approaches and nanotechnologies are under extensive exploration for modelling these interactions and measuring them in situ and in vivo to investigate therapeutic vulnerabilities in cancer and extend a systemic view of tumour ecosystems. Here we highlight the greatest opportunities for improving the understanding of tumour ecosystems using microfluidic devices, bioprinting or organ-on-a-chip approaches. We also discuss the potential of nanosensors that can transmit information from within the TME or elsewhere in the body to address scientific and clinical questions about changes in chemical gradients, enzymatic activities, metabolic and immune profiles of the TME and circulating analytes. This Review aims to connect the cancer biology and engineering communities, presenting biomedical technologies that may expand the methodologies of the former, while inspiring the latter to develop approaches for interrogating cancer ecosystems.
Collapse
Affiliation(s)
- Mijin Kim
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA
| | | | - Chen Chen
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional PhD Program in Chemical Biology, Sloan Kettering Institute, New York, NY, USA
| | - Stephen B Ruiz
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Karuna Ganesh
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Tuomas Tammela
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
- Cancer Biology and Genetics Program, Sloan Kettering Institute, New York, NY, USA
| | - Daniel A Heller
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA.
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
14
|
Afrin H, Geetha Bai R, Kumar R, Ahmad SS, Agarwal SK, Nurunnabi M. Oral delivery of RNAi for cancer therapy. Cancer Metastasis Rev 2023; 42:699-724. [PMID: 36971908 PMCID: PMC10040933 DOI: 10.1007/s10555-023-10099-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/14/2023] [Indexed: 03/29/2023]
Abstract
Cancer is a major health concern worldwide and is still in a continuous surge of seeking for effective treatments. Since the discovery of RNAi and their mechanism of action, it has shown promises in targeted therapy for various diseases including cancer. The ability of RNAi to selectively silence the carcinogenic gene makes them ideal as cancer therapeutics. Oral delivery is the ideal route of administration of drug administration because of its patients' compliance and convenience. However, orally administered RNAi, for instance, siRNA, must cross various extracellular and intracellular biological barriers before it reaches the site of action. It is very challenging and important to keep the siRNA stable until they reach to the targeted site. Harsh pH, thick mucus layer, and nuclease enzyme prevent siRNA to diffuse through the intestinal wall and thereby induce a therapeutic effect. After entering the cell, siRNA is subjected to lysosomal degradation. Over the years, various approaches have been taken into consideration to overcome these challenges for oral RNAi delivery. Therefore, understanding the challenges and recent development is crucial to offer a novel and advanced approach for oral RNAi delivery. Herein, we have summarized the delivery strategies for oral delivery RNAi and recent advancement towards the preclinical stages.
Collapse
Affiliation(s)
- Humayra Afrin
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
| | - Renu Geetha Bai
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
- Chair of Biosystems Engineering, Institute of Forestry and Engineering, Estonian University of Life Sciences, Kreutzwaldi 56/1, 51006, Tartu, Estonia
| | - Raj Kumar
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
| | - Sheikh Shafin Ahmad
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
- Aerospace Center (cSETR), University of Texas at El Paso, El Paso, TX, 79965, USA
| | - Sandeep K Agarwal
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Md Nurunnabi
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA.
- Aerospace Center (cSETR), University of Texas at El Paso, El Paso, TX, 79965, USA.
- Biomedical Engineering, College of Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA.
| |
Collapse
|
15
|
Louis L, Chee BS, McAfee M, Nugent M. Electrospun Drug-Loaded and Gene-Loaded Nanofibres: The Holy Grail of Glioblastoma Therapy? Pharmaceutics 2023; 15:1649. [PMID: 37376095 DOI: 10.3390/pharmaceutics15061649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
To date, GBM remains highly resistant to therapies that have shown promising effects in other cancers. Therefore, the goal is to take down the shield that these tumours are using to protect themselves and proliferate unchecked, regardless of the advent of diverse therapies. To overcome the limitations of conventional therapy, the use of electrospun nanofibres encapsulated with either a drug or gene has been extensively researched. The aim of this intelligent biomaterial is to achieve a timely release of encapsulated therapy to exert the maximal therapeutic effect simultaneously eliminating dose-limiting toxicities and activating the innate immune response to prevent tumour recurrence. This review article is focused on the developing field of electrospinning and aims to describe the different types of electrospinning techniques in biomedical applications. Each technique describes how not all drugs or genes can be electrospun with any method; their physico-chemical properties, site of action, polymer characteristics and the desired drug or gene release rate determine the strategy used. Finally, we discuss the challenges and future perspectives associated with GBM therapy.
Collapse
Affiliation(s)
- Lynn Louis
- Materials Research Institute, Faculty of Engineering, Technological University of the Shannon, Midlands Midwest, Athlone Main Campus, N37HD68 Athlone, Ireland
| | - Bor Shin Chee
- Materials Research Institute, Faculty of Engineering, Technological University of the Shannon, Midlands Midwest, Athlone Main Campus, N37HD68 Athlone, Ireland
| | - Marion McAfee
- Centre for Mathematical Modelling and Intelligent Systems for Health and Environment (MISHE), Atlantic Technological University, F91YW50 Sligo, Ireland
| | - Michael Nugent
- Materials Research Institute, Faculty of Engineering, Technological University of the Shannon, Midlands Midwest, Athlone Main Campus, N37HD68 Athlone, Ireland
| |
Collapse
|
16
|
Zaszczyńska A, Niemczyk-Soczynska B, Sajkiewicz P. A Comprehensive Review of Electrospun Fibers, 3D-Printed Scaffolds, and Hydrogels for Cancer Therapies. Polymers (Basel) 2022; 14:5278. [PMID: 36501672 PMCID: PMC9736375 DOI: 10.3390/polym14235278] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/09/2022] Open
Abstract
Anticancer therapies and regenerative medicine are being developed to destroy tumor cells, as well as remodel, replace, and support injured organs and tissues. Nowadays, a suitable three-dimensional structure of the scaffold and the type of cells used are crucial for creating bio-inspired organs and tissues. The materials used in medicine are made of non-degradable and degradable biomaterials and can serve as drug carriers. Developing flexible and properly targeted drug carrier systems is crucial for tissue engineering, regenerative medicine, and novel cancer treatment strategies. This review is focused on presenting innovative biomaterials, i.e., electrospun nanofibers, 3D-printed scaffolds, and hydrogels as a novel approach for anticancer treatments which are still under development and awaiting thorough optimization.
Collapse
Affiliation(s)
| | | | - Paweł Sajkiewicz
- Laboratory of Polymers & Biomaterials, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, 02-106 Warsaw, Poland
| |
Collapse
|
17
|
Fu L, Feng Q, Chen Y, Fu J, Zhou X, He C. Nanofibers for the Immunoregulation in Biomedical Applications. ADVANCED FIBER MATERIALS 2022; 4:1334-1356. [DOI: 10.1007/s42765-022-00191-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/19/2022] [Indexed: 01/06/2025]
|
18
|
Neira-Carrillo A, Zárate IA, Nieto E, Butto-Miranda N, Lobos-González L, Del Campo-Smith M, Palacio DA, Urbano BF. Electrospun Poly(acrylic acid- co-4-styrene sulfonate) as Potential Drug-Eluting Scaffolds for Targeted Chemotherapeutic Delivery Systems on Gastric (AGS) and Breast (MDA-Mb-231) Cancer Cell Lines. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3903. [PMID: 36364679 PMCID: PMC9657868 DOI: 10.3390/nano12213903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 06/16/2023]
Abstract
Potential drug-eluting scaffolds of electrospun poly(acrylic acid-co-styrene sulfonate) P(AA-co-SS) in clonogenic assays using tumorigenic gastric and ovarian cancer cells were tested in vitro. Electrospun polymer nanofiber (EPnF) meshes of PAA and PSSNa homo- and P(AA-co-SS) copolymer composed of 30:70, 50:50, 70:30 acrylic acid (AA) and sodium 4-styrene sulfonate (SSNa) units were performed by electrospinning (ES). The synthesis, structural and morphological characterization of all EPnF meshes were analyzed by optical and electron microscopy (SEM-EDS), infrared spectroscopy (FTIR), contact angle, and X-ray diffraction (XRD) measurements. This study shows that different ratio of AA and SSNa of monomers in P(AA-co-SS) EPnF play a crucial role in clonogenic in vitro assays. We found that 50:50 P(AA-co-SS) EPnF mesh loaded with antineoplastic drugs can be an excellent suppressor of growth-independent anchored capacities in vitro assays and a good subcutaneous drug delivery system for chemotherapeutic medication in vivo model for surgical resection procedures in cancer research.
Collapse
Affiliation(s)
- Andrónico Neira-Carrillo
- Department of Biological and Animal Sciences, Faculty of Veterinary and Animal Sciences, University of Chile, Santa Rosa 11735, La Pintana, Santiago 8820808, Chile
- Advanced Center for Chronic Diseases (ACCDIS), Santiago 380492, Chile
| | - Ignacio A. Zárate
- Department of Biological and Animal Sciences, Faculty of Veterinary and Animal Sciences, University of Chile, Santa Rosa 11735, La Pintana, Santiago 8820808, Chile
| | - Eddie Nieto
- Department of Biological and Animal Sciences, Faculty of Veterinary and Animal Sciences, University of Chile, Santa Rosa 11735, La Pintana, Santiago 8820808, Chile
| | - Nicole Butto-Miranda
- Department of Biological and Animal Sciences, Faculty of Veterinary and Animal Sciences, University of Chile, Santa Rosa 11735, La Pintana, Santiago 8820808, Chile
| | - Lorena Lobos-González
- Advanced Center for Chronic Diseases (ACCDIS), Santiago 380492, Chile
- Center for Regenerative Medicine, Faculty of Medicine, Universidad del Desarrollo, Clínica Alemana, Santiago 7610658, Chile
| | - Matias Del Campo-Smith
- Advanced Center for Chronic Diseases (ACCDIS), Santiago 380492, Chile
- Center for Regenerative Medicine, Faculty of Medicine, Universidad del Desarrollo, Clínica Alemana, Santiago 7610658, Chile
| | - Daniel A. Palacio
- Department of Polymer Chemistry, Faculty of Chemical Science, University of Concepción, Concepción 3349001, Chile
| | - Bruno F. Urbano
- Department of Polymer Chemistry, Faculty of Chemical Science, University of Concepción, Concepción 3349001, Chile
| |
Collapse
|
19
|
Indrakumar J, Sankar S, Madhyastha H, Muthukaliannan GK. Progressive Application of Marine Biomaterials in Targeted Cancer Nanotherapeutics. Curr Pharm Des 2022; 28:3337-3350. [PMID: 35466870 DOI: 10.2174/1381612828666220422091611] [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: 10/12/2021] [Revised: 12/15/2021] [Accepted: 01/04/2022] [Indexed: 01/28/2023]
Abstract
The marine microenvironment harbors many unique species of organisms that produce a plethora of compounds that help mankind cure a wide range of diseases. The diversity of products from the ocean bed serves as potentially healing materials and inert vehicles carrying the drug of interest to the target site. Several composites still lay undiscovered under the blue canopy, which can provide treatment for untreated diseases that keep haunting the earth periodically. Cancer is one such disease that has been of interest to several eminent scientists worldwide due to the heterogenic complexity involved in the disease's pathophysiology. Due to extensive globalization and environmental changes, cancer has become a lifestyle disease continuously increasing exponentially in the current decade. This ailment requires a definite remedy that treats by causing minimal damage to the body's normal cells. The application of nanotechnology in medicine has opened up new avenues of research in targeted therapeutics due to their highly malleable characteristics. Marine waters contain an immense ionic environment that succors the production of distinct nanomaterials with exceptional character, yielding highly flexible molecules to modify, thus facilitating the engineering of targeted biomolecules. This review provides a short insight into an array of marine biomolecules that can be probed into cancer nanotherapeutics sparing healthy cells.
Collapse
Affiliation(s)
- Janani Indrakumar
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Srivarshini Sankar
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Harishkumar Madhyastha
- Department of Medical Sciences, Division of Cardio-Vascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | | |
Collapse
|
20
|
Erickson A, Chiarelli PA, Huang J, Levengood SL, Zhang M. Electrospun nanofibers for 3-D cancer models, diagnostics, and therapy. NANOSCALE HORIZONS 2022; 7:1279-1298. [PMID: 36106417 DOI: 10.1039/d2nh00328g] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
As one of the leading causes of global mortality, cancer has prompted extensive research and development to advance efficacious drug discovery, sustained drug delivery and improved sensitivity in diagnosis. Towards these applications, nanofibers synthesized by electrospinning have exhibited great clinical potential as a biomimetic tumor microenvironment model for drug screening, a controllable platform for localized, prolonged drug release for cancer therapy, and a highly sensitive cancer diagnostic tool for capture and isolation of circulating tumor cells in the bloodstream and for detection of cancer-associated biomarkers. This review provides an overview of applied nanofiber design with focus on versatile electrospinning fabrication techniques. The influence of topographical, physical, and biochemical properties on the function of nanofiber assemblies is discussed, as well as current and foreseeable barriers to the clinical translation of applied nanofibers in the field of oncology.
Collapse
Affiliation(s)
- Ariane Erickson
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA.
| | - Peter A Chiarelli
- The Saban Research Institute, University of Southern California, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Jianxi Huang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA.
| | - Sheeny Lan Levengood
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA.
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
21
|
Gopalakrishnan Usha P, Jalajakumari S, Babukuttan Sheela U, Meena Gopalakrishnan A, Therakathinal Thankappan Nair S. Polysaccharide nanofibers and hydrogel: A comparative evaluation on
3D
cell culture and tumor reduction. J Appl Polym Sci 2022. [DOI: 10.1002/app.53044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Preethi Gopalakrishnan Usha
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research Regional Cancer Centre Thiruvananthapuram Kerala India
| | - Sreekutty Jalajakumari
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research Regional Cancer Centre Thiruvananthapuram Kerala India
| | - Unnikrishnan Babukuttan Sheela
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research Regional Cancer Centre Thiruvananthapuram Kerala India
| | - Archana Meena Gopalakrishnan
- Laboratory of Biopharmaceuticals and Nanomedicine, Division of Cancer Research Regional Cancer Centre Thiruvananthapuram Kerala India
| | | |
Collapse
|
22
|
Pebdeni AB, Hosseini M, Barkhordari A. Smart fluorescence aptasensor using nanofiber functionalized with carbon quantum dot for specific detection of pathogenic bacteria in the wound. Talanta 2022; 246:123454. [DOI: 10.1016/j.talanta.2022.123454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 01/23/2023]
|
23
|
Antunes N, Kundu B, Kundu SC, Reis RL, Correlo V. In Vitro Cancer Models: A Closer Look at Limitations on Translation. Bioengineering (Basel) 2022; 9:166. [PMID: 35447726 PMCID: PMC9029854 DOI: 10.3390/bioengineering9040166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/02/2022] [Accepted: 04/05/2022] [Indexed: 12/18/2022] Open
Abstract
In vitro cancer models are envisioned as high-throughput screening platforms for potential new therapeutic discovery and/or validation. They also serve as tools to achieve personalized treatment strategies or real-time monitoring of disease propagation, providing effective treatments to patients. To battle the fatality of metastatic cancers, the development and commercialization of predictive and robust preclinical in vitro cancer models are of urgent need. In the past decades, the translation of cancer research from 2D to 3D platforms and the development of diverse in vitro cancer models have been well elaborated in an enormous number of reviews. However, the meagre clinical success rate of cancer therapeutics urges the critical introspection of currently available preclinical platforms, including patents, to hasten the development of precision medicine and commercialization of in vitro cancer models. Hence, the present article critically reflects the difficulty of translating cancer therapeutics from discovery to adoption and commercialization in the light of in vitro cancer models as predictive tools. The state of the art of in vitro cancer models is discussed first, followed by identifying the limitations of bench-to-bedside transition. This review tries to establish compatibility between the current findings and obstacles and indicates future directions to accelerate the market penetration, considering the niche market.
Collapse
Affiliation(s)
- Nina Antunes
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, AvePark, Zona Industrial da Gandra, 4805-017 Barco, Portugal; (N.A.); (B.K.); (S.C.K.); (R.L.R.)
- ICVS/3 B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Banani Kundu
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, AvePark, Zona Industrial da Gandra, 4805-017 Barco, Portugal; (N.A.); (B.K.); (S.C.K.); (R.L.R.)
- ICVS/3 B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Subhas C. Kundu
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, AvePark, Zona Industrial da Gandra, 4805-017 Barco, Portugal; (N.A.); (B.K.); (S.C.K.); (R.L.R.)
- ICVS/3 B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Rui L. Reis
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, AvePark, Zona Industrial da Gandra, 4805-017 Barco, Portugal; (N.A.); (B.K.); (S.C.K.); (R.L.R.)
- ICVS/3 B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Vítor Correlo
- Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, AvePark, Zona Industrial da Gandra, 4805-017 Barco, Portugal; (N.A.); (B.K.); (S.C.K.); (R.L.R.)
- ICVS/3 B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| |
Collapse
|
24
|
Hauck M, Hellmold D, Kubelt C, Synowitz M, Adelung R, Schütt F, Held‐Feindt J. Localized Drug Delivery Systems in High‐Grade Glioma Therapy – From Construction to Application. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Margarethe Hauck
- Functional Nanomaterials, Institute for Materials Science Kiel University Kiel 24143 Germany
| | - Dana Hellmold
- Department of Neurosurgery University Medical Center Schleswig‐Holstein UKSH Campus Kiel Kiel 24105 Germany
| | - Carolin Kubelt
- Department of Neurosurgery University Medical Center Schleswig‐Holstein UKSH Campus Kiel Kiel 24105 Germany
| | - Michael Synowitz
- Department of Neurosurgery University Medical Center Schleswig‐Holstein UKSH Campus Kiel Kiel 24105 Germany
| | - Rainer Adelung
- Functional Nanomaterials, Institute for Materials Science Kiel University Kiel 24143 Germany
| | - Fabian Schütt
- Functional Nanomaterials, Institute for Materials Science Kiel University Kiel 24143 Germany
| | - Janka Held‐Feindt
- Department of Neurosurgery University Medical Center Schleswig‐Holstein UKSH Campus Kiel Kiel 24105 Germany
| |
Collapse
|
25
|
Fabrication and Modelling of a Reservoir-Based Drug Delivery System for Customizable Release. Pharmaceutics 2022; 14:pharmaceutics14040777. [PMID: 35456611 PMCID: PMC9025308 DOI: 10.3390/pharmaceutics14040777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 01/27/2023] Open
Abstract
Localized therapy approaches have emerged as an alternative drug administration route to overcome the limitations of systemic therapies, such as the crossing of the blood–brain barrier in the case of brain tumor treatment. For this, implantable drug delivery systems (DDS) have been developed and extensively researched. However, to achieve an effective localized treatment, the release kinetics of DDS needs to be controlled in a defined manner, so that the concentration at the tumor site is within the therapeutic window. Thus, a DDS, with patient-specific release kinetics, is crucial for the improvement of therapy. Here, we present a computationally supported reservoir-based DDS (rDDS) development towards patient-specific release kinetics. The rDDS consists of a reservoir surrounded by a polydimethylsiloxane (PDMS) microchannel membrane. By tailoring the rDDS, in terms of membrane porosity, geometry, and drug concentration, the release profiles can be precisely adapted, with respect to the maximum concentration, release rate, and release time. The release is investigated using a model dye for varying parameters, leading to different distinct release profiles, with a maximum release of up to 60 days. Finally, a computational simulation, considering exemplary in vivo conditions (e.g., exchange of cerebrospinal fluid), is used to study the resulting drug release profiles, demonstrating the customizability of the system. The establishment of a computationally supported workflow, for development towards a patient-specific rDDS, in combination with the transfer to suitable drugs, could significantly improve the efficacy of localized therapy approaches.
Collapse
|
26
|
Recent advancements of electrospun nanofibers for cancer therapy. Polym Bull (Berl) 2022. [DOI: 10.1007/s00289-022-04153-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
27
|
Qu Y, Lu K, Zheng Y, Huang C, Wang G, Zhang Y, Yu Q. Photothermal scaffolds/surfaces for regulation of cell behaviors. Bioact Mater 2022; 8:449-477. [PMID: 34541413 PMCID: PMC8429475 DOI: 10.1016/j.bioactmat.2021.05.052] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/18/2021] [Accepted: 05/31/2021] [Indexed: 12/22/2022] Open
Abstract
Regulation of cell behaviors and even cell fates is of great significance in diverse biomedical applications such as cancer treatment, cell-based therapy, and tissue engineering. During the past decades, diverse methods have been developed to regulate cell behaviors such as applying external stimuli, delivering exogenous molecules into cell interior and changing the physicochemical properties of the substrates where cells adhere. Photothermal scaffolds/surfaces refer to a kind of materials embedded or coated with photothermal agents that can absorb light with proper wavelength (usually in near infrared region) and convert light energy to heat; the generated heat shows great potential for regulation of cell behaviors in different ways. In the current review, we summarize the recent research progress, especially over the past decade, of using photothermal scaffolds/surfaces to regulate cell behaviors, which could be further categorized into three types: (i) killing the tumor cells via hyperthermia or thermal ablation, (ii) engineering cells by intracellular delivery of exogenous molecules via photothermal poration of cell membranes, and (iii) releasing a single cell or an intact cell sheet via modulation of surface physicochemical properties in response to heat. In the end, challenges and perspectives in these areas are commented.
Collapse
Affiliation(s)
- Yangcui Qu
- College of Biomedical Engineering & the Key Laboratory for Medical Functional Nanomaterials, Jining Medical University, Jining, 272067, PR China
| | - Kunyan Lu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, PR China
| | - Yanjun Zheng
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, PR China
| | - Chaobo Huang
- Joint Laboratory of Advanced Biomedical Materials (NFU-UGent), College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, PR China
| | - Guannan Wang
- College of Biomedical Engineering & the Key Laboratory for Medical Functional Nanomaterials, Jining Medical University, Jining, 272067, PR China
| | - Yanxia Zhang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, 215006, PR China
| | - Qian Yu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, PR China
| |
Collapse
|
28
|
Jao D, Hu X, Beachley V. Bioinspired Silk Fiber Spinning System via Automated Track-Drawing. ACS APPLIED BIO MATERIALS 2021; 4:8192-8204. [DOI: 10.1021/acsabm.1c00630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
Safwat S, Hathout RM, Ishak RA, Mortada ND. Elaborated survey in the scope of nanocarriers engineering for boosting chemotherapy cytotoxicity: A meta-analysis study. Int J Pharm 2021; 610:121268. [PMID: 34748812 DOI: 10.1016/j.ijpharm.2021.121268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/24/2021] [Accepted: 11/01/2021] [Indexed: 02/02/2023]
Abstract
Cancer is the prime cause of mortality throughout the world. Although the conventional chemotherapeutic agents damage the cancerous cells, they exert prominent injury to the normal cells owing to their lack of specificity. With advances in science, many research studies have been established to boost the cytotoxic effect of the chemotherapeutic agents via innovating novel nano-formulations having different variables. In the current meta-analysis study, combined data from different research articles were gathered for the evidence-based proof of the superiority of drug loaded nanocarriers over their corresponding conventional solutions in boosting the cytotoxic effect of chemotherapy in terms of IC50 values. The meta-analysis was subdivided into three subgroups; nanoparticles versus nanofibers, surface functionalized nanocarriers versus naked ones, and protein versus non-protein-based platforms. The different subgroups interestingly showed distinct scoring outcome data paving the road for cytotoxicity enhancement of the anti-cancer drugs in an evidence-based manner.
Collapse
Affiliation(s)
- Sally Safwat
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, African Union Organization Street, Abbassia, Cairo, Egypt
| | - Rania M Hathout
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, African Union Organization Street, Abbassia, Cairo, Egypt.
| | - Rania A Ishak
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, African Union Organization Street, Abbassia, Cairo, Egypt
| | - Nahed D Mortada
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, African Union Organization Street, Abbassia, Cairo, Egypt
| |
Collapse
|
30
|
Zhang M, Zhang J, Ran S, Sun W, Zhu Z. Polydopamine-assisted decoration of Se nanoparticles on curcumin-incorporated nanofiber matrices for localized synergistic tumor-wound therapy. Biomater Sci 2021; 10:536-548. [PMID: 34904972 DOI: 10.1039/d1bm01607e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The management of surgical wounds incurred during tumor removal procedures has become a non-negligible issue. Herein, for the first time, an implantable polymer-based nanofiber matrix is developed for postoperative tumor management by promoting wound healing and preventing cancer recurrence. The multifunctional matrix is successfully prepared by assembling chitosan-stabilized Se nanoparticles (SeNPs) at the surface of polydopamine (PDA) modified poly(ε-caprolactone)/curcumin fibres (PCL/CUR), denoted as PCL/CUR/PDA@Se. In this system, PDA as functionalized layers coated onto the PCL/CUR surface favors the effective immobilization of SeNPs through a covalent bond, as well as acts as a gatekeeper guaranteeing the sustained release of CUR. The CUR/SeNPs present excellent antitumor efficacy, respectively, which supports the nanocomposite matrix to efficiently kill cancer cells in vitro by inducing mitochondrial dysfunction caused by the ROS overproduction, and significantly suppressing the tumor growth in vivo. Additionally, due to the synergistic antioxidant activity of CUR and SeNPs, the nanofibrous matrix distinctly facilitates the adhesion and proliferation of normal fibroblast cells, and simultaneously accelerates wound healing during tumor treatments in tumor-bearing mice. These results suggest that the PCL/CUR/PDA@Se matrix with bifunctional properties is a promising candidate for local tumor-wound therapy. This work offers an innovative strategy to develop new improved post-surgery therapies for cancer patients.
Collapse
Affiliation(s)
- Meng Zhang
- Institute of Nano-Science and Nano-Technology, College of Physical Science and Technology, Central China Normal University, Wuhan, 430079, China.
| | - Jiting Zhang
- Institute of Nano-Science and Nano-Technology, College of Physical Science and Technology, Central China Normal University, Wuhan, 430079, China.
| | - Siyi Ran
- Institute of Nano-Science and Nano-Technology, College of Physical Science and Technology, Central China Normal University, Wuhan, 430079, China.
| | - Wei Sun
- Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China
| | - Zhihong Zhu
- Institute of Nano-Science and Nano-Technology, College of Physical Science and Technology, Central China Normal University, Wuhan, 430079, China.
| |
Collapse
|
31
|
Hussain T, Ramakrishna S, Abid S. Nanofibrous drug delivery systems for breast cancer: a review. NANOTECHNOLOGY 2021; 33:102001. [PMID: 34757956 DOI: 10.1088/1361-6528/ac385c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 11/10/2021] [Indexed: 06/13/2023]
Abstract
Breast cancer is the most common type of cancer among women. Breast-conserving surgery (BCS) is one of the preferred approaches for treating non-invasive or early-stage breast cancers. However, local-regional recurrence (LRR) is one of the critical risk factors after BCS. As many as 10%-20% of BCS cases may show LRR within 5 years and almost 50% within 10 years after surgery. Radiation therapy is one of the preferred treatments used to prevent LRR after BCS. However, because of possible side-effects of radiation therapy, targeted drug delivery systems (DDS) based on nanofibers loaded with anti-cancer drugs have been explored in recent years to control LRR. This paper aims to review different polymers and anti-cancer drugs used for developing nanofibrous DDS against various breast cancer cell lines for their efficacy and advantages. It was observed that the utilization of nanofibers scaffolds after mastectomy could decrease the recurrence of breast cancer cells to a great extent as these nanofibrous scaffolds release drugs in a sustained manner for a prolonged time eliminating the need for radiations. Besides, the side effects of chemotherapy or other aggressive anticancer treatment on healthy cells could also be avoided.
Collapse
Affiliation(s)
- Tanveer Hussain
- Electrospun Materials & Polymeric Membranes Research Group, National Textile University, Pakistan
| | - Seeram Ramakrishna
- Center for Nanofibers & Nanotechnology (CNN), National University of Singapore (NUS), Singapore
| | - Sharjeel Abid
- Electrospun Materials & Polymeric Membranes Research Group, National Textile University, Pakistan
| |
Collapse
|
32
|
|
33
|
Multifunctional Electrospun Nanofibers Based on Biopolymer Blends and Magnetic Tubular Halloysite for Medical Applications. Polymers (Basel) 2021; 13:polym13223870. [PMID: 34833169 PMCID: PMC8624944 DOI: 10.3390/polym13223870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 11/17/2022] Open
Abstract
Tubular halloysite (HNT) is a naturally occurring aluminosilicate clay with a unique combination of natural availability, good biocompatibility, high mechanical strength, and functionality. This study explored the effects of magnetically responsive halloysite (MHNT) on the structure, morphology, chemical composition, and magnetic and mechanical properties of electrospun nanofibers based on polycaprolactone (PCL) and gelatine (Gel) blends. MHNT was prepared via a simple modification of HNT with a perchloric-acid-stabilized magnetic fluid–methanol mixture. PCL/Gel nanofibers containing 6, 9, and 12 wt.% HNT and MHNT were prepared via an electrospinning process, respecting the essential rules for medical applications. The structure and properties of the prepared nanofibers were studied using infrared spectroscopy (ATR-FTIR) and electron microscopy (SEM, STEM) along with energy-dispersive X-ray spectroscopy (EDX), magnetometry, and mechanical analysis. It was found that the incorporation of the studied concentrations of MHNT into PCL/Gel nanofibers led to soft magnetic biocompatible materials with a saturation magnetization of 0.67 emu/g and coercivity of 15 Oe for nanofibers with 12 wt.% MHNT. Moreover, by applying both HNT and MHNT, an improvement of the nanofibers structure was observed, together with strong reinforcing effects. The greatest improvement was observed for nanofibers containing 9 wt.% MHNT when increases in tensile strength reached more than two-fold and the elongation at break reached a five-fold improvement.
Collapse
|
34
|
Mohseni Garakani M, Ahangar P, Watson S, Nisol B, Wertheimer MR, Rosenzweig DH, Ajji A. A novel 3D co-culture platform for integrating tissue interfaces for tumor growth, migration and therapeutic sensitivity: “PP-3D-S”. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 134:112566. [DOI: 10.1016/j.msec.2021.112566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/08/2021] [Accepted: 11/22/2021] [Indexed: 02/07/2023]
|
35
|
Hussein MAM, Su S, Ulag S, Woźniak A, Grinholc M, Erdemir G, Erdem Kuruca S, Gunduz O, Muhammed M, El-Sherbiny IM, Megahed M. Development and In Vitro Evaluation of Biocompatible PLA-Based Trilayer Nanofibrous Membranes for the Delivery of Nanoceria: A Novel Approach for Diabetic Wound Healing. Polymers (Basel) 2021; 13:3630. [PMID: 34771187 PMCID: PMC8587307 DOI: 10.3390/polym13213630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 01/22/2023] Open
Abstract
The attempts to explore and optimize the efficiency of diabetic wound healing's promotors are still in progress. Incorporation of cerium oxide nanoparticles (nCeO2) in appropriate nanofibers (NFs) can prolong and maximize their promoting effect for the healing of diabetic wounds, through their sustained releases, as well as the nanofibers role in mimicking of the extra cellular matrix (ECM). The as-prepared nCeO2 were analyzed by using UV-Vis spectroscopy, XRD, SEM-EDX, TEM and FTIR, where TEM and SEM images of both aqueous suspension and powder showed spherical/ovoid-shaped particles. Biodegradable trilayer NFs with cytobiocompatibility were developed to sandwich nCeO2 in PVA NFs as a middle layer where PLA NFs were electrospun as outer bilayer. The nCeO2-loaded trilayer NFs were characterized by SEM, XRD, FTIR and DSC. A two-stage release behavior was observed when the nanoceria was released from the trilayer-based nanofibers; an initial burst release took place, and then it was followed by a sustained release pattern. The mouse embryo fibroblasts, i.e., 3T3 cells, were seeded over the nCeO2-loaded NFs mats to investigate their cyto-biocompatibility. The presence and sustained release of nCeO2 efficiently enhance the adhesion, growth and proliferation of the fibroblasts' populations. Moreover, the incorporation of nCeO2 with a higher amount into the designed trilayer NFs demonstrated a significant improvement in morphological, mechanical, thermal and cyto-biocompatibility properties than lower doses. Overall, the obtained results suggest that designated trilayer nanofibrous membranes would offer a specific approach for the treatment of diabetic wounds through an effective controlled release of nCeO2.
Collapse
Affiliation(s)
- Mohamed Ahmed Mohamady Hussein
- Clinic of Dermatology, University Hospital of RWTH Aachen, 52074 Aachen, Germany;
- Department of Pharmacology, Medical Research Division, National Research Center, Dokki, Cairo 12622, Egypt
| | - Sena Su
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Istanbul 34722, Turkey; (S.S.); (S.U.); (O.G.)
| | - Songul Ulag
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Istanbul 34722, Turkey; (S.S.); (S.U.); (O.G.)
| | - Agata Woźniak
- Laboratory of Molecular Diagnostics, Department of Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk, 80-307 Gdansk, Poland; (A.W.); (M.G.)
| | - Mariusz Grinholc
- Laboratory of Molecular Diagnostics, Department of Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdansk, 80-307 Gdansk, Poland; (A.W.); (M.G.)
| | - Gökce Erdemir
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul 34390, Turkey;
- Molecular Cancer Research Center (ISUMKAM), Istinye University, Istanbul 34010, Turkey
| | - Serap Erdem Kuruca
- Department of Physiology, Faculty of Medicine, Istanbul University, Istanbul 34390, Turkey;
| | - Oguzhan Gunduz
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Istanbul 34722, Turkey; (S.S.); (S.U.); (O.G.)
- Department of Metallurgical and Materials Engineering, Faculty of Technology, Marmara University, Istanbul 34722, Turkey
| | - Mamoun Muhammed
- KTH Royal Institute of Technology, SE-100 44 Stockholm, Sweden;
| | - Ibrahim M. El-Sherbiny
- Nanomedicine Laboratory, Center for Materials Science (CMS), Zewail City of Science and Technology, Giza 12578, Egypt
| | - Mosaad Megahed
- Clinic of Dermatology, University Hospital of RWTH Aachen, 52074 Aachen, Germany;
| |
Collapse
|
36
|
Cheng X, Wei J, Ge Q, Xing D, Zhou X, Qian Y, Jiang G. The optimized drug delivery systems of treating cancer bone metastatic osteolysis with nanomaterials. Drug Deliv 2021; 28:37-53. [PMID: 33336610 PMCID: PMC7751395 DOI: 10.1080/10717544.2020.1856225] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Some cancers such as human breast cancer, prostate cancer, and lung cancer easily metastasize to bone, leading to osteolysis and bone destruction accompanied by a complicated microenvironment. Systemic administration of bisphosphonates (BP) or denosumab is the routine therapy for osteolysis but with non-negligible side effects such as mandibular osteonecrosis and hypocalcemia. Thus, it is imperative to exploit optimized drug delivery systems, and some novel nanotechnology and nanomaterials have opened new horizons for scientists. Targeted and local drug delivery systems can optimize biodistribution depending on nanoparticles (NPs) or microspheres (MS) and implantable biomaterials with the controllable property. Drug delivery kinetics can be optimized by smart and sustained/local drug delivery systems for responsive delivery and sustained delivery. These delicately fabricated drug delivery systems with special matrix, structure, morphology, and modification can minimize unexpected toxicity caused by systemic delivery and achieve desired effects through integrating multiple drugs or multiple functions. This review summarized recent studies about optimized drug delivery systems for the treatment of cancer metastatic osteolysis, aimed at giving some inspiration in designing efficient multifunctional drug delivery systems.
Collapse
Affiliation(s)
- Xi Cheng
- Department of Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jinrong Wei
- Department of Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Qi Ge
- Department of Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Danlei Xing
- Department of Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xuefeng Zhou
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, People's Republic of China
| | - Yunzhu Qian
- Center of Stomatology, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Guoqin Jiang
- Department of Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
37
|
Kaur H, Kesharwani P. Advanced nanomedicine approaches applied for treatment of skin carcinoma. J Control Release 2021; 337:589-611. [PMID: 34364919 DOI: 10.1016/j.jconrel.2021.08.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022]
Abstract
Skin-cancer is the commonest malignancy affecting huge proportion of the population, reaching heights in terms of morbidity. The treatment strategies are presently focusing on surgery, radiation and chemotherapy, which eventually cause destruction to unaffected cells. To overcome this limitation, wide range of nanoscaled materials have been recognized as potential carriers for delivering selective response to cancerous cells and neoplasms. Nanotechnological approach has been tremendously exploited in several areas, owing to their functional nanometric dimensions. The alarming incidence of skin cancer engenders burdensome effects worldwide, which is further awakening innovational medicinal approaches, accompanying target specific drug delivery tools for coveted benefits to provide reduced toxicity and tackle proliferative episodes of skin cancer. The developed nanosystems for anti-cancer agents include liposomes, ethosomes, nanofibers, solid lipid nanoparticles and metallic nanoparticles, which exhibit pronounced outcomes for skin carcinoma. In this review, skin cancer with its sub-types is explained in nutshell, followed by compendium of specific nanotechnological tools presented, in addition to therapeutic applications of drug-loaded nano systems for skin cancer.
Collapse
Affiliation(s)
- Harsimran Kaur
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
38
|
Xu L, Li W, Sadeghi-Soureh S, Amirsaadat S, Pourpirali R, Alijani S. Dual drug release mechanisms through mesoporous silica nanoparticle/electrospun nanofiber for enhanced anticancer efficiency of curcumin. J Biomed Mater Res A 2021; 110:316-330. [PMID: 34378328 DOI: 10.1002/jbm.a.37288] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/18/2021] [Accepted: 07/23/2021] [Indexed: 12/11/2022]
Abstract
Electrospun nanofibers (NFs)-based drug delivery approaches are of particular interest as a hopeful implantable nanoplatform for localized cancer therapy and treating tissue defect after resection, allowing the on-site drug delivery with minimal side effect to healthy cells. To maintain therapeutic concentrations of anticancer molecules for a relatively long time through a combination of burst and sustained drug release mechanisms, a hybrid of polycaprolactone and gelatin (PCL/GEL) was used for co-encapsulation of free curcumin (CUR) and CUR-loaded mesoporous silica nanoparticles (CUR@MSNs) via electrospinning, resulting in a novel drug-loaded nanofibrous scaffold, CUR/CUR@MSNs-NFs. The as-prepared MSNs and composite NFs were characterized via TGA, FTIR, FE-SEM, TEM, and BET. In vitro release profile of CUR from CUR/CUR@MSNs-NFs was examined, and the in vitro antitumor efficacy against MDA-MB-231 breast cancer cells was also evaluated through MTT, scratch assay, DAPI staining, and real-time PCR. The results disclosed that the smooth, bead-free, and randomly oriented CUR/CUR@MSNs-NFs displayed a combination of initial rapid discharge and sustained release for CUR, which led to higher cytotoxicity, lower migration as well as a more pronounced effect on apoptosis induction than CUR-NFs and CUR@MSNs-NFs. The present study illustrated that the dual drug release mechanisms through MSN/NF-mediated drug delivery systems might have a highly hopeful application as a localized implantable scaffold for potential postoperative breast cancer therapy.
Collapse
Affiliation(s)
- Liguo Xu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Wei Li
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | | | - Soumaye Amirsaadat
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raheleh Pourpirali
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Alijani
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
39
|
Hot or cold: Bioengineering immune contextures into in vitro patient-derived tumor models. Adv Drug Deliv Rev 2021; 175:113791. [PMID: 33965462 DOI: 10.1016/j.addr.2021.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
In the past decade, immune checkpoint inhibitors (ICI) have proven to be tremendously effective for a subset of cancer patients. However, it is difficult to predict the response of individual patients and efforts are now directed at understanding the mechanisms of ICI resistance. Current models of patient tumors poorly recapitulate the immune contexture, which describe immune parameters that are associated with patient survival. In this Review, we discuss parameters that influence the induction of different immune contextures found within tumors and how engineering strategies may be leveraged to recapitulate these contextures to develop the next generation of immune-competent patient-derived in vitro models.
Collapse
|
40
|
Affolter A, Lammert A, Kern J, Scherl C, Rotter N. Precision Medicine Gains Momentum: Novel 3D Models and Stem Cell-Based Approaches in Head and Neck Cancer. Front Cell Dev Biol 2021; 9:666515. [PMID: 34307351 PMCID: PMC8296983 DOI: 10.3389/fcell.2021.666515] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
Despite the current progress in the development of new concepts of precision medicine for head and neck squamous cell carcinoma (HNSCC), in particular targeted therapies and immune checkpoint inhibition (CPI), overall survival rates have not improved during the last decades. This is, on the one hand, caused by the fact that a significant number of patients presents with late stage disease at the time of diagnosis, on the other hand HNSCC frequently develop therapeutic resistance. Distinct intratumoral and intertumoral heterogeneity is one of the strongest features in HNSCC and has hindered both the identification of specific biomarkers and the establishment of targeted therapies for this disease so far. To date, there is a paucity of reliable preclinical models, particularly those that can predict responses to immune CPI, as these models require an intact tumor microenvironment (TME). The "ideal" preclinical cancer model is supposed to take both the TME as well as tumor heterogeneity into account. Although HNSCC patients are frequently studied in clinical trials, there is a lack of reliable prognostic biomarkers allowing a better stratification of individuals who might benefit from new concepts of targeted or immunotherapeutic strategies. Emerging evidence indicates that cancer stem cells (CSCs) are highly tumorigenic. Through the process of stemness, epithelial cells acquire an invasive phenotype contributing to metastasis and recurrence. Specific markers for CSC such as CD133 and CD44 expression and ALDH activity help to identify CSC in HNSCC. For the majority of patients, allocation of treatment regimens is simply based on histological diagnosis and on tumor location and disease staging (clinical risk assessments) rather than on specific or individual tumor biology. Hence there is an urgent need for tools to stratify HNSCC patients and pave the way for personalized therapeutic options. This work reviews the current literature on novel approaches in implementing three-dimensional (3D) HNSCC in vitro and in vivo tumor models in the clinical daily routine. Stem-cell based assays will be particularly discussed. Those models are highly anticipated to serve as a preclinical prediction platform for the evaluation of stable biomarkers and for therapeutic efficacy testing.
Collapse
Affiliation(s)
- Annette Affolter
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | | | | | | |
Collapse
|
41
|
Hsu MY, Hsieh CH, Huang YT, Chu SY, Chen CM, Lee WJ, Liu SJ. Enhanced Paclitaxel Efficacy to Suppress Triple-Negative Breast Cancer Progression Using Metronomic Chemotherapy with a Controlled Release System of Electrospun Poly-d-l-Lactide-Co-Glycolide (PLGA) Nanofibers. Cancers (Basel) 2021; 13:cancers13133350. [PMID: 34283075 PMCID: PMC8268060 DOI: 10.3390/cancers13133350] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Treatment of metastatic triple-negative breast cancer (TNBC) relies on chemotherapy. To improve the efficacy of chemotherapy and avoid systemic toxicity, metronomic chemotherapy using continuous administration of low-dose chemotherapy could be a solution. The paclitaxel-loaded PLGA nanofibers allow for continuous and prolonged drug release, which is compatible with the concept of metronomic chemotherapy. The animal study revealed that the strategy successfully inhibited the growth of the primary tumor and distant metastasis without sarcopenia. These data offer new insights into the role of drug-loaded nanofibers in the treatment of metastatic TNBC. Abstract Triple-negative breast cancer (TNBC) is highly aggressive and responds poorly to conventional chemotherapy. The challenge of TNBC therapy is to maximize the efficacies of conventional chemotherapeutic agents and reduce their toxicities. Metronomic chemotherapy using continuous low-dose chemotherapy has been proposed as a new treatment option, but this approach is limited by the selection of drugs. To improve antitumor therapeutic effects, we developed electrospun paclitaxel-loaded poly-d-l-lactide-co-glycolide (PLGA) nanofibers as a topical implantable delivery device for controlled drug release and site-specific treatment. The subcutaneously implanted paclitaxel-loaded nanofibrous membrane in mice was compatible with the concept of metronomic chemotherapy; it significantly enhanced antitumor activity, inhibited local tumor growth, constrained distant metastasis, and prolonged survival compared with intraperitoneal paclitaxel injection. Furthermore, under paclitaxel-loaded nanofiber treatment, systemic toxicity was low with a persistent increase in lean body weight in mice; in contrast, body weight decreased in other groups. The paclitaxel-loaded nanofibrous membranes provided sustained drug release and site-specific treatment by directly targeting and changing the tumor microenvironment, resulting in low systemic toxicity and a significant improvement in the therapeutic effect and safety compared with conventional chemotherapy. Thus, metronomic chemotherapy with paclitaxel-loaded nanofibrous membranes offers a promising strategy for the treatment of TNBC.
Collapse
Affiliation(s)
- Ming-Yi Hsu
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (M.-Y.H.); (Y.-T.H.); (S.-Y.C.); (C.-M.C.)
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Cheng-Hsien Hsieh
- Department of Emergency Medicine, En-Chu-Kong Hospital, New Taipei City 23741, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Ting Huang
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (M.-Y.H.); (Y.-T.H.); (S.-Y.C.); (C.-M.C.)
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Sung-Yu Chu
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (M.-Y.H.); (Y.-T.H.); (S.-Y.C.); (C.-M.C.)
| | - Chien-Ming Chen
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (M.-Y.H.); (Y.-T.H.); (S.-Y.C.); (C.-M.C.)
| | - Wei-Jiunn Lee
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei 11695, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (W.-J.L.); (S.-J.L.); Tel.: +886-2-2930-7930 (ext. 2551/2547) (W.-J.L.); +886-3-2118166 (S.-J.L.)
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
- Correspondence: (W.-J.L.); (S.-J.L.); Tel.: +886-2-2930-7930 (ext. 2551/2547) (W.-J.L.); +886-3-2118166 (S.-J.L.)
| |
Collapse
|
42
|
Wang H, Jin Y, Chen Y, Luo Y, Lv S, Li M, Tao Y. Multifunctional hybrid sponge for in situ postoperative management to inhibit tumor recurrence. Biomater Sci 2021; 9:4066-4075. [PMID: 33908452 DOI: 10.1039/d1bm00085c] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Disseminated tumor cells in bleeding and residual tumor cells in the resection tumor site are the primary factors that result in tumor recurrence after surgery. Safe and efficient local implantation of the drug depot system into the resection cavity to inhibit tumor recurrence would be of great benefit to reduce the mortality of postoperative patients. Here, a sandwich-like doxorubicin-triptolide-loaded fiber/(chitosan/gelatin) sponge, DTF/CGS, is fabricated, combining hemostatic, antibacterial, and chemotherapeutic capability. The CGS obtained via freeze-drying can efficiently prevent bleeding; meanwhile, the metastatic residual tumor cells are stuck with the clotted absorbed blood. Subsequently, dual drugs released from the electrospun fiber can further kill the stuck tumor cells in CGS and the disseminated tumor cells to significantly inhibit the tumor recurrence. This antitumor recurrence strategy by immediately implanting a multifunctional hybrid sponge for in situ postoperative management may possess great potential for preventing tumor recurrence.
Collapse
Affiliation(s)
- Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Yuanyuan Jin
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Yanyan Chen
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Yun Luo
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Shixian Lv
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. and Guangdong Provincial Key Laboratory of Liver Disease, Guangzhou 510630, China
| |
Collapse
|
43
|
Su CY, Burchett A, Dunworth M, Choi JS, Ewald AJ, Ahn EH, Kim DH. Engineering a 3D collective cancer invasion model with control over collagen fiber alignment. Biomaterials 2021; 275:120922. [PMID: 34126408 DOI: 10.1016/j.biomaterials.2021.120922] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 12/21/2022]
Abstract
Prior to cancer cell invasion, the structure of the extracellular matrix (ECM) surrounding the tumor is remodeled, such that circumferentially oriented matrix fibers become radially aligned. This predisposed radially aligned matrix structure serves as a critical regulator of cancer invasion. However, a biomimetic 3D model recapitulating a tumor's behavioral response to these ECM structures is not yet available. In this study, we have developed a phase-specific, force-guided method to establish a 3D dual topographical tumor model in which each tumor spheroid/organoid is surrounded by radially aligned collagen I fibers on one side and circumferentially oriented fibers on the opposite side. A coaxial rotating cylinder system was employed to construct the dual fiber topography and to pre-seed tumor spheroids/organoids within a single device. This system enables the application of different force mechanisms in the nucleation and elongation phases of collagen fiber polymerization to guide fiber alignment. In the nucleation phase, fiber alignment is enhanced by a horizontal laminar Couette flow driven by the inner cylinder rotation. In the elongation phase, fiber growth is guided by a vertical gravitational force to form a large aligned collagen matrix gel (35 × 25 × 0.5 mm) embedded with >1000 tumor spheroids. The fibers above each tumor spheroid are radially aligned along the direction of gravitational force in contrast to the circumferentially oriented fibers beneath each tumor spheroid/organoid, where the presence of the tumor interferes with the gravity-induced fiber alignment. After tumor invasion, there are more disseminated multicellular clusters on the radially aligned side, compared to the side of the tumor spheroid/organoid facing circumferentially oriented fibers. These results indicate that our 3D dual topographical model recapitulates the preference of tumors to invade and disseminate along radially aligned fibers. We anticipate that this 3D dual topographical model will have broad utility to those studying collective tumor invasion and that it has the potential to identify cancer invasion-targeted therapeutic agents.
Collapse
Affiliation(s)
- Chia-Yi Su
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alice Burchett
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Matthew Dunworth
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jong Seob Choi
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Andrew J Ewald
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Cell Biology and Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Eun Hyun Ahn
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
44
|
Akpan UM, Pellegrini M, Salifu AA, Obayemi JD, Ezenwafor T, Browe D, Ani CJ, Danyuo Y, Dozie-Nwachukwu S, Odusanya OS, Freeman J, Soboyejo WO. In vitro studies of Annona muricata L. extract-loaded electrospun scaffolds for localized treatment of breast cancer. J Biomed Mater Res B Appl Biomater 2021; 109:2041-2056. [PMID: 33960623 DOI: 10.1002/jbm.b.34852] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 04/02/2021] [Accepted: 04/08/2021] [Indexed: 01/05/2023]
Abstract
This paper presents in vitro studies of the sustained release of Annona muricata leaf extracts (AME) from hybrid electrospun fibers for breast cancer treatment. Electrospun hybrid scaffolds were fabricated from crude AME extracts, poly(lactic-co-glycolic acid)/gelatin (PLGA/Ge) and pluronic F127. The physicochemical properties of the AME extract and scaffolds were studied. The antiproliferative effects of the scaffolds were also assessed on breast cancer (MCF-7 and MDA-MB-231) and non-tumorigenic breast (MCF10A) cell lines. Scanning electron microscope micrographs revealed a random network of micro- and submicron fibers. In vitro drug release profiles, governed by quasi-Fickian diffusion at pH 7.4 and non-Fickian super case II at pH 6.7, showed initial burst AME release from the PLGA/Ge-AME and PLGA/Ge-F127/AME fibers at pH 7.4, and burst release from PLGA/Ge-F127/AME (not observed from PLGA/Ge-AME) at pH 6.7. Then, a slower, sustained release of the remaining AME from the fibers, attributed to the onset of degradation of the PLGA/Ge backbone, was observed for the next 72 hr. The cumulative release of AME was 89.33 ± 0.73% (PLGA/Ge-AME) and 51.17 ± 7.96% (PLGA/Ge-F127/AME) at pH 7.4, and 9.27 ± 2.3% and 73.5 ± 4.5%, respectively, at pH 6.7. Pluronic F127 addition increased the drug loading capacity and prolonged the sustained AME release from the fibers. The released AME significantly inhibited the in vitro growth of the breast cancer cells more than the non-tumorigenic cells, due to the induction of apoptosis, providing evidence for using pluronic F127-containing electrospun fibers for sustained and localized AME delivery to breast cancer cells.
Collapse
Affiliation(s)
- Udom M Akpan
- Department of Materials Science and Engineering, African University of Science and Technology, Abuja, Nigeria.,Scientific Equipment Development Institute, Minna, Niger State (SEDI-M), National Agency for Science and Engineering Infrastructure (NASENI), Abuja, Nigeria
| | - Michael Pellegrini
- Department of Biomedical Engineering, Rutgers University, New Brunswick, New Jersey, USA
| | - Ali A Salifu
- Department of Mechanical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA.,Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - John D Obayemi
- Department of Mechanical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA.,Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Theresa Ezenwafor
- Department of Materials Science and Engineering, African University of Science and Technology, Abuja, Nigeria
| | - Daniel Browe
- Department of Biomedical Engineering, Rutgers University, New Brunswick, New Jersey, USA
| | - Chukwuemeka J Ani
- Department of Theoretical and Applied Physics, African University of Science and Technology, Abuja, Nigeria
| | - Yiporo Danyuo
- Department of Mechanical Engineering, Ashesi University, Accra, Ghana
| | - Stella Dozie-Nwachukwu
- Biotechnology and Genetic Engineering Advanced Laboratory, Sheda Science and Technology Complex (SHESTCO), Abuja, Nigeria
| | - Olushola S Odusanya
- Biotechnology and Genetic Engineering Advanced Laboratory, Sheda Science and Technology Complex (SHESTCO), Abuja, Nigeria
| | - Joseph Freeman
- Department of Biomedical Engineering, Rutgers University, New Brunswick, New Jersey, USA
| | - Winston O Soboyejo
- Department of Mechanical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA.,Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| |
Collapse
|
45
|
Doostmohammadi M, Forootanfar H, Ramakrishna S. New Strategies for Safe Cancer Therapy Using Electrospun Nanofibers: A Short Review. Mini Rev Med Chem 2021; 20:1272-1286. [PMID: 32400330 DOI: 10.2174/1389557520666200513120924] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/10/2019] [Accepted: 02/14/2020] [Indexed: 12/26/2022]
Abstract
Electrospun nanofibers regarding their special features, including high drug loading capacity, high surface to volume area, flexibility, and ease of production and operation, are of great interest for being used in tissue engineering, and drug delivery approaches. In this context, several studies have been done for the production of biodegradable and biocompatible scaffolds containing different anticancer agents for fighting with solid tumors. Surprisingly, these scaffolds are able to deliver different combinations of drugs and agents, such as nanoparticles and release them in a time dependent manner. Here in this review, we summarize the principles of electrospinning and their uses in entrapment of drugs and anti-proliferative agents suitable for cancer therapy. The latest studies performed on treating cancer using electrospinning are mentioned and their advantages and disadvantages over conventional treatment methods are discussed.
Collapse
Affiliation(s)
- Mohsen Doostmohammadi
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Forootanfar
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| |
Collapse
|
46
|
Frazier T, Williams C, Henderson M, Duplessis T, Rogers E, Wu X, Hamel K, Martin EC, Mohiuddin O, Shaik S, Devireddy R, Rowan BG, Hayes DJ, Gimble JM. Breast Cancer Reconstruction: Design Criteria for a Humanized Microphysiological System. Tissue Eng Part A 2021; 27:479-488. [PMID: 33528293 PMCID: PMC8196546 DOI: 10.1089/ten.tea.2020.0372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 01/21/2021] [Indexed: 11/12/2022] Open
Abstract
International regulatory agencies such as the Food and Drug Administration have mandated that the scientific community develop humanized microphysiological systems (MPS) as an in vitro alternative to animal models in the near future. While the breast cancer research community has long appreciated the importance of three-dimensional growth dynamics in their experimental models, there are remaining obstacles preventing a full conversion to humanized MPS for drug discovery and pathophysiological studies. This perspective evaluates the current status of human tissue-derived cells and scaffolds as building blocks for an "idealized" breast cancer MPS based on bioengineering design principles. It considers the utility of adipose tissue as a potential source of endothelial, lymphohematopoietic, and stromal cells for the support of breast cancer epithelial cells. The relative merits of potential MPS scaffolds derived from adipose tissue, blood components, and synthetic biomaterials is evaluated relative to the current "gold standard" material, Matrigel, a murine chondrosarcoma-derived basement membrane-enriched hydrogel. The advantages and limitations of a humanized breast cancer MPS are discussed in the context of in-process and destructive read-out assays. Impact statement Regulatory authorities have highlighted microphysiological systems as an emerging tool in breast cancer research. This has been led by calls for more predictive human models and reduced animal experimentation. This perspective describes how human-derived cells, extracellular matrices, and hydrogels will provide the building blocks to create breast cancer models that accurately reflect diversity at multiple levels, that is, patient ethnicity, pathophysiology, and metabolic status.
Collapse
Affiliation(s)
| | - Christopher Williams
- Division of Basic Pharmaceutical Sciences, Xavier University of Louisiana, New Orleans, Louisiana, USA
| | | | - Tamika Duplessis
- Department of Physical Sciences, Delgado Community College, New Orleans, Louisiana, USA
| | - Emma Rogers
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
| | - Xiying Wu
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
| | - Katie Hamel
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Elizabeth C. Martin
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Omair Mohiuddin
- Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Science, University of Karachi, Karachi, Pakistan
| | - Shahensha Shaik
- Cell and Molecular Biology Core Laboratory, Xavier University of Louisiana, New Orleans, Louisiana, USA
| | - Ram Devireddy
- Department of Mechanical Engineering, Louisiana State University, New Orleans, Louisiana, USA
| | - Brian G. Rowan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Daniel J. Hayes
- Department of Biomedical Engineering, Pennsylvania State University, State College, Pennsylvania, USA
| | | |
Collapse
|
47
|
Tavakoli F, Ghasemikhah R, Shafiee H. Customizing the Protoscolicidal Activity by a Drug Delivery System: Application of Guar Gum in Electrospun Nanofibers. IRANIAN JOURNAL OF PARASITOLOGY 2021; 16:136-145. [PMID: 33786055 PMCID: PMC7988667 DOI: 10.18502/ijpa.v16i1.5532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background The present study aimed to control mebendazole drug release from ethyl cellulose nanofibers containing guar gum produced by Electrospinning Method (ESM) on mortality of hydatid cyst protoscoleces under laboratory conditions. Methods The study was conducted in Arak Islamic Azad University, 2019. After preparation of ethyl cellulose nanofibers containing guar gum with concentrations 10, 250, 50 and 500 ppm with ESM, the uniformity and fineness of nanofibers were investigated by electron microscope. By determining the absorption of nanofibers during 312 h via spectrophotometry method, the amount of drug release was obtained. Then, the mortality of live protoscoleces in-vitro with nanofibers made with different concentrations was studied during 13 days. Results Guar gum nanofiber with four concentrations of 10, 50, 250 and 500 ppm had 0.78512, 0.83729, 1.0098 and 1.0633 absorption respectively and showed drug release 42.09%, 39.95%, 33.05% and 30.96% after 312 hours. Therefore, the survival of protoscoleces in the presence of guar gum with four concentrations was zero after 3, 6, 11 and 13 days (P<0.05). Conclusion To produce nanofibers carrying the drug for research related to the treatment of hydatid cysts, the electrospinning technique can be considered as a reliable method.
Collapse
Affiliation(s)
- Fatemeh Tavakoli
- Department of Chemistry, Faculty of Science, Arak Branch, Islamic Azad University, Arak, Iran
| | - Reza Ghasemikhah
- Department of Parasitology & Mycology, School of Medicine, Arak University of Medical Sciences, Arak. Iran
| | - Hadi Shafiee
- Department of Chemistry, Faculty of Science, Arak Branch, Islamic Azad University, Arak, Iran.,Department of Parasitology & Mycology, School of Medicine, Arak University of Medical Sciences, Arak. Iran
| |
Collapse
|
48
|
Blanco‐Fernandez B, Gaspar VM, Engel E, Mano JF. Proteinaceous Hydrogels for Bioengineering Advanced 3D Tumor Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003129. [PMID: 33643799 PMCID: PMC7887602 DOI: 10.1002/advs.202003129] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/13/2020] [Indexed: 05/14/2023]
Abstract
The establishment of tumor microenvironment using biomimetic in vitro models that recapitulate key tumor hallmarks including the tumor supporting extracellular matrix (ECM) is in high demand for accelerating the discovery and preclinical validation of more effective anticancer therapeutics. To date, ECM-mimetic hydrogels have been widely explored for 3D in vitro disease modeling owing to their bioactive properties that can be further adapted to the biochemical and biophysical properties of native tumors. Gathering on this momentum, herein the current landscape of intrinsically bioactive protein and peptide hydrogels that have been employed for 3D tumor modeling are discussed. Initially, the importance of recreating such microenvironment and the main considerations for generating ECM-mimetic 3D hydrogel in vitro tumor models are showcased. A comprehensive discussion focusing protein, peptide, or hybrid ECM-mimetic platforms employed for modeling cancer cells/stroma cross-talk and for the preclinical evaluation of candidate anticancer therapies is also provided. Further development of tumor-tunable, proteinaceous or peptide 3D microtesting platforms with microenvironment-specific biophysical and biomolecular cues will contribute to better mimic the in vivo scenario, and improve the predictability of preclinical screening of generalized or personalized therapeutics.
Collapse
Affiliation(s)
- Barbara Blanco‐Fernandez
- Department of Chemistry, CICECO – Aveiro Institute of Materials, University of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute of Science and TechnologyBaldiri Reixac 10–12Barcelona08028Spain
| | - Vítor M. Gaspar
- Department of Chemistry, CICECO – Aveiro Institute of Materials, University of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| | - Elisabeth Engel
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute of Science and TechnologyBaldiri Reixac 10–12Barcelona08028Spain
- Materials Science and Metallurgical EngineeringPolytechnical University of Catalonia (UPC)Eduard Maristany 16Barcelona08019Spain
- CIBER en BioingenieríaBiomateriales y NanomedicinaCIBER‐BBNMadrid28029Spain
| | - João F. Mano
- Department of Chemistry, CICECO – Aveiro Institute of Materials, University of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| |
Collapse
|
49
|
Xu L, Li R, Wang Z, Cui H, Li W, Yu M, Guo SS, Zhao XZ. Electrospun degradable Zn-Mn oxide hierarchical nanofibers for specific capture and efficient release of circulating tumor cells. NANOTECHNOLOGY 2020; 31:495102. [PMID: 32990263 DOI: 10.1088/1361-6528/abb48b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Constructing biological affinity devices is considered as an effective strategy for isolating circulating tumor cells (CTCs), and electrospun nanofibers (ESNFs) have recently received attention. However, the current research focuses on polymer fibers, and fabricating stimuli-responsive inorganic nanofibers for cancer diagnosis and analysis is still challenging. In this work, Zn-Mn oxide nanofibers (ZnMnNFs) are used to capture and purify cancer cells after modification with specific antibodies. Then, the hierarchical nanofibers are degraded by reductive weak acid to release the captured cells efficiently without residues. Fusion of Zn and Mn, two transition metals, enhances the surface activity of oxides so that ZnMnNFs are easier to be degraded and modified. By using MCF-7 cancer cells, the cell capture efficiency of ZnMnNFs is up to 88.2%. Furthermore, by using citric acid, it is discovered that, by comparison with Mn oxide nanofibers, the cell release efficiency of ZnMnNFs is improved to 95.1% from 15.4%. In addition, the viability of released cells exceeds 90%. Lastly, the robustness of ZnMnNFs substrates is tested in peripheral blood from breast cancer patients (BCP) and colorectal cancer patients (CCP). Combined with fluorescence labeling, CTCs are confirmed to be isolated from all the clinical samples. This is the first trial of using ternary inorganic ESNFs for cancer cell capture. It is anticipated that the degradable ESNFs will provide biocompatible theranostic platforms and overcome the current limitations of cell release for high-precision gene analysis.
Collapse
Affiliation(s)
- Longguang Xu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Rui Li
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Zixiang Wang
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Heng Cui
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Wei Li
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Mingxia Yu
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, People's Republic of China
| | - Shi-Shang Guo
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | - Xing-Zhong Zhao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| |
Collapse
|
50
|
Vanza JD, Patel RB, Patel MR. Nanocarrier centered therapeutic approaches: Recent developments with insight towards the future in the management of lung cancer. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.102070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|