1
|
Hu X, Hu J, Pang Y, Wang M, Zhou W, Xie X, Zhu C, Wang X, Sun X. Application of nano-radiosensitizers in non-small cell lung cancer. Front Oncol 2024; 14:1372780. [PMID: 38646428 PMCID: PMC11027897 DOI: 10.3389/fonc.2024.1372780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/07/2024] [Indexed: 04/23/2024] Open
Abstract
Radiotherapy stands as a cornerstone in the treatment of numerous malignant tumors, including non-small cell lung cancer. However, the critical challenge of amplifying the tumoricidal effectiveness of radiotherapy while minimizing collateral damage to healthy tissues remains an area of significant research interest. Radiosensitizers, by methods such as amplifying DNA damage and fostering the creation of free radicals, play a pivotal role in enhancing the destructive impact of radiotherapy on tumors. Over recent decades, nano-dimensional radiosensitizers have emerged as a notable advancement. Their mechanisms include cell cycle arrest in the G2/M phase, combating tumor hypoxia, and others, thereby enhancing the efficacy of radiotherapy. This review delves into the evolving landscape of nanomaterials used for radiosensitization in non-small cell lung cancer. It provides insights into the current research progress and critically examines the challenges and future prospects within this burgeoning field.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiaonan Sun
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| |
Collapse
|
2
|
Zhou Z, Zhang Y, Zeng Y, Yang D, Mo J, Zheng Z, Zhang Y, Xiao P, Zhong X, Yan W. Effects of Nanomaterials on Synthesis and Degradation of the Extracellular Matrix. ACS NANO 2024; 18:7688-7710. [PMID: 38436232 DOI: 10.1021/acsnano.3c09954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Extracellular matrix (ECM) remodeling is accompanied by the continuous synthesis and degradation of the ECM components. This dynamic process plays an important role in guiding cell adhesion, migration, proliferation, and differentiation, as well as in tissue development, body repair, and maintenance of homeostasis. Nanomaterials, due to their photoelectric and catalytic properties and special structure, have garnered much attention in biomedical fields for use in processes such as tissue engineering and disease treatment. Nanomaterials can reshape the cell microenvironment by changing the synthesis and degradation of ECM-related proteins, thereby indirectly changing the behavior of the surrounding cells. This review focuses on the regulatory role of nanomaterials in the process of cell synthesis of different ECM-related proteins and extracellular protease. We discuss influencing factors and possible related mechanisms of nanomaterials in ECM remodeling, which may provide different insights into the design and development of nanomaterials for the treatment of ECM disorder-related diseases.
Collapse
Affiliation(s)
- Zhiyan Zhou
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yanli Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510260, China
| | - Yuting Zeng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Dehong Yang
- Department of Orthopedics - Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiayao Mo
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ziting Zheng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuxin Zhang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ping Xiao
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xincen Zhong
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenjuan Yan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
3
|
Tutty MA, Prina-Mello A. Three-Dimensional Spheroids for Cancer Research. Methods Mol Biol 2023; 2645:65-103. [PMID: 37202612 DOI: 10.1007/978-1-0716-3056-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
In vitro cell culture is one of the most widely used tools used today for increasing our understanding of various things such as protein production, mechanisms of drug action, tissue engineering, and overall cellular biology. For the past decades, however, cancer researchers have relied heavily on conventional two-dimensional (2D) monolayer culture techniques to test a variety of aspects of cancer research ranging from the cytotoxic effects of antitumor drugs to the toxicity of diagnostic dyes and contact tracers. However, many promising cancer therapies have either weak or no efficacy in real-life conditions, therefore delaying or stopping altogether their translating to the clinic. This is, in part, due to the reductionist 2D cultures used to test these materials, which lack appropriate cell-cell contacts, have altered signaling, do not represent the natural tumor microenvironment, and have different drug responses, due to their reduced malignant phenotype when compared to real in vivo tumors. With the most recent advances, cancer research has moved into 3D biological investigation. Three-dimensional (3D) cultures of cancer cells not only recapitulate the in vivo environment better than their 2D counterparts, but they have, in recent years, emerged as a relatively low-cost and scientifically accurate methodology for studying cancer. In this chapter, we highlight the importance of 3D culture, specifically 3D spheroid culture, reviewing some key methodologies for forming 3D spheroids, discussing the experimental tools that can be used in conjunction with 3D spheroids and finally their applications in cancer research.
Collapse
Affiliation(s)
- Melissa Anne Tutty
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland.
| | - Adriele Prina-Mello
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland
- Nanomedicine and Molecular Imaging Group, Trinity Translational Medicine Institute, (TTMI), School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
4
|
Lysyl Oxidase Family Proteins: Prospective Therapeutic Targets in Cancer. Int J Mol Sci 2022; 23:ijms232012270. [PMID: 36293126 PMCID: PMC9602794 DOI: 10.3390/ijms232012270] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/03/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
Abstract
The lysyl oxidase (LOX) family, consisting of LOX and LOX-like proteins 1–4 (LOXL1–4), is responsible for the covalent crosslinking of collagen and elastin, thus maintaining the stability of the extracellular matrix (ECM) and functioning in maintaining connective tissue function, embryonic development, and wound healing. Recent studies have found the aberrant expression or activity of the LOX family occurs in various types of cancer. It has been proved that the LOX family mainly performs tumor microenvironment (TME) remodeling function and is extensively involved in tumor invasion and metastasis, immunomodulation, proliferation, apoptosis, etc. With relevant translational research in progress, the LOX family is expected to be an effective target for tumor therapy. Here, we review the research progress of the LOX family in tumor progression and therapy to provide novel insights for future exploration of relevant tumor mechanism and new therapeutic targets.
Collapse
|
5
|
Zhou W, Tang X, Huang J, Wang J, Zhao J, Zhang L, Wang Z, Li P, Li R. Dual-imaging magnetic nanocatalysis based on Fenton-like reaction for tumor therapy. J Mater Chem B 2022; 10:3462-3473. [PMID: 35403639 DOI: 10.1039/d1tb02308j] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Sequential nano-catalytic therapy has emerged as a novel therapeutic modality for cancer treatment as it utilizes the unique tumor microenvironment for selective tumor treatment. This study reports a magnetic nanoparticle to achieve Fenton-like reaction and dual-imaging guidance/monitoring. Natural glucose oxidase (GOx) and superparamagnetic Fe3O4 nanoparticles have been integrated into poly(lactic-co-glycolic acid) (PLGA) to fabricate a sequential nanocatalyst (designated as GOx@PLGA-Fe3O4). This nanocatalyst can functionally deplete glucose in tumor tissues, producing a considerable amount of highly cytotoxic hydroxyl radicals via the sequential Fenton-like reaction, and meanwhile maximizing the potential imaging capability as a contrast agent for magnetic resonance imaging and photoacoustic imaging. By ribonucleic acid sequencing (RNA-seq) technology, GOx@PLGA-Fe3O4 nanoparticles are demonstrated to induce tumor cell death by inhibiting multiple gene regulation pathways involving tumor growth and recurrence. Therefore, this finding provides a novel strategy to achieve promising therapeutic efficacy by the rational design of multifunctional nanoparticles with various features, including magnetic targeting, sequential nano-catalytic therapy, and dual-imaging guidance/monitoring.
Collapse
Affiliation(s)
- Weicheng Zhou
- Department of Ultrasound, The Third Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P. R. China
| | - Xinyi Tang
- Department of Ultrasound, The Third Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P. R. China
| | - Ju Huang
- Department of Ultrasound, The Third Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P. R. China
| | - Jingxue Wang
- Department of Ultrasound, The Third Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P. R. China
| | - Jiawen Zhao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P. R. China
| | - Liang Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P. R. China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P. R. China
| | - Pan Li
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P. R. China
| | - Rui Li
- Department of Ultrasound, The Third Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P. R. China
| |
Collapse
|
6
|
Zoi V, Galani V, Tsekeris P, Kyritsis AP, Alexiou GA. Radiosensitization and Radioprotection by Curcumin in Glioblastoma and Other Cancers. Biomedicines 2022; 10:312. [PMID: 35203521 PMCID: PMC8869399 DOI: 10.3390/biomedicines10020312] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/25/2022] Open
Abstract
Radiation therapy plays an important role in almost every cancer treatment. However, radiation toxicity to normal tissues, mainly due to the generation of reactive free radicals, has limited the efficacy of radiotherapy in clinical practice. Curcumin has been reported to possess significant antitumor properties. Although curcumin can sensitize cancer cells to irradiation, healthy cells are much less sensitive to this effect, and thus, curcumin is thought to be a potent, yet safe anti-cancer agent. In this review, a summary of the role of curcumin as both a radiosensitizer and radioprotector has been presented, based on the most recent data from the experimental and clinical evaluation of curcumin in different cancer cell lines, animal models, and human patients.
Collapse
Affiliation(s)
- Vasiliki Zoi
- Neurosurgical Institute, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (A.P.K.)
| | - Vasiliki Galani
- Department of Anatomy Histology-Embryology, School of Medicine, University of Ioannina, 45110 Ioannina, Greece;
| | - Pericles Tsekeris
- Department of Radiation Oncology, University of Ioannina, 45110 Ioannina, Greece;
| | - Athanasios P. Kyritsis
- Neurosurgical Institute, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (A.P.K.)
| | - George A. Alexiou
- Neurosurgical Institute, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (A.P.K.)
| |
Collapse
|
7
|
Zhu J, Luo C, Zhao J, Zhu X, Lin K, Bu F, Yu Z, Zou F, Zhu Z. Expression of LOX Suggests Poor Prognosis in Gastric Cancer. Front Med (Lausanne) 2021; 8:718986. [PMID: 34595188 PMCID: PMC8476844 DOI: 10.3389/fmed.2021.718986] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/24/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Lysyl oxidase (LOX) is a key enzyme for the cross-linking of collagen and elastin in the extracellular matrix. This study evaluated the prognostic role of LOX in gastric cancer (GC) by analyzing the data of The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) dataset. Methods: The Wilcoxon rank-sum test was used to calculate the expression difference of LOX gene in gastric cancer and normal tissues. Western blot and immunohistochemical staining were used to evaluate the expression level of LOX protein in gastric cancer. Kaplan-Meier analysis was used to calculate the survival difference between the high expression group and the low expression group in gastric cancer. The relationship between statistical clinicopathological characteristics and LOX gene expression was analyzed by Wilcoxon or Kruskal-Wallis test and logistic regression. Univariate and multivariate Cox regression analysis was used to find independent risk factors affecting the prognosis of GC patients. Gene set enrichment analysis (GSEA) was used to screen the possible mechanisms of LOX and GC. The CIBERSORT calculation method was used to evaluate the distribution of tumor-infiltrating immune cell (TIC) abundance. Results: LOX is highly expressed in gastric cancer tissues and is significantly related to poor overall survival. Wilcoxon or Kruskal-Wallis test and Logistic regression analysis showed, LOX overexpression is significantly correlated with T-stage progression in gastric cancer. Multivariate Cox regression analysis on TCGA and GEO data found that LOX (all p < 0.05) is an independent factor for poor GC prognosis. GSEA showed that high LOX expression is related to ECM receptor interaction, cancer, Hedgehog, TGF-beta, JAK-STAT, MAPK, Wnt, and mTOR signaling pathways. The expression level of LOX affects the immune activity of the tumor microenvironment in gastric cancer. Conclusion: High expression of LOX is a potential molecular indicator for poor prognosis of gastric cancer.
Collapse
Affiliation(s)
- Jinfeng Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chen Luo
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiefeng Zhao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaojian Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kang Lin
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fanqin Bu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhonglin Yu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Feilong Zou
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhengming Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Yi X, Shen M, Liu X, Gu J. Emerging strategies based on nanomaterials for ionizing radiation-optimized drug treatment of cancer. NANOSCALE 2021; 13:13943-13961. [PMID: 34477676 DOI: 10.1039/d1nr03034e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Drug-radiotherapy is a common and effective combinational treatment for cancer. This study aimed to explore the ionizing radiation-optimized drug treatment based on nanomaterials so as to improve the synergistic efficacy of drug-radiotherapy against cancer and limit the adverse effect on healthy organs. In this review, these emerging strategies were divided into four parts. First, the delivery of the drug-loaded nanoparticles was optimized owing to the strengthened passive targeting process, active targeting process, and cell targeting process of nanoparticles after ionizing radiation exposure. Second, nanomaterials were designed to respond to the ionizing radiation, thus leading to the release of the loading drugs controllably. Third, radiation-activated pro-drugs were loaded onto nanoparticles for radiation-triggered drug therapy. In particular, nontoxic nanoparticles with radiosensitization capability and innocuous radio-dynamic contrast agents can be considered as radiation-activated drugs, which were discussed in this review. Fourth, according to the various synergetic mechanisms, radiotherapy could improve the drug response of cancer, obtaining optimized drug-radiotherapy. Finally, relative suggestions were provided to further optimize these aforementioned strategies. Therefore, a novel topic was selected and the emerging strategies in this region were discussed, aiming to stimulate the inspiration for the development of ionizing radiation-optimized drug treatment based on nanomaterials.
Collapse
Affiliation(s)
- Xuan Yi
- School of Pharmacy, Jiangsu Key Laboratory of Inflammation and Molecular Drug Targets, Nantong University, Nantong, Jiangsu 226001, China.
| | | | | | | |
Collapse
|
9
|
3D Modeling of Epithelial Tumors-The Synergy between Materials Engineering, 3D Bioprinting, High-Content Imaging, and Nanotechnology. Int J Mol Sci 2021; 22:ijms22126225. [PMID: 34207601 PMCID: PMC8230141 DOI: 10.3390/ijms22126225] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022] Open
Abstract
The current statistics on cancer show that 90% of all human cancers originate from epithelial cells. Breast and prostate cancer are examples of common tumors of epithelial origin that would benefit from improved drug treatment strategies. About 90% of preclinically approved drugs fail in clinical trials, partially due to the use of too simplified in vitro models and a lack of mimicking the tumor microenvironment in drug efficacy testing. This review focuses on the origin and mechanism of epithelial cancers, followed by experimental models designed to recapitulate the epithelial cancer structure and microenvironment, such as 2D and 3D cell culture models and animal models. A specific focus is put on novel technologies for cell culture of spheroids, organoids, and 3D-printed tissue-like models utilizing biomaterials of natural or synthetic origins. Further emphasis is laid on high-content imaging technologies that are used in the field to visualize in vitro models and their morphology. The associated technological advancements and challenges are also discussed. Finally, the review gives an insight into the potential of exploiting nanotechnological approaches in epithelial cancer research both as tools in tumor modeling and how they can be utilized for the development of nanotherapeutics.
Collapse
|
10
|
Zhao Y, Zheng X, Zheng Y, Chen Y, Fei W, Wang F, Zheng C. Extracellular Matrix: Emerging Roles and Potential Therapeutic Targets for Breast Cancer. Front Oncol 2021; 11:650453. [PMID: 33968752 PMCID: PMC8100244 DOI: 10.3389/fonc.2021.650453] [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: 01/08/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence shows that the extracellular matrix (ECM) is an important regulator of breast cancer (BC). The ECM comprises of highly variable and dynamic components. Compared with normal breast tissue under homeostasis, the ECM undergoes many changes in composition and organization during BC progression. Induced ECM proteins, including fibrinogen, fibronectin, hyaluronic acid, and matricellular proteins, have been identified as important components of BC metastatic cells in recent years. These proteins play major roles in BC progression, invasion, and metastasis. Importantly, several specific ECM molecules, receptors, and remodeling enzymes are involved in promoting resistance to therapeutic intervention. Additional analysis of these ECM proteins and their downstream signaling pathways may reveal promising therapeutic targets against BC. These potential drug targets may be combined with new nanoparticle technologies. This review summarizes recent advances in functional nanoparticles that target the ECM to treat BC. Accurate nanomaterials may offer a new approach to BC treatment.
Collapse
Affiliation(s)
- Yunchun Zhao
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Lab Women's Reproductive Health, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoling Zheng
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Lab Women's Reproductive Health, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongquan Zheng
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Lab Women's Reproductive Health, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yue Chen
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Lab Women's Reproductive Health, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Lab Women's Reproductive Health, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fengmei Wang
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Lab Women's Reproductive Health, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Lab Women's Reproductive Health, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Mó I, Sabino IJ, Melo-Diogo DD, Lima-Sousa R, Alves CG, Correia IJ. The importance of spheroids in analyzing nanomedicine efficacy. Nanomedicine (Lond) 2020; 15:1513-1525. [PMID: 32552537 DOI: 10.2217/nnm-2020-0054] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The use of nanomedicines for cancer treatment holds a great potential due to their improved efficacy and safety. During the nanomedicine preclinical in vitro evaluation stage, these are mainly tested on cell culture monolayers. However, these 2D models are an unrealistic representation of the in vivo tumors, leading to an inaccurate screening of the candidate formulations. To address this problem, spheroids are emerging as an additional tool to validate the efficacy of new therapeutics due to the ability of these 3D in vitro cancer models to mimic the key features displayed by in vivo solid tumors. In this review, the application of spheroids for the evaluation of nanomedicines' physicochemical properties and therapeutic efficacy is discussed.
Collapse
Affiliation(s)
- Inês Mó
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506, Covilhã, Portugal
| | - Ivo J Sabino
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506, Covilhã, Portugal
| | - Duarte de Melo-Diogo
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506, Covilhã, Portugal
| | - Rita Lima-Sousa
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506, Covilhã, Portugal
| | - Cátia G Alves
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506, Covilhã, Portugal
| | - Ilídio J Correia
- CICS-UBI - Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506, Covilhã, Portugal.,CIEPQPF - Departamento de Engenharia Química, Universidade de Coimbra, 3030-790, Coimbra, Portugal
| |
Collapse
|