1
|
Alsharabasy AM, Pandit A. Hyaluronan-Based Hydrogels for 3D Modeling of Tumor Tissues. Tissue Eng Part C Methods 2024; 30:452-499. [PMID: 39345138 DOI: 10.1089/ten.tec.2024.0271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024] Open
Abstract
Although routine two-dimensional (2D) cell culture techniques have advanced basic cancer research owing to their simplicity, cost-effectiveness, and reproducibility, they have limitations that necessitate the development of advanced three-dimensional (3D) tumor models that better recapitulate the tumor microenvironment. Various biomaterials have been used to establish these 3D models, enabling the study of cancer cell behavior within different matrices. Hyaluronic acid (HA), a key component of the extracellular matrix (ECM) in tumor tissues, has been widely studied and employed in the development of multiple cancer models. This review first examines the role of HA in tumors, including its function as an ECM component and regulator of signaling pathways that affect tumor progression. It then explores HA-based models for various cancers, focusing on HA as a central component of the 3D matrix and its mobilization within the matrix for targeted studies of cell behavior and drug testing. The tumor models discussed included those for breast cancer, glioblastoma, fibrosarcoma, gastric cancer, hepatocellular carcinoma, and melanoma. The review concludes with a discussion of future prospects for developing more robust and high-throughput HA-based models to more accurately mimic the tumor microenvironment and improve drug testing. Impact Statement This review underscores the transformative potential of hyaluronic acid (HA)-based hydrogels in developing advanced tumor models. By exploring HA's dual role as a critical extracellular matrix component and a regulator of cancer cell dynamics, we highlight its unique contributions to replicating the tumor microenvironment. The recent advancements in HA-based models provide new opportunities for more accurate studies of cancer cell behavior and drug responses. Looking ahead, these innovations pave the way for high-throughput, biomimetic platforms that could revolutionize drug testing and accelerate the discovery of effective cancer therapies.
Collapse
Affiliation(s)
- Amir M Alsharabasy
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| |
Collapse
|
2
|
Gholamali I, Vu TT, Jo SH, Park SH, Lim KT. Exploring the Progress of Hyaluronic Acid Hydrogels: Synthesis, Characteristics, and Wide-Ranging Applications. MATERIALS (BASEL, SWITZERLAND) 2024; 17:2439. [PMID: 38793505 PMCID: PMC11123044 DOI: 10.3390/ma17102439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/30/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
This comprehensive review delves into the world of hyaluronic acid (HA) hydrogels, exploring their creation, characteristics, research methodologies, and uses. HA hydrogels stand out among natural polysaccharides due to their distinct features. Their exceptional biocompatibility makes them a top choice for diverse biomedical purposes, with a great ability to coexist harmoniously with living cells and tissues. Furthermore, their biodegradability permits their gradual breakdown by bodily enzymes, enabling the creation of temporary frameworks for tissue engineering endeavors. Additionally, since HA is a vital component of the extracellular matrix (ECM) in numerous tissues, HA hydrogels can replicate the ECM's structure and functions. This mimicry is pivotal in tissue engineering applications by providing an ideal setting for cellular growth and maturation. Various cross-linking techniques like chemical, physical, enzymatic, and hybrid methods impact the mechanical strength, swelling capacity, and degradation speed of the hydrogels. Assessment tools such as rheological analysis, electron microscopy, spectroscopy, swelling tests, and degradation studies are employed to examine their attributes. HA-based hydrogels feature prominently in tissue engineering, drug distribution, wound recovery, ophthalmology, and cartilage mending. Crafting HA hydrogels enables the production of biomaterials with sought-after qualities, offering avenues for advancements in the realm of biomedicine.
Collapse
Affiliation(s)
- Iman Gholamali
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Republic of Korea; (I.G.); (S.-H.J.)
| | - Trung Thang Vu
- Department of Smart Green Technology Engineering, Pukyong National University, Busan 48513, Republic of Korea;
| | - Sung-Han Jo
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Republic of Korea; (I.G.); (S.-H.J.)
| | - Sang-Hyug Park
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Republic of Korea; (I.G.); (S.-H.J.)
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence, Pukyong National University, Busan 48513, Republic of Korea
| | - Kwon Taek Lim
- Institute of Display Semiconductor Technology, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
3
|
Zhao Y, Wu Y, Islam K, Paul R, Zhou Y, Qin X, Li Q, Liu Y. Microphysiologically Engineered Vessel-Tumor Model to Investigate Vascular Transport Dynamics of Immune Cells. ACS APPLIED MATERIALS & INTERFACES 2024; 16. [PMID: 38652824 PMCID: PMC11082852 DOI: 10.1021/acsami.4c00391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Cancer immunotherapy has emerged as a promising therapeutic strategy to combat cancer effectively. However, it is hard to observe and quantify how this in vivo process happens. Three-dimensional (3D) microfluidic vessel-tumor models offer valuable capability to study how immune cells transport during cancer progression. We presented an advanced 3D vessel-supported tumor model consisting of the endothelial lumen and vessel network for the study of T cells' transportation. The process of T cell transport through the vessel network and interaction with tumor spheroids was represented and monitored in vitro. Specifically, we demonstrate that the endothelial glycocalyx serving in the T cells' transport can influence the endothelium-immune interaction. Furthermore, after vascular transport, how programmed cell death protein 1 (PD-1) immune checkpoint inhibition influences the delivered activated-T cells on tumor killing was evaluated. Our in vitro vessel-tumor model provides a microphysiologically engineered platform to represent T cell vascular transportation during tumor immunotherapy. The reported innovative vessel-tumor platform is believed to have the potential to explore the tumor-induced immune response mechanism and preclinically evaluate immunotherapy's effectiveness.
Collapse
Affiliation(s)
- Yuwen Zhao
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Yue Wu
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Khayrul Islam
- Department
of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Ratul Paul
- Department
of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Yuyuan Zhou
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Xiaochen Qin
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Qiying Li
- Department
of Electrical and Computer Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Yaling Liu
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
- Department
of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
4
|
He X, Hu S, Wang C, Yang Y, Li Z, Zeng M, Song G, Li Y, Lu Q. Predicting prostate cancer recurrence: Introducing PCRPS, an advanced online web server. Heliyon 2024; 10:e28878. [PMID: 38623253 PMCID: PMC11016622 DOI: 10.1016/j.heliyon.2024.e28878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/17/2024] Open
Abstract
Background Prostate cancer (PCa) is one of the leading causes of cancer death in men. About 30% of PCa will develop a biochemical recurrence (BCR) following initial treatment, which significantly contributes to prostate cancer-related deaths. In clinical practice, accurate prediction of PCa recurrence is crucial for making informed treatment decisions. However, the development of reliable models and biomarkers for predicting PCa recurrence remains a challenge. In this study, the aim is to establish an effective and reliable tool for predicting the recurrence of PCa. Methods We systematically screened and analyzed potential datasets to predict PCa recurrence. Through quality control analysis, low-quality datasets were removed. Using meta-analysis, differential expression analysis, and feature selection, we identified key genes associated with recurrence. We also evaluated 22 previously published signatures for PCa recurrence prediction. To assess prediction performance, we employed nine machine learning algorithms. We compared the predictive capabilities of models constructed using clinical variables, expression data, and their combinations. Subsequently, we implemented these machine learning models into a user-friendly web server freely accessible to all researchers. Results Based on transcriptomic data derived from eight multicenter studies consisting of 733 PCa patients, we screened 23 highly influential genes for predicting prostate cancer recurrence. These genes were used to construct the Prostate Cancer Recurrence Prediction Signature (PCRPS). By comparing with 22 published signatures and four important clinicopathological features, the PCRPS exhibited a robust and significantly improved predictive capability. Among the tested algorithms, Random Forest demonstrated the highest AUC value of 0.72 in predicting PCa recurrence in the testing dataset. To facilitate access and usage of these machine learning models by all researchers and clinicians, we also developed an online web server (https://urology1926.shinyapps.io/PCRPS/) where the PCRPS model can be freely utilized. The tool can also be used to (1) predict the PCa recurrence by clinical information or expression data with high accuracy. (2) provide the possibility of PCa recurrence by nine machine learning algorithms. Furthermore, using the PCRPS scores, we predicted the sensitivity of 22 drugs from GDSC2 and 95 drugs from CTRP2 to the samples. These predictions provide valuable insights into potential drug sensitivities related to the PCRPS score groups. Conclusion Overall, our study provides an attractive tool to further guide the clinical management and individualized treatment for PCa.
Collapse
Affiliation(s)
| | | | - Chen Wang
- Department of Urology, Hunan Provincial People's Hospital (The 1st Affiliated Hospital of Hunan Normal University), China
| | - Yongjun Yang
- Department of Urology, Hunan Provincial People's Hospital (The 1st Affiliated Hospital of Hunan Normal University), China
| | - Zhuo Li
- Department of Urology, Hunan Provincial People's Hospital (The 1st Affiliated Hospital of Hunan Normal University), China
| | - Mingqiang Zeng
- Department of Urology, Hunan Provincial People's Hospital (The 1st Affiliated Hospital of Hunan Normal University), China
| | - Guangqing Song
- Department of Urology, Hunan Provincial People's Hospital (The 1st Affiliated Hospital of Hunan Normal University), China
| | - Yuanwei Li
- Department of Urology, Hunan Provincial People's Hospital (The 1st Affiliated Hospital of Hunan Normal University), China
| | - Qiang Lu
- Department of Urology, Hunan Provincial People's Hospital (The 1st Affiliated Hospital of Hunan Normal University), China
| |
Collapse
|
5
|
De Vitis E, Stanzione A, Romano A, Quattrini A, Gigli G, Moroni L, Gervaso F, Polini A. The Evolution of Technology-Driven In Vitro Models for Neurodegenerative Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304989. [PMID: 38366798 DOI: 10.1002/advs.202304989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/15/2024] [Indexed: 02/18/2024]
Abstract
The alteration in the neural circuits of both central and peripheral nervous systems is closely related to the onset of neurodegenerative disorders (NDDs). Despite significant research efforts, the knowledge regarding NDD pathological processes, and the development of efficacious drugs are still limited due to the inability to access and reproduce the components of the nervous system and its intricate microenvironment. 2D culture systems are too simplistic to accurately represent the more complex and dynamic situation of cells in vivo and have therefore been surpassed by 3D systems. However, both models suffer from various limitations that can be overcome by employing two innovative technologies: organ-on-chip and 3D printing. In this review, an overview of the advantages and shortcomings of both microfluidic platforms and extracellular matrix-like biomaterials will be given. Then, the combination of microfluidics and hydrogels as a new synergistic approach to study neural disorders by analyzing the latest advances in 3D brain-on-chip for neurodegenerative research will be explored.
Collapse
Affiliation(s)
- Eleonora De Vitis
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Antonella Stanzione
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Romano
- IRCCS San Raffaele Scientific Institute, Division of Neuroscience, Institute of Experimental Neurology, Milan, 20132, Italy
| | - Angelo Quattrini
- IRCCS San Raffaele Scientific Institute, Division of Neuroscience, Institute of Experimental Neurology, Milan, 20132, Italy
| | - Giuseppe Gigli
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
- Dipartimento di Medicina Sperimentale, Università Del Salento, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Lorenzo Moroni
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
- Complex Tissue Regeneration, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, Netherlands
| | - Francesca Gervaso
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Polini
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| |
Collapse
|
6
|
Karimifard SA, Salehzadeh-Yazdi A, Taghizadeh-Tabarsi R, Akbari-Birgani S. Mechanical effects modulate drug resistance in MCF-7-derived organoids: Insights into the wnt/β-catenin pathway. Biochem Biophys Res Commun 2024; 695:149420. [PMID: 38154263 DOI: 10.1016/j.bbrc.2023.149420] [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: 10/14/2023] [Revised: 11/29/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023]
Abstract
Addressing drug resistance poses a significant challenge in cancer treatment, as cancer cells develop diverse mechanisms to evade chemotherapy drugs, leading to treatment failure and disease relapse. Three-dimensional (3D) cell culture has emerged as a valuable model for studying drug resistance, although the underlying mechanisms remain elusive. By obtaining a better understanding of drug resistance within the 3D culture environment, we can develop more effective strategies to overcome it and improve the success of cancer treatments. Notably, the physical structure undergoes notable changes in 3D culture, with mechanical effects believed to play a pivotal role in drug resistance. Hence, our study aimed to explore the influence of mechanical effects on drug resistance by analyzing data related to "drug resistance" and "mechanobiology". Through this analysis, we identified β-catenin and JNK1 as potential factors, which were further examined in MCF-7 cells cultivated under both 2D and 3D culture conditions. Our findings demonstrate that β-catenin is activated through canonical and non-canonical pathways and associated with the drug resistance, particularly in organoids obtained under 3D culture.
Collapse
Affiliation(s)
- Seyed Ali Karimifard
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 45137-66731, Iran
| | | | - Reza Taghizadeh-Tabarsi
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 45137-66731, Iran
| | - Shiva Akbari-Birgani
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 45137-66731, Iran; Research Center for Basic Sciences and Modern Technologies (RBST), Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 45137-66731, Iran.
| |
Collapse
|
7
|
Hassani I, Anbiah B, Moore AL, Abraham PT, Odeniyi IA, Habbit NL, Greene MW, Lipke EA. Establishment of a tissue-engineered colon cancer model for comparative analysis of cancer cell lines. J Biomed Mater Res A 2024; 112:231-249. [PMID: 37927200 DOI: 10.1002/jbm.a.37611] [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: 11/10/2022] [Revised: 08/13/2023] [Accepted: 08/30/2023] [Indexed: 11/07/2023]
Abstract
To overcome the limitations of in vitro two-dimensional (2D) cancer models in mimicking the complexities of the native tumor milieu, three-dimensional (3D) engineered cancer models using biomimetic materials have been introduced to more closely recapitulate the key attributes of the tumor microenvironment. Specifically, for colorectal cancer (CRC), a few studies have developed 3D engineered tumor models to investigate cell-cell interactions or efficacy of anti-cancer drugs. However, recapitulation of CRC cell line phenotypic differences within a 3D engineered matrix has not been systematically investigated. Here, we developed an in vitro 3D engineered CRC (3D-eCRC) tissue model using the natural-synthetic hybrid biomaterial PEG-fibrinogen and three CRC cell lines, HCT 116, HT-29, and SW480. To better recapitulate native tumor conditions, our 3D-eCRC model supported higher cell density encapsulation (20 × 106 cells/mL) and enabled longer term maintenance (29 days) as compared to previously reported in vitro CRC models. The 3D-eCRCs formed using each cell line demonstrated line-dependent differences in cellular and tissue properties, including cellular growth and morphology, cell subpopulations, cell size, cell granularity, migration patterns, tissue growth, gene expression, and tissue stiffness. Importantly, these differences were found to be most prominent from Day 22 to Day 29, thereby indicating the importance of long-term culture of engineered CRC tissues for recapitulation and investigation of mechanistic differences and drug response. Our 3D-eCRC tissue model showed high potential for supporting future in vitro comparative studies of disease progression, metastatic mechanisms, and anti-cancer drug candidate response in a CRC cell line-dependent manner.
Collapse
Affiliation(s)
- Iman Hassani
- Department of Chemical Engineering, Auburn University, Auburn, Alabama, USA
- Department of Chemical Engineering, Tuskegee University, Tuskegee, Alabama, USA
| | - Benjamin Anbiah
- Department of Chemical Engineering, Auburn University, Auburn, Alabama, USA
| | - Andrew L Moore
- Department of Chemical Engineering, Auburn University, Auburn, Alabama, USA
| | - Peter T Abraham
- Department of Chemical Engineering, Auburn University, Auburn, Alabama, USA
| | - Ifeoluwa A Odeniyi
- Department of Nutritional Sciences, Auburn University, Auburn, Alabama, USA
| | - Nicole L Habbit
- Department of Chemical Engineering, Auburn University, Auburn, Alabama, USA
| | - Michael W Greene
- Department of Nutritional Sciences, Auburn University, Auburn, Alabama, USA
| | - Elizabeth A Lipke
- Department of Chemical Engineering, Auburn University, Auburn, Alabama, USA
| |
Collapse
|
8
|
Lee SY, Koo IS, Hwang HJ, Lee DW. WITHDRAWN: In Vitro three-dimensional (3D) cell culture tools for spheroid and organoid models. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023:100131. [PMID: 38101575 DOI: 10.1016/j.slasd.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 12/17/2023]
Abstract
The Publisher regrets that this article is an accidental duplication of an article previously published at http://dx.doi.org/10.1016/j.slasd.2023.03.006. This duplication was due to an error in the publishing workflow and was not the responsibility of the authors or editors. As a result, the duplicate article has been withdrawn. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Sang-Yun Lee
- Department of Biomedical Engineering, Gachon University, Seongnam, 13120, Republic of Korea; Central R & D Center, Medical & Bio Decision (MBD) Co., Ltd, Suwon, 16229, Republic of Korea
| | - In-Seong Koo
- Department of Biomedical Engineering, Gachon University, Seongnam, 13120, Republic of Korea
| | - Hyun Ju Hwang
- Central R & D Center, Medical & Bio Decision (MBD) Co., Ltd, Suwon, 16229, Republic of Korea
| | - Dong Woo Lee
- Department of Biomedical Engineering, Gachon University, Seongnam, 13120, Republic of Korea.
| |
Collapse
|
9
|
Thilakan AT, Nandakumar N, Balakrishnan AR, Pooleri GK, Nair SV, Sathy BN. Development and characterisation of suitably bioengineered microfibrillar matrix-based 3D prostate cancer model for in vitrodrug testing. Biomed Mater 2023; 18:065016. [PMID: 37738986 DOI: 10.1088/1748-605x/acfc8e] [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: 06/11/2023] [Accepted: 09/22/2023] [Indexed: 09/24/2023]
Abstract
Bioengineered 3D models that can mimic patient-specific pathologiesin vitroare valuable tools for developing and validating anticancer therapeutics. In this study, microfibrillar matrices with unique structural and functional properties were fabricated as 3D spherical and disc-shaped scaffolds with highly interconnected pores and the potential of the newly developed scaffolds for developing prostate cancer model has been investigated. The newly developed scaffolds showed improved cell retention upon seeding with cancer cells compared to conventional electrospun scaffolds. They facilitated rapid growth and deposition of cancer-specific extracellular matrix through-the-thickness of the scaffold. Compared to the prostate cancer cells grown in 2D culture, the newly developed prostate cancer model showed increased resistance to the chemodrug Docetaxel regardless of the drug concentration or the treatment frequency. A significant reduction in the cell number was observed within one week after the drug treatment in the 2D culture for both PC3 and patient-derived cells. Interestingly, almost 20%-30% of the cancer cells in the newly developed 3D model survived the drug treatment, and the patient-derived cells were more resistant than the tested cell line PC3. The results from this study indicate the potential of the newly developed prostate cancer model forin vitrodrug testing.
Collapse
Affiliation(s)
- Akhil T Thilakan
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Niji Nandakumar
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Arvind R Balakrishnan
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Ginil K Pooleri
- Department of Urology and Renal Transplantation, Amrita Institute of Medical Sciences and Research, Kochi, Kerala, India
| | - Shantikumar V Nair
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Binulal N Sathy
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| |
Collapse
|
10
|
Pol M, Gao H, Zhang H, George OJ, Fox JM, Jia X. Dynamic modulation of matrix adhesiveness induces epithelial-to-mesenchymal transition in prostate cancer cells in 3D. Biomaterials 2023; 299:122180. [PMID: 37267701 PMCID: PMC10330660 DOI: 10.1016/j.biomaterials.2023.122180] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 04/27/2023] [Accepted: 05/25/2023] [Indexed: 06/04/2023]
Abstract
Synthetic matrices with dynamic presentation of cell guidance cues are needed for the development of physiologically relevant in vitro tumor models. Towards the goal of mimicking prostate cancer progression and metastasis, we engineered a tunable hyaluronic acid-based hydrogel platform with protease degradable and cell adhesive properties employing bioorthogonal tetrazine ligation with strained alkenes. The synthetic matrix was first fabricated via a slow tetrazine-norbornene reaction, then temporally modified via a diffusion-controlled method using trans-cyclooctene, a fierce dienophile that reacts with tetrazine with an unusually fast rate. The encapsulated DU145 prostate cancer single cells spontaneously formed multicellular tumoroids after 7 days of culture. In situ modification of the synthetic matrix via covalent tagging of cell adhesive RGD peptide induced tumoroid decompaction and the development of cellular protrusions. RGD tagging did not compromise the overall cell viability, nor did it induce cell apoptosis. In response to increased matrix adhesiveness, DU145 cells dynamically loosen cell-cell adhesion and strengthen cell-matrix interactions to promote an invasive phenotype. Characterization of the 3D cultures by immunocytochemistry and gene expression analyses demonstrated that cells invaded into the matrix via a mesenchymal like migration, with upregulation of major mesenchymal markers, and down regulation of epithelial markers. The tumoroids formed cortactin positive invadopodia like structures, indicating active matrix remodeling. Overall, the engineered tumor model can be utilized to identify potential molecular targets and test pharmacological inhibitors, thereby accelerating the design of innovative strategies for cancer therapeutics.
Collapse
Affiliation(s)
- Mugdha Pol
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Hanyuan Gao
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, USA
| | - He Zhang
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, USA
| | - Olivia J George
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, USA
| | - Joseph M Fox
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, USA; Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| | - Xinqiao Jia
- Department of Biological Sciences, University of Delaware, Newark, DE, USA; Department of Materials Science and Engineering, University of Delaware, Newark, DE, USA; Department of Biomedical Engineering, University of Delaware, Newark, DE, USA; Delaware Biotechnology Institute, University of Delaware, Newark, DE, USA.
| |
Collapse
|
11
|
Park DH, He MT, Cho EJ, Morten K, Go JS. Development of a novel microfluidic perfusion 3D cell culture system for improved neuronal cell differentiation. Biomed Microdevices 2023; 25:22. [PMID: 37310518 DOI: 10.1007/s10544-023-00660-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2023] [Indexed: 06/14/2023]
Abstract
Three-dimensional (3D) cell cultures have recently gained popularity in the biomedical sciences because of their similarity to the in vivo environment. SH-SY5Y cells, which are neuronal cells and are commonly used to investigate neurodegenerative diseases, have particularly been reported to be differentiated as neuron-like cells expressing neuron-specific markers of mature neurons in static 3D culture environments when compared to static 2D environments, and those in perfusion environments have not yet been investigated. Microfluidic technology has provided perfusion environment which has more similarity to in vivo through mimicking vascular transportation of nutrients, but air bubbles entering into microchannels drastically increase instability of the flow. Furthermore, static incubation commonly used is incompatible with perfusion setup due to its air conditions, which is a critical huddle to the biologists. In the present study, we developed a novel microfluidic perfusion 3D cell culture system that overcomes the disturbance from air bubbles and intuitionally sets the incubation with the perfusion 3D culture. The system is capable of generating concentration gradients between 5 and 95% and air bubble traps were included to increase stability during incubation by collecting air bubbles. To evaluate the perfusion 3D culture, SH-SY5Y differentiation was examined in static 2D, static 3D, and perfusion 3D cultures. Our system supported significantly increased clustering of SH-SY5Y compared to static 2D and 3D methods, as well as increasing neurite growth rate. This novel system therefore supports differentiation of SH-SY5Y and can be used to more accurately model the in vivo environment during cell culture experiments.
Collapse
Affiliation(s)
- Dong Hyeok Park
- School of Mechanical Engineering, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan, 46241, Republic of Korea
| | - Mei Tong He
- Department of Food Science and Nutrition & Kimchi Research Institute, Pusan National University, Busan, Republic of Korea
| | - Eun Ju Cho
- Department of Food Science and Nutrition & Kimchi Research Institute, Pusan National University, Busan, Republic of Korea
| | - Karl Morten
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Jeung Sang Go
- School of Mechanical Engineering, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan, 46241, Republic of Korea.
| |
Collapse
|
12
|
Goel R, Gulwani D, Upadhyay P, Sarangthem V, Singh TD. Unsung versatility of elastin-like polypeptide inspired spheroid fabrication: A review. Int J Biol Macromol 2023; 234:123664. [PMID: 36791934 DOI: 10.1016/j.ijbiomac.2023.123664] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/23/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023]
Abstract
Lately, 3D cell culture technique has gained a lot of appreciation as a research model. Augmented with technological advancements, the area of 3D cell culture is growing rapidly with a diverse array of scaffolds being tested. This is especially the case for spheroid cultures. The culture of cells as spheroids provides opportunities for unanticipated vision into biological phenomena with its application to drug discovery, metabolic profiling, stem cell research as well as tumor, and disease biology. Spheroid fabrication techniques are broadly categorised into matrix-dependent and matrix-independent techniques. While there is a profusion of spheroid fabrication substrates with substantial biological relevance, an economical, modular, and bio-compatible substrate for high throughput production of spheroids is lacking. In this review, we posit the prospects of elastin-like polypeptides (ELPs) as a broad-spectrum spheroid fabrication platform. Elastin-like polypeptides are nature inspired, size-tunable genetically engineered polymers with wide applicability in various arena of biological considerations, has been employed for spheroid culture with profound utility. The technology offers a cheap, high-throughput, reproducible alternative for spheroid culture with exquisite adaptability. Here, we will brief the applicability of 3D cultures as compared to 2D cultures with spheroids being the focal point of the review. Common approaches to spheroid fabrication are discussed with existential limitations. Finally, the versatility of elastin-like polypeptide inspired substrates for spheroid culture has been discussed.
Collapse
Affiliation(s)
- Ridhima Goel
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Deepak Gulwani
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Priyanka Upadhyay
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Vijaya Sarangthem
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Thoudam Debraj Singh
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
13
|
Luo Y, Tan J, Zhou Y, Guo Y, Liao X, He L, Li D, Li X, Liu Y. From crosslinking strategies to biomedical applications of hyaluronic acid-based hydrogels: A review. Int J Biol Macromol 2023; 231:123308. [PMID: 36669634 DOI: 10.1016/j.ijbiomac.2023.123308] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
Hyaluronic acid (HA) is not only a natural anionic polysaccharide with excellent biocompatibility, biodegradability, and moisturizing effect, but also an essential factor that can affect angiogenesis, inflammation, cell behavior, which has a wide range of applications in the biomedical field. Among them, HA-based hydrogels formed by various physical or chemical crosslinking strategies are particularly striking. They not only retain the physiological function of HA, but also have the skeleton function of hydrogel, which further expands the application of HA. However, HA-based natural hydrogels generally have problems such as insufficient mechanical strength and susceptibility to degradation by hyaluronidase, which limits their application to a certain extent. To solve such problems, researchers have prepared a variety of HA-based multifunctional hydrogels with remarkable properties in recent years by adopting various structural modification methods or novel crosslinking strategies, as well as introducing functionally reactive molecules or moieties, which have extended the application scope. This manuscript systematically introduced common crosslinking strategies of HA-based hydrogels and highlighted the development of novel HA-based hydrogels in anticancer drug delivery, cartilage repair, three-dimensional cell culture, skin dressing and other fields. We hope to provide some references for the subsequent development of HA-based hydrogels in the biomedical field.
Collapse
Affiliation(s)
- Yuning Luo
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Junyan Tan
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yue Zhou
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yuqiong Guo
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Xinying Liao
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Li He
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Dingxilei Li
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Xinxin Li
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yang Liu
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmacology, Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
14
|
Lee SY, Koo IS, Hwang HJ, Lee DW. In Vitro Three-dimensional (3D) Cell Culture Tools for Spheroid and Organoid Models. SLAS DISCOVERY 2023:S2472-5552(23)00028-X. [PMID: 36997090 DOI: 10.1016/j.slasd.2023.03.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023]
Abstract
Three-dimensional (3D) cell culture technology has been steadily studied since the 1990's due to its superior biocompatibility compared to the conventional two-dimensional (2D) cell culture technology, and has recently developed into an organoid culture technology that further improved biocompatibility. Since the 3D culture of human cell lines in artificial scaffolds was demonstrated in the early 90's, 3D cell culture technology has been actively developed owing to various needs in the areas of disease research, precision medicine, new drug development, and some of these technologies have been commercialized. In particular, 3D cell culture technology is actively being applied and utilized in drug development and cancer-related precision medicine research. Drug development is a long and expensive process that involves multiple steps-from target identification to lead discovery and optimization, preclinical studies, and clinical trials for approval for clinical use. Cancer ranks first among life-threatening diseases owing to intra-tumoral heterogeneity associated with metastasis, recurrence, and treatment resistance, ultimately contributing to treatment failure and adverse prognoses. Therefore, there is an urgent need for the development of efficient drugs using 3D cell culture techniques that can closely mimic in vivo cellular environments and customized tumor models that faithfully represent the tumor heterogeneity of individual patients. This review discusses 3D cell culture technology focusing on research trends, commercialization status, and expected effects developed until recently. We aim to summarize the great potential of 3D cell culture technology and contribute to expanding the base of this technology.
Collapse
|
15
|
Tian T, Liu J, Zhu H. Organ Chips and Visualization of Biological Systems. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1199:155-183. [PMID: 37460731 DOI: 10.1007/978-981-32-9902-3_8] [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: 07/20/2023]
Abstract
Organ-on-a-chip (OOC) is an emerging frontier cross-cutting science and technology developed in the past 10 years. It was first proposed by the Wyss Institute for Biologically Inspired Engineering of Harvard Medical School. It consists of a transparent flexible polymer the size of a computer memory stick, with hollow microfluidic channels lined with living human cells. Researchers used bionics methods to simulate the microenvironment of human cells on microfluidic chips, so as to realize the basic physiological functions of corresponding tissues and organs in vitro. Transparent chip materials can perform real-time visualization and high-resolution analysis of various human life processes in a way that is impossible in animal models, so as to better reproduce the microenvironment of human tissue and simulate biological systems in vitro to observe drug metabolism and other life processes. It provides innovative research systems and system solutions for in vitro bionics of biological systems. It also has gradually become a new tool for disease mechanism research and new drug development. In this chapter, we will take the current research mature single-organ-on-a-chip and multi-organ human-on-a-chip as examples; give an overview of the research background and underlying technologies in this field, especially the application of in vitro bionic models in visualized medicine; and look forward to the foreseeable future development prospects after the integration of organ-on-chip and organoid technology.
Collapse
Affiliation(s)
- Tian Tian
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.
| | - Jun Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - He Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
16
|
Anthon SG, Valente KP. Vascularization Strategies in 3D Cell Culture Models: From Scaffold-Free Models to 3D Bioprinting. Int J Mol Sci 2022; 23:14582. [PMID: 36498908 PMCID: PMC9737506 DOI: 10.3390/ijms232314582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
The discrepancies between the findings in preclinical studies, and in vivo testing and clinical trials have resulted in the gradual decline in drug approval rates over the past decades. Conventional in vitro drug screening platforms employ two-dimensional (2D) cell culture models, which demonstrate inaccurate drug responses by failing to capture the three-dimensional (3D) tissue microenvironment in vivo. Recent advancements in the field of tissue engineering have made possible the creation of 3D cell culture systems that can accurately recapitulate the cell-cell and cell-extracellular matrix interactions, as well as replicate the intricate microarchitectures observed in native tissues. However, the lack of a perfusion system in 3D cell cultures hinders the establishment of the models as potential drug screening platforms. Over the years, multiple techniques have successfully demonstrated vascularization in 3D cell cultures, simulating in vivo-like drug interactions, proposing the use of 3D systems as drug screening platforms to eliminate the deviations between preclinical and in vivo testing. In this review, the basic principles of 3D cell culture systems are briefly introduced, and current research demonstrating the development of vascularization in 3D cell cultures is discussed, with a particular focus on the potential of these models as the future of drug screening platforms.
Collapse
Affiliation(s)
- Shamapto Guha Anthon
- Department of Biomedical Engineering, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | | |
Collapse
|
17
|
Comparison of EMT-Related and Multi-Drug Resistant Gene Expression, Extracellular Matrix Production, and Drug Sensitivity in NSCLC Spheroids Generated by Scaffold-Free and Scaffold-Based Methods. Int J Mol Sci 2022; 23:ijms232113306. [PMID: 36362093 PMCID: PMC9657250 DOI: 10.3390/ijms232113306] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/27/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022] Open
Abstract
Multicellular 3D tumor models are becoming a powerful tool for testing of novel drug products and personalized anticancer therapy. Tumor spheroids, a commonly used 3D multicellular tumor model, more closely reproduce the tumor microenvironment than conventional 2D cell cultures. It should be noted that spheroids can be produced using different techniques, which can be subdivided into scaffold-free (SF) and scaffold-based (SB) methods. However, it remains unclear, to what extent spheroid properties depend on the method of their generation. In this study, we aimed to carry out a head-to-head comparison of drug sensitivity and molecular expression profile in SF and SB spheroids along with a monolayer (2D) cell culture. Here, we produced non-small cell lung cancer (NSCLC) spheroids based on human lung adenocarcinoma cell line A549. Drug sensitivity analysis of the tested cell cultures to five different chemotherapeutics resulted in IC50 (A549-SB) > IC50 (A549-SF) > IC50 (A549-2D) trend. It was found that SF and SB A549 spheroids displayed elevated expression levels of epithelial-to-mesenchymal transition (EMT) markers and proteins associated with drug resistance compared with the monolayer A549 cell culture. Enhanced drug resistance of A549-SB spheroids can be a result of larger diameters and elevated deposition of extracellular matrix (ECM) that impairs drug penetration into spheroids. Thus, the choice of the spheroid production method can influence the properties of the generated 3D cell culture and their drug resistance. This fact should be considered for correct interpretation of drug testing results.
Collapse
|
18
|
Bhatt R, Ravi D, Evens AM, Parekkadan B. Scaffold-mediated switching of lymphoma metabolism in culture. Cancer Metab 2022; 10:15. [PMID: 36224623 PMCID: PMC9559005 DOI: 10.1186/s40170-022-00291-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 09/22/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Diffuse large B cell lymphoma (DLBCL) is an aggressive subtype of non-Hodgkin lymphoma (NHL) and accounts for about a third of all NHL cases. A significant proportion (~40%) of treated DLBCL patients develop refractory or relapsed disease due to drug resistance which can be attributed to metabolomic and genetic variations amongst diverse DLBCL subtypes. An assay platform that reproduces metabolic patterns of DLBCL in vivo could serve as a useful model for DLBCL. METHODS This report investigated metabolic functions in 2D and 3D cell cultures using parental and drug-resistant DLBCL cell lines as compared to patient biopsy tissue. RESULTS A 3D culture model controlled the proliferation of parental and drug-resistant DLBCL cell lines, SUDHL-10, SUDHL-10 RR (rituximab resistant), and SUDHL-10 OR (obinutuzumab resistant), as well as retained differential sensitivity to CHOP. The results from metabolic profiling and isotope tracer studies with D-glucose-13C6 indicated metabolic switching in 3D culture when compared with a 2D environment. Analysis of DLBCL patient tumor tissue revealed that the metabolic changes in 3D grown cells were shifted towards that of clinical specimens. CONCLUSION 3D culture restrained DLBCL cell line growth and modulated metabolic pathways that trend towards the biological characteristics of patient tumors. Counter-intuitively, this research thereby contends that 3D matrices can be a tool to control tumor function towards a slower growing and metabolically dormant state that better reflects in vivo tumor physiology.
Collapse
Affiliation(s)
- Rachana Bhatt
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Dashnamoorthy Ravi
- Division of Blood Disorders, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Andrew M Evens
- Division of Blood Disorders, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Biju Parekkadan
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
- Department of Medicine, Rutgers Biomedical Health Sciences, The State University of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
19
|
Ingavle G, Das M. Bench to Bedside: New Therapeutic Approaches with Extracellular Vesicles and Engineered Biomaterials for Targeting Therapeutic Resistance of Cancer Stem Cells. ACS Biomater Sci Eng 2022; 8:4673-4696. [PMID: 36194142 DOI: 10.1021/acsbiomaterials.2c00484] [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: 11/28/2022]
Abstract
Cancer has recently been the second leading cause of death worldwide, trailing only cardiovascular disease. Cancer stem cells (CSCs), represented as tumor-initiating cells (TICs), are mainly liable for chemoresistance and disease relapse due to their self-renewal capability and differentiating capacity into different types of tumor cells. The intricate molecular mechanism is necessary to elucidate CSC's chemoresistance properties and cancer recurrence. Establishing efficient strategies for CSC maintenance and enrichment is essential to elucidate the mechanisms and properties of CSCs and CSC-related therapeutic measures. Current approaches are insufficient to mimic the in vivo chemical and physical conditions for the maintenance and growth of CSC and yield unreliable research results. Biomaterials are now widely used for simulating the bone marrow microenvironment. Biomaterial-based three-dimensional (3D) approaches for the enrichment of CSC provide an excellent promise for future drug discovery and elucidation of molecular mechanisms. In the future, the biomaterial-based model will contribute to a more operative and predictive CSC model for cancer therapy. Design strategies for materials, physicochemical cues, and morphology will offer a new direction for future modification and new methods for studying the CSC microenvironment and its chemoresistance property. This review highlights the critical roles of the microenvironmental cues that regulate CSC function and endow them with drug resistance properties. This review also explores the latest advancement and challenges in biomaterial-based scaffold structure for therapeutic approaches against CSC chemoresistance. Since the recent entry of extracellular vesicles (EVs), cell-derived nanostructures, have opened new avenues of investigation into this field, which, together with other more conventionally studied signaling pathways, play an important role in cell-to-cell communication. Thus, this review further explores the subject of EVs in-depth. This review also discusses possible future biomaterial and biomaterial-EV-based models that could be used to study the tumor microenvironment (TME) and will provide possible therapeutic approaches. Finally, this review concludes with potential perspectives and conclusions in this area.
Collapse
Affiliation(s)
- Ganesh Ingavle
- Symbiosis Centre for Stem Cell Research (SCSCR) and Symbiosis School of Biological Sciences (SSBS), SIU, Lavale, Pune 412115, India
| | - Madhurima Das
- Symbiosis Centre for Stem Cell Research (SCSCR) and Symbiosis School of Biological Sciences (SSBS), SIU, Lavale, Pune 412115, India
| |
Collapse
|
20
|
Jubelin C, Muñoz-Garcia J, Griscom L, Cochonneau D, Ollivier E, Heymann MF, Vallette FM, Oliver L, Heymann D. Three-dimensional in vitro culture models in oncology research. Cell Biosci 2022; 12:155. [PMID: 36089610 PMCID: PMC9465969 DOI: 10.1186/s13578-022-00887-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 08/18/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractCancer is a multifactorial disease that is responsible for 10 million deaths per year. The intra- and inter-heterogeneity of malignant tumors make it difficult to develop single targeted approaches. Similarly, their diversity requires various models to investigate the mechanisms involved in cancer initiation, progression, drug resistance and recurrence. Of the in vitro cell-based models, monolayer adherent (also known as 2D culture) cell cultures have been used for the longest time. However, it appears that they are often less appropriate than the three-dimensional (3D) cell culture approach for mimicking the biological behavior of tumor cells, in particular the mechanisms leading to therapeutic escape and drug resistance. Multicellular tumor spheroids are widely used to study cancers in 3D, and can be generated by a multiplicity of techniques, such as liquid-based and scaffold-based 3D cultures, microfluidics and bioprinting. Organoids are more complex 3D models than multicellular tumor spheroids because they are generated from stem cells isolated from patients and are considered as powerful tools to reproduce the disease development in vitro. The present review provides an overview of the various 3D culture models that have been set up to study cancer development and drug response. The advantages of 3D models compared to 2D cell cultures, the limitations, and the fields of application of these models and their techniques of production are also discussed.
Collapse
|
21
|
Bayareh M. Active cell capturing for organ-on-a-chip systems: a review. BIOMED ENG-BIOMED TE 2022; 67:443-459. [PMID: 36062551 DOI: 10.1515/bmt-2022-0232] [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: 06/16/2022] [Accepted: 08/25/2022] [Indexed: 11/15/2022]
Abstract
Organ-on-a-chip (OOC) is an emerging technology that has been proposed as a new powerful cell-based tool to imitate the pathophysiological environment of human organs. For most OOC systems, a pivotal step is to culture cells in microfluidic devices. In active cell capturing techniques, external actuators, such as electrokinetic, magnetic, acoustic, and optical forces, or a combination of these forces, can be applied to trap cells after ejecting cell suspension into the microchannel inlet. This review paper distinguishes the characteristics of biomaterials and evaluates microfluidic technology. Besides, various types of OOC and their fabrication techniques are reported and various active cell capture microstructures are analyzed. Furthermore, their constraints, challenges, and future perspectives are provided.
Collapse
Affiliation(s)
- Morteza Bayareh
- Department of Mechanical Engineering, Shahrekord University, Shahrekord, Iran
| |
Collapse
|
22
|
Tamayo-Angorrilla M, López de Andrés J, Jiménez G, Marchal JA. The biomimetic extracellular matrix: a therapeutic tool for breast cancer research. Transl Res 2022; 247:117-136. [PMID: 34844003 DOI: 10.1016/j.trsl.2021.11.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 11/17/2021] [Accepted: 11/21/2021] [Indexed: 12/14/2022]
Abstract
A deeper knowledge of the functional versatility and dynamic nature of the ECM has improved the understanding of cancer biology. Translational Significance: This work provides an in-depth view of the importance of the ECM to develop more mimetic breast cancer models, which aim to recreate the components and architecture of tumor microenvironment. Special focus is placed on decellularized matrices derived from tissue and cell culture, both in procurement and applications, as they have achieved great success in cancer research and pharmaceutical sector. The extracellular matrix (ECM) is increasingly recognized as a master regulator of cell behavior and response to breast cancer (BC) treatment. During BC progression, the mammary gland ECM is remodeled and altered in the composition and organization. Accumulated evidence suggests that changes in the composition and mechanics of ECM, orchestrated by tumor-stromal interactions along with ECM remodeling enzymes, are actively involved in BC progression and metastasis. Understanding how specific ECM components modulate the tumorigenic process has led to an increased interest in the development of biomaterial-based biomimetic ECM models to recapitulate key tumor characteristics. The decellularized ECMs (dECMs) have emerged as a promising in vitro 3D tumor model, whose recent advances in the processing and application could become the biomaterial by excellence for BC research and the pharmaceutical industry. This review offers a detailed view of the contribution of ECM in BC progression, and highlights the application of dECM-based biomaterials as promising personalized tumor models that more accurately mimic the tumorigenic mechanisms of BC and the response to treatment. This will allow the design of targeted therapeutic approaches adapted to the specific characteristics of each tumor that will have a great impact on the precision medicine applied to BC patients.
Collapse
Affiliation(s)
- Marta Tamayo-Angorrilla
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Julia López de Andrés
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria, ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain
| | - Gema Jiménez
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria, ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria, ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.
| |
Collapse
|
23
|
Sahan AZ, Baday M, Patel CB. Biomimetic Hydrogels in the Study of Cancer Mechanobiology: Overview, Biomedical Applications, and Future Perspectives. Gels 2022; 8:gels8080496. [PMID: 36005097 PMCID: PMC9407355 DOI: 10.3390/gels8080496] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/26/2022] [Accepted: 07/02/2022] [Indexed: 11/18/2022] Open
Abstract
Hydrogels are biocompatible polymers that are tunable to the system under study, allowing them to be widely used in medicine, bioprinting, tissue engineering, and biomechanics. Hydrogels are used to mimic the three-dimensional microenvironment of tissues, which is essential to understanding cell–cell interactions and intracellular signaling pathways (e.g., proliferation, apoptosis, growth, and survival). Emerging evidence suggests that the malignant properties of cancer cells depend on mechanical cues that arise from changes in their microenvironment. These mechanobiological cues include stiffness, shear stress, and pressure, and have an impact on cancer proliferation and invasion. The hydrogels can be tuned to simulate these mechanobiological tissue properties. Although interest in and research on the biomedical applications of hydrogels has increased in the past 25 years, there is still much to learn about the development of biomimetic hydrogels and their potential applications in biomedical and clinical settings. This review highlights the application of hydrogels in developing pre-clinical cancer models and their potential for translation to human disease with a focus on reviewing the utility of such models in studying glioblastoma progression.
Collapse
Affiliation(s)
- Ayse Z. Sahan
- Biomedical Sciences Graduate Program, Department of Pharmacology, School of Medicine, University California at San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Murat Baday
- Department of Neurology and Neurological Sciences, School of Medicine, Stanford University, Stanford, CA 94305, USA
- Precision Health and Integrated Diagnostics Center, School of Medicine, Stanford University, Stanford, CA 94305, USA
- Correspondence: (M.B.); (C.B.P.)
| | - Chirag B. Patel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX 77030, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX 77030, USA
- Correspondence: (M.B.); (C.B.P.)
| |
Collapse
|
24
|
Kim D, Hwang KS, Seo EU, Seo S, Lee BC, Choi N, Choi J, Kim HN. Vascularized Lung Cancer Model for Evaluating the Promoted Transport of Anticancer Drugs and Immune Cells in an Engineered Tumor Microenvironment. Adv Healthc Mater 2022; 11:e2102581. [PMID: 35286780 PMCID: PMC11468795 DOI: 10.1002/adhm.202102581] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/08/2022] [Indexed: 01/09/2023]
Abstract
The tumor microenvironment (TME) is the environment around the tumor, including blood vessels, immune cells, fibroblasts, signaling molecules, and the extracellular matrix (ECM). Owing to its component interactions, the TME influences tumor growth and drug delivery in a highly complex manner. Although several vascularized cancer models are developed to mimic the TME in vitro, these models cannot comprehensively reflect blood vessel-tumor spheroid interactions. Here, a method for inducing controlled tumor angiogenesis by engineering the microenvironment is presented. The interstitial flow direction regulates the direction of capillary sprouting, showing that angiogenesis occurs in the opposite direction of flow, while the existence of lung fibroblasts affects the continuity and lumen formation of sprouted capillaries. The vascularized tumor model shows enhanced delivery of anticancer drugs and immune cells to the tumor spheroids because of the perfusable vascular networks. The possibility of capillary embolism using anticancer drug-conjugated liquid metal nanoparticles is investigated using the vascularized tumor model. This vascularized tumor platform can aid in the development of effective anticancer drugs and cancer immunotherapy.
Collapse
Affiliation(s)
- Dasom Kim
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
| | - Kyeong Seob Hwang
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- School of Mechanical EngineeringYonsei UniversitySeoul03722Republic of Korea
| | - Eun U Seo
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and Technology (UST)Seoul02792Republic of Korea
| | - Suyeong Seo
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Program in Nano Science and TechnologyGraduate School of Convergence Science and TechnologySeoul National UniversitySeoul08826Republic of Korea
| | - Byung Chul Lee
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and Technology (UST)Seoul02792Republic of Korea
| | - Nakwon Choi
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and Technology (UST)Seoul02792Republic of Korea
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
| | - Jonghoon Choi
- School of Integrative EngineeringChung‐Ang UniversitySeoul06974Republic of Korea
| | - Hong Nam Kim
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- School of Mechanical EngineeringYonsei UniversitySeoul03722Republic of Korea
- Division of Bio‐Medical Science and TechnologyKIST SchoolKorea University of Science and Technology (UST)Seoul02792Republic of Korea
- Yonsei‐KIST Convergence Research InstituteYonsei UniversitySeoul03722Republic of Korea
| |
Collapse
|
25
|
Shao R, Wang Y, Li L, Dong Y, Zhao J, Liang W. Bone tumors effective therapy through functionalized hydrogels: current developments and future expectations. Drug Deliv 2022; 29:1631-1647. [PMID: 35612368 PMCID: PMC9154780 DOI: 10.1080/10717544.2022.2075983] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Primary bone tumors especially, sarcomas affect adolescents the most because it originates from osteoblasts cells responsible for bone growth. Chemotherapy, surgery, and radiation therapy are the most often used clinical treatments. Regrettably, surgical resection frequently fails to entirely eradicate the tumor, which is the primary cause of metastasis and postoperative recurrence, leading to a high death rate. Additionally, bone tumors frequently penetrate significant regions of bone, rendering them incapable of self-repair, and impairing patients' quality of life. As a result, treating bone tumors and regenerating bone in the clinic is difficult. In recent decades, numerous sorts of alternative therapy approaches have been investigated due to a lack of approved treatments. Among the novel therapeutic approaches, hydrogel-based anticancer therapy has cleared the way for the development of new targeted techniques for treating bone cancer and bone regeneration. They include strategies such as co-delivery of several drug payloads, enhancing their biodistribution and transport capabilities, normalizing accumulation, and optimizing drug release profiles to decrease the limitations of current therapy. This review discusses current advances in functionalized hydrogels to develop a new technique for treating bone tumors by reducing postoperative tumor recurrence and promoting tissue repair.
Collapse
Affiliation(s)
- Ruyi Shao
- Department of Orthopedics, Zhuji People's Hospital, Shaoxing, Zhejiang, China
| | - Yeben Wang
- Department of Traumatic Orthopedics, Affiliated Jinan Third Hospital of Jining Medical University, Jinan, Shandong, China
| | - Laifeng Li
- Department of Traumatic Orthopedics, Affiliated Jinan Third Hospital of Jining Medical University, Jinan, Shandong, China
| | - Yongqiang Dong
- Department of Orthopaedics, Xinchang People's Hospital, Shaoxing, Zhejiang, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, Zhejiang, China
| | - Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, Zhejiang, China
| |
Collapse
|
26
|
Azees PAA, Natarajan S, Amaechi BT, Thajuddin N, Raghavendra VB, Brindhadevi K, Pugazhendhi A. An empirical review on the risk factors, therapeutic strategies and materials at nanoscale for the treatment of oral malignancies. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
27
|
Biomimetic hydrogel supports initiation and growth of patient-derived breast tumor organoids. Nat Commun 2022; 13:1466. [PMID: 35304464 PMCID: PMC8933543 DOI: 10.1038/s41467-022-28788-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 02/01/2022] [Indexed: 12/15/2022] Open
Abstract
Patient-derived tumor organoids (PDOs) are a highly promising preclinical model that recapitulates the histology, gene expression, and drug response of the donor patient tumor. Currently, PDO culture relies on basement-membrane extract (BME), which suffers from batch-to-batch variability, the presence of xenogeneic compounds and residual growth factors, and poor control of mechanical properties. Additionally, for the development of new organoid lines from patient-derived xenografts, contamination of murine host cells poses a problem. We propose a nanofibrillar hydrogel (EKGel) for the initiation and growth of breast cancer PDOs. PDOs grown in EKGel have histopathologic features, gene expression, and drug response that are similar to those of their parental tumors and PDOs in BME. In addition, EKGel offers reduced batch-to-batch variability, a range of mechanical properties, and suppressed contamination from murine cells. These results show that EKGel is an improved alternative to BME matrices for the initiation, growth, and maintenance of breast cancer PDOs. Patient-derived tumour organoids are important preclinical models but suffer from variability from the use of basement-membrane extract and cell contamination. Here, the authors report on the development of mimetic nanofibrilar hydrogel which supports tumour organoid growth with reduced batch variability and cell contamination.
Collapse
|
28
|
Law AMK, Rodriguez de la Fuente L, Grundy TJ, Fang G, Valdes-Mora F, Gallego-Ortega D. Advancements in 3D Cell Culture Systems for Personalizing Anti-Cancer Therapies. Front Oncol 2021; 11:782766. [PMID: 34917509 PMCID: PMC8669727 DOI: 10.3389/fonc.2021.782766] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/11/2021] [Indexed: 01/09/2023] Open
Abstract
Over 90% of potential anti-cancer drug candidates results in translational failures in clinical trials. The main reason for this failure can be attributed to the non-accurate pre-clinical models that are being currently used for drug development and in personalised therapies. To ensure that the assessment of drug efficacy and their mechanism of action have clinical translatability, the complexity of the tumor microenvironment needs to be properly modelled. 3D culture models are emerging as a powerful research tool that recapitulates in vivo characteristics. Technological advancements in this field show promising application in improving drug discovery, pre-clinical validation, and precision medicine. In this review, we discuss the significance of the tumor microenvironment and its impact on therapy success, the current developments of 3D culture, and the opportunities that advancements that in vitro technologies can provide to improve cancer therapeutics.
Collapse
Affiliation(s)
- Andrew M K Law
- Tumour Development Group, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Randwick, NSW, Australia
| | - Laura Rodriguez de la Fuente
- Tumour Development Group, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Randwick, NSW, Australia.,Cancer Epigenetic Biology and Therapeutics Lab, Children's Cancer Institute, Randwick, NSW, Australia
| | - Thomas J Grundy
- Life Sciences, Inventia Life Science Pty Ltd, Alexandria, NSW, Australia
| | - Guocheng Fang
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, Australia
| | - Fatima Valdes-Mora
- Cancer Epigenetic Biology and Therapeutics Lab, Children's Cancer Institute, Randwick, NSW, Australia.,School of Women's and Children's Health, Faculty of Medicine, University of New South Wales Sydney, Randwick, NSW, Australia
| | - David Gallego-Ortega
- Tumour Development Group, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Randwick, NSW, Australia.,School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, Australia
| |
Collapse
|
29
|
Lee SY, Teng Y, Son M, Ku B, Moon HS, Tergaonkar V, Chow PKH, Lee DW, Nam DH. High-dose drug heat map analysis for drug safety and efficacy in multi-spheroid brain normal cells and GBM patient-derived cells. PLoS One 2021; 16:e0251998. [PMID: 34855773 PMCID: PMC8638871 DOI: 10.1371/journal.pone.0251998] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 11/01/2021] [Indexed: 11/19/2022] Open
Abstract
To test the safety and efficacy of drugs via a high does drug heat map, a multi-spheroids array chip was developed by adopting a micropillar and microwell structure. In the chip, patient-derived cells were encapsulated in alginate and grown to maturity for more than 7 days to form cancer multi-spheroids. Multi-spheroids grown in conventional well plates require many cells and are easily damaged as a result of multiple pipetting during maintenance culture or experimental procedures. To address these issues, we applied a micropillar and microwell structure to the multi-spheroids array. Patient-derived cells from patients with Glioblastoma (GBM), the most common and lethal form of central nervous system cancer, were used to validate the array chip performance. After forming multi-spheroids with a diameter greater than 100μm in a 12×36 pillar array chip (25mm × 75mm), we tested 70 drug compounds (6 replicates) using a high-dose to determine safety and efficacy for drug candidates. Comparing the drug response of multi-spheroids derived from normal cells and cancer cells, we found that four compounds (Dacomitinib, Cediranib, LY2835219, BGJ398) did not show toxicity to astrocyte cell and were efficacious to patient-derived GBM cells.
Collapse
Affiliation(s)
- Sang-Yun Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
- Central R & D Center, Medical & Bio Device (MBD) Co., Ltd, Suwon, Republic of Korea
| | - Yvonne Teng
- Research & Development Department, AVATAMED Pte. Ltd., Singapore, Singapore
| | - Miseol Son
- Research & Development Department, AVATAMED Pte. Ltd., Singapore, Singapore
| | - Bosung Ku
- Central R & D Center, Medical & Bio Device (MBD) Co., Ltd, Suwon, Republic of Korea
| | - Ho Sang Moon
- Central R & D Center, Medical & Bio Device (MBD) Co., Ltd, Suwon, Republic of Korea
| | - Vinay Tergaonkar
- Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
| | - Pierce Kah-Hoe Chow
- Division of Surgery and Surgical Oncology, National Cancer Centre Singapore (NCCS), Singapore, Singapore
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital (SGH), Singapore, Singapore
- Surgery Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore
- Faculty (Senior Group Leader), Genome Institute of Singapore (GIS), Singapore, Singapore
- Research Director, Institute of Molecular Cell Biology (IMCB), Singapore, Singapore
| | - Dong Woo Lee
- Department of Biomedical Engineering, Konyang University, Daejon, Korea
| | - Do-Hyun Nam
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
30
|
Deng M, Rao JD, Guo R, Li M, He Q. Size-Adjustable Nano-Drug Delivery Systems for Enhanced Tumor Retention and Penetration. PHARMACEUTICAL FRONTS 2021. [DOI: 10.1055/s-0041-1736474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Over the past decades, nano-drug delivery systems have shown great potential in improving tumor treatment. And the controllability and design flexibility of nanoparticles endow them a broad development space. The particle size is one of the most important factors affecting the potency of nano-drug delivery systems. Large-size (100–200 nm) nanoparticles are more conducive to long circulation and tumor retention, but have poor tumor penetration; small-size (<50 nm) nanoparticles can deeply penetrate tumor but are easily cleared. Most of the current fixed-size nanoparticles are difficult to balance the retention and penetration, while the proposal of size-adjustable nano-drug delivery systems offers a solution to this paradox. Many endogenous and exogenous stimuli, such as acidic pH, upregulated enzymes, temperature, light, catalysts, redox conditions, and reactive oxygen species, can trigger the in situ transformation of nanoparticles based on protonation, hydrolysis, click reaction, phase transition, photoisomerization, redox reaction, etc. In this review, we summarize the principles and applications of stimuli-responsive size-adjustable strategies, including size-enlargement strategies and size-shrinkage strategies. We also propose the challenges faced by size-adjustable nano-drug delivery systems, hoping to promote the development of this strategy.
Collapse
Affiliation(s)
- Miao Deng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| | - Jing-Dong Rao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| | - Rong Guo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
31
|
Habanjar O, Diab-Assaf M, Caldefie-Chezet F, Delort L. 3D Cell Culture Systems: Tumor Application, Advantages, and Disadvantages. Int J Mol Sci 2021; 22:12200. [PMID: 34830082 PMCID: PMC8618305 DOI: 10.3390/ijms222212200] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 01/09/2023] Open
Abstract
The traditional two-dimensional (2D) in vitro cell culture system (on a flat support) has long been used in cancer research. However, this system cannot be fully translated into clinical trials to ideally represent physiological conditions. This culture cannot mimic the natural tumor microenvironment due to the lack of cellular communication (cell-cell) and interaction (cell-cell and cell-matrix). To overcome these limitations, three-dimensional (3D) culture systems are increasingly developed in research and have become essential for tumor research, tissue engineering, and basic biology research. 3D culture has received much attention in the field of biomedicine due to its ability to mimic tissue structure and function. The 3D matrix presents a highly dynamic framework where its components are deposited, degraded, or modified to delineate functions and provide a platform where cells attach to perform their specific functions, including adhesion, proliferation, communication, and apoptosis. So far, various types of models belong to this culture: either the culture based on natural or synthetic adherent matrices used to design 3D scaffolds as biomaterials to form a 3D matrix or based on non-adherent and/or matrix-free matrices to form the spheroids. In this review, we first summarize a comparison between 2D and 3D cultures. Then, we focus on the different components of the natural extracellular matrix that can be used as supports in 3D culture. Then we detail different types of natural supports such as matrigel, hydrogels, hard supports, and different synthetic strategies of 3D matrices such as lyophilization, electrospiding, stereolithography, microfluid by citing the advantages and disadvantages of each of them. Finally, we summarize the different methods of generating normal and tumor spheroids, citing their respective advantages and disadvantages in order to obtain an ideal 3D model (matrix) that retains the following characteristics: better biocompatibility, good mechanical properties corresponding to the tumor tissue, degradability, controllable microstructure and chemical components like the tumor tissue, favorable nutrient exchange and easy separation of the cells from the matrix.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie Moléculaire et Anticancéreuse, Faculté des Sciences II, Université Libanaise Fanar, Beyrouth 1500, Liban;
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| |
Collapse
|
32
|
Phon BWS, Kamarudin MNA, Bhuvanendran S, Radhakrishnan AK. Transitioning pre-clinical glioblastoma models to clinical settings with biomarkers identified in 3D cell-based models: A systematic scoping review. Biomed Pharmacother 2021; 145:112396. [PMID: 34775238 DOI: 10.1016/j.biopha.2021.112396] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/25/2021] [Accepted: 11/02/2021] [Indexed: 11/02/2022] Open
Abstract
Glioblastoma (GBM) remains incurable despite the overwhelming discovery of 2-dimensional (2D) cell-based potential therapeutics since the majority of them have met unsatisfactory results in animal and clinical settings. Incremental empirical evidence has laid the widespread need of transitioning 2D to 3-dimensional (3D) cultures that better mimic GBM's complex and heterogenic nature to allow better translation of pre-clinical results. This systematic scoping review analyses the transcriptomic data involving 3D models of GBM against 2D models from 22 studies identified from four databases (PubMed, ScienceDirect, Medline, and Embase). From a total of 499 genes reported in these studies, 313 (63%) genes were upregulated across 3D models cultured using different scaffolds. Our analysis showed that 4 of the replicable upregulated genes are associated with GBM stemness, epithelial to mesenchymal transition (EMT), hypoxia, and migration-related genes regardless of the type of scaffolds, displaying close resemblances to primitive undifferentiated tumour phenotypes that are associated with decreased overall survival and increased hazard ratio in GBM patients. The upregulation of drug response and drug efflux genes (e.g. cytochrome P450s and ABC transporters) mirrors the GBM genetic landscape that contributes to in vivo and clinical treatment resistance. These upregulated genes displayed strong protein-protein interactions when analysed using an online bioinformatics software (STRING). These findings reinforce the need for widespread transition to 3D GBM models as a relatively inexpensive humanised pre-clinical tool with suitable genetic biomarkers to bridge clinical gaps in potential therapeutic evaluations.
Collapse
Affiliation(s)
- Brandon Wee Siang Phon
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Muhamad N A Kamarudin
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor, Malaysia.
| | - Saatheeyavaane Bhuvanendran
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor, Malaysia
| | - Ammu K Radhakrishnan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
33
|
Pais AS, Reis S, Laranjo M, Caramelo F, Silva F, Botelho MF, Almeida-Santos T. The challenge of ovarian tissue culture: 2D versus 3D culture. J Ovarian Res 2021; 14:147. [PMID: 34724957 PMCID: PMC8561954 DOI: 10.1186/s13048-021-00892-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cryopreservation of ovarian tissue is a powerful technique for preserving female fertility, as it can restore fertility and endocrine function. To increase the longevity of the transplant and decrease the risk of reimplantation of neoplastic cells, several studies have been carried out with culture of ovarian tissue. The aim of this study was to compare a conventional (2D) culture with an alginate matrix three-dimensional (3D) model for ovarian tissue culture. RESULTS The ovarian tissue culture within the alginate matrix (3D) was similar to 2D culture, regarding follicular density and cell apoptosis in follicles and stroma. The proliferation rate remained stable in both models for follicles, but for stromal cell proliferation it decreased only in 3D culture (p = 0.001). At 24 h of culture, cytotoxicity was lower in the 3D model (p = 0.006). As culture time increased, cytotoxicity seemed similar. Degradation of the tissue was suggested by the histological score analysis of tissue morphology after 72 h of culture. Tissue injury was greater (p = 0.01) in 3D culture due to higher interstitial oedema (p = 0.017) and tissue necrosis (p = 0.035). CONCLUSION According to our results, 3D culture of ovarian tissue has no advantage over 2Dculture; it is more time consuming and difficult to perform and has worse reproducibility.
Collapse
Affiliation(s)
- Ana Sofia Pais
- Reproductive Medicine Unit, Centro Hospitalar e Universitário de Coimbra (CHUC), E.P.E., Coimbra, Portugal.
- Obstetrics Department, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal.
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.
| | - Sandra Reis
- Reproductive Medicine Unit, Centro Hospitalar e Universitário de Coimbra (CHUC), E.P.E., Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), CIBB, Azinhaga de Santa Comba, Celas, University of Coimbra, Coimbra, Portugal
- PhD Programme in Experimental Biology and Biomedicine, IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Mafalda Laranjo
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Francisco Caramelo
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Fátima Silva
- Pathology Unit, Centro Hospitalar e Universitário de Coimbra (CHUC), E.P.E., Coimbra, Portugal
| | - Maria Filomena Botelho
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Teresa Almeida-Santos
- Reproductive Medicine Unit, Centro Hospitalar e Universitário de Coimbra (CHUC), E.P.E., Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), CIBB, Azinhaga de Santa Comba, Celas, University of Coimbra, Coimbra, Portugal
- University of Coimbra, Faculty of Medicine, Coimbra, Portugal
| |
Collapse
|
34
|
Costard LS, Hosn RR, Ramanayake H, O'Brien FJ, Curtin CM. Influences of the 3D microenvironment on cancer cell behaviour and treatment responsiveness: A recent update on lung, breast and prostate cancer models. Acta Biomater 2021; 132:360-378. [PMID: 33484910 DOI: 10.1016/j.actbio.2021.01.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/21/2022]
Abstract
The majority of in vitro studies assessing cancer treatments are performed in two-dimensional (2D) monolayers and are subsequently validated in in vivo animal models. However, 2D models fail to accurately model the tumour microenvironment. Furthermore, animal models are not directly applicable to mimic the human scenario. Three-dimensional (3D) culture models may help to address the discrepancies of 2D and animal models. When cancer cells escape the primary tumour, they can invade at distant organs building secondary tumours, called metastasis. The development of metastasis leads to a dramatic decrease in the life expectancy of patients. Therefore, 3D systems to model the microenvironment of metastasis have also been developed. Several studies have demonstrated changes in cell behaviour and gene expression when cells are cultured in 3D compared to 2D and concluded a better comparability to cells in vivo. Of special importance is the effect seen in response to anti-cancer treatments as models are built primarily to serve as drug-testing platforms. This review highlights these changes between cancer cells grown in 2D and 3D models for some of the most common cancers including lung, breast and prostate tumours. In addition to models aiming to mimic the primary tumour site, the effects of 3D cell culturing in bone metastasis models are also described. STATEMENT OF SIGNIFICANCE: Most in vitro studies in cancer research are performed in 2D and are subsequently validated in in vivo animal models. However, both models possess numerous limitations: 2D models fail to accurately model the tumour microenvironment while animal models are expensive, time-consuming and can differ considerably from humans. It is accepted that the cancer microenvironment plays a critical role in the disease, thus, 3D models have been proposed as a potential solution to address the discrepancies of 2D and animal models. This review highlights changes in cell behaviour, including proliferation, gene expression and chemosensitivity, between cancer cells grown in 2D and 3D models for some of the most common cancers including lung, breast and prostate cancer as well as bone metastasis.
Collapse
|
35
|
Ding Y, Li Z, Hu W, Feng X, Chen Y, Yan G, Wang Y, Zhu B, Yao W, Zheng L, He M, Gao M, Zhao J. Carbazate-modified cross-linked dextran microparticles suppress the progression of osteoarthritis by ROS scavenging. Biomater Sci 2021; 9:6236-6250. [PMID: 34365495 DOI: 10.1039/d1bm00743b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A series of modified polysaccharide microparticles have been fabricated and their potential application for scavenging reactive oxygen species (ROS) and their derivatives to achieve osteoarthritis (OA) treatment has been explored. These microparticles were cross-linked dextran (Sephadex) with different carbazate substitution ratios determined by the TNBS assay and elemental analysis. It has been demonstrated that they could effectively scavenge carbonylated proteins and ROS including hydroxyl radicals (˙OH), superoxide anions (˙O2-) and H2O2 and their derivatives with high efficiency, improve the viability of H2O2-treated chondrocytes by reducing their ROS levels, as well as lower their inflammatory factors. The above ability of antioxidation and inflammation resistance improved with the increase of carbazate substitution ratio. Significantly, this work provided the proof that modified Sephadex successfully alleviated the deterioration of cartilage and the progression of OA in vivo. The proposed microparticles showed a very promising capability for reducing ROS levels and further treating OA.
Collapse
Affiliation(s)
- Yanfeng Ding
- Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China. .,Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Zhimin Li
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Division of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| | - Wenwen Hu
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| | - Xianjing Feng
- Pharmaceutical College, Guangxi Medical University, Nanning, 530021, China
| | - Ying Chen
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| | - Guohua Yan
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Yonglin Wang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Bo Zhu
- Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China. .,Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Wei Yao
- Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China. .,Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Li Zheng
- Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China. .,Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Maolin He
- Division of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| | - Ming Gao
- Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China. .,Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Jinmin Zhao
- Guangxi Collaborative Innovation Center for Biomedicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China. .,Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.,Guangxi Key Liboratory of Regenerative Medicine, Guangxi Medical University, Nanning, 530021, China
| |
Collapse
|
36
|
Chang CY, Johnson HC, Babb O, Fishel ML, Lin CC. Biomimetic stiffening of cell-laden hydrogels via sequential thiol-ene and hydrazone click reactions. Acta Biomater 2021; 130:161-171. [PMID: 34087443 DOI: 10.1016/j.actbio.2021.05.054] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/11/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022]
Abstract
Hydrogels with dynamically tunable crosslinking are invaluable for directing stem cell fate and mimicking a stiffening matrix during fibrosis or tumor development. The increases in matrix stiffness during tissue development are often accompanied by the accumulation of extracellular matrices (e.g., collagen, hyaluronic acid (HA)), a phenomenon that has received little attention in the development of dynamic hydrogels. In this contribution, we present a gelatin-based cell-laden hydrogel system capable of being dynamically stiffened while accumulating HA, a key glycosaminoglycans (GAG) increasingly deposited by stromal cells during tumor progression. Central to this strategy is the synthesis of a dually-modified gelatin macromer - gelatin-norbornene-carbohydrazide (GelNB-CH), which is susceptible to both thiol-norbornene photopolymerization and hydrazone click chemistry. We demonstrate that the crosslinking density of cell-laden thiol-norbornene hydrogels can be dynamically tuned via simple incubation with aldehyde-bearing macromers (e.g., oxidized dextran (oDex) or oHA). The GelNB-CH hydrogel system is highly cytocompatible, as demonstrated by in situ encapsulation of pancreatic cancer cells (PCC) and cancer-associated fibroblasts (CAF). This unique dynamic stiffening scheme provides a platform to study tandem accumulation of HA and elevation in matrix stiffness in the pancreatic tumor microenvironment. STATEMENT OF SIGNIFICANCE: Hydrogels permitting on-demand and secondary crosslinking are ideal for mimicking a stiffening tumor microenvironment (TME). However, none of the current dynamic hydrogels account for both stiffening and accumulation of hyaluronic acid (HA), a major extracellular matrix component increasingly deposited in tumor stromal tissues, including pancreatic ductal adenocarcinoma (PDAC). The current work addresses this gap by developing a dynamic hydrogel system capable of simultaneously increasing stiffness and HA accumulation. This is achieved by a new gelatin macromer permitting sequential thiol-norbornene (for primary network crosslinking) and hydrazone click chemistry (for bioinert or biomimetic stiffening with oxidized dextran (oDex) or oHA, respectively). The results of this study provide new insights into how dynamically changing physicochemical matrix properties guide cancer cell fate processes.
Collapse
Affiliation(s)
- Chun-Yi Chang
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Hunter C Johnson
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Olivia Babb
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Melissa L Fishel
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Chien-Chi Lin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA; Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN, USA.
| |
Collapse
|
37
|
Goodarzi K, Rao SS. Hyaluronic acid-based hydrogels to study cancer cell behaviors. J Mater Chem B 2021; 9:6103-6115. [PMID: 34259709 DOI: 10.1039/d1tb00963j] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Hyaluronic acid (HA) is a natural polysaccharide and a key component of the extracellular matrix (ECM) in many tissues. Therefore, HA-based biomaterials are extensively utilized to create three dimensional ECM mimics to study cell behaviors in vitro. Specifically, derivatives of HA have been commonly used to fabricate hydrogels with controllable properties. In this review, we discuss the various chemistries employed to fabricate HA-based hydrogels as a tunable matrix to mimic the cancer microenvironment and subsequently study cancer cell behaviors in vitro. These include Michael-addition reactions, photo-crosslinking, carbodiimide chemistry, and Diels-Alder chemistry. The utility of these HA-based hydrogels to examine cancer cell behaviors such as proliferation, migration, and invasion in vitro in various types of cancer are highlighted. Overall, such hydrogels provide a biomimetic material-based platform to probe cell-matrix interactions in cancer cells in vitro and study the mechanisms associated with cancer progression.
Collapse
Affiliation(s)
- Kasra Goodarzi
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487-0203, USA.
| | | |
Collapse
|
38
|
Transcending toward Advanced 3D-Cell Culture Modalities: A Review about an Emerging Paradigm in Translational Oncology. Cells 2021; 10:cells10071657. [PMID: 34359827 PMCID: PMC8304089 DOI: 10.3390/cells10071657] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer is a disorder characterized by an uncontrollable overgrowth and a fast-moving spread of cells from a localized tissue to multiple organs of the body, reaching a metastatic state. Throughout years, complexity of cancer progression and invasion, high prevalence and incidence, as well as the high rise in treatment failure cases leading to a poor patient prognosis accounted for continuous experimental investigations on animals and cellular models, mainly with 2D- and 3D-cell culture. Nowadays, these research models are considered a main asset to reflect the physiological events in many cancer types in terms of cellular characteristics and features, replication and metastatic mechanisms, metabolic pathways, biomarkers expression, and chemotherapeutic agent resistance. In practice, based on research perspective and hypothesis, scientists aim to choose the best model to approach their understanding and to prove their hypothesis. Recently, 3D-cell models are seen to be highly incorporated as a crucial tool for reflecting the true cancer cell microenvironment in pharmacokinetic and pharmacodynamics studies, in addition to the intensity of anticancer drug response in pharmacogenomics trials. Hence, in this review, we shed light on the unique characteristics of 3D cells favoring its promising usage through a comparative approach with other research models, specifically 2D-cell culture. Plus, we will discuss the importance of 3D models as a direct reflector of the intrinsic cancer cell environment with the newest multiple methods and types available for 3D-cells implementation.
Collapse
|
39
|
Abstract
Recreating human organ-level function in vitro is a rapidly evolving field that integrates tissue engineering, stem cell biology, and microfluidic technology to produce 3D organoids. A critical component of all organs is the vasculature. Herein, we discuss general strategies to create vascularized organoids, including common source materials, and survey previous work using vascularized organoids to recreate specific organ functions and simulate tumor progression. Vascularization is not only an essential component of individual organ function but also responsible for coupling the fate of all organs and their functions. While some success in coupling two or more organs together on a single platform has been demonstrated, we argue that the future of vascularized organoid technology lies in creating organoid systems complete with tissue-specific microvasculature and in coupling multiple organs through a dynamic vascular network to create systems that can respond to changing physiological conditions.
Collapse
Affiliation(s)
- Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Davis, California 95616, USA;
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California 92697, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, California 95616, USA;
| |
Collapse
|
40
|
Han SJ, Kwon S, Kim KS. Challenges of applying multicellular tumor spheroids in preclinical phase. Cancer Cell Int 2021; 21:152. [PMID: 33663530 PMCID: PMC7934264 DOI: 10.1186/s12935-021-01853-8] [Citation(s) in RCA: 176] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
The three-dimensional (3D) multicellular tumor spheroids (MCTs) model is becoming an essential tool in cancer research as it expresses an intermediate complexity between 2D monolayer models and in vivo solid tumors. MCTs closely resemble in vivo solid tumors in many aspects, such as the heterogeneous architecture, internal gradients of signaling factors, nutrients, and oxygenation. MCTs have growth kinetics similar to those of in vivo tumors, and the cells in spheroid mimic the physical interaction of the tumors, such as cell-to-cell and cell-to-extracellular matrix interactions. These similarities provide great potential for studying the biological properties of tumors and a promising platform for drug screening and therapeutic efficacy evaluation. However, MCTs are not well adopted as preclinical tools for studying tumor behavior and therapeutic efficacy up to now. In this review, we addressed the challenges with MCTs application and discussed various efforts to overcome the challenges.
Collapse
Affiliation(s)
- Se Jik Han
- Department of Biomedical Engineering, Graduate School, Kyung Hee University, Seoul, 02447, Korea
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, Seoul, 02447, Korea
| | - Sangwoo Kwon
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, Seoul, 02447, Korea
| | - Kyung Sook Kim
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, Seoul, 02447, Korea.
| |
Collapse
|
41
|
Distinct roles for the hypoxia-inducible transcription factors HIF-1α and HIF-2α in human osteoclast formation and function. Sci Rep 2020; 10:21072. [PMID: 33273561 PMCID: PMC7713367 DOI: 10.1038/s41598-020-78003-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/19/2020] [Indexed: 01/04/2023] Open
Abstract
Bone homeostasis is maintained by a balance between osteoblast-mediated bone formation and osteoclast-driven bone resorption. Hypoxia modulates this relationship partially via direct and indirect effects of the hypoxia-inducible factor-1 alpha (HIF-1α) transcription factor on osteoclast formation and bone resorption. Little data is available on the role(s) of the HIF-2α isoform of HIF in osteoclast biology. Here we describe induction of HIF-1α and HIF-2α during the differentiation of human CD14+ monocytes into osteoclasts. Knockdown of HIF-1α did not affect osteoclast differentiation but prevented the increase in bone resorption that occurs under hypoxic conditions. HIF-2α knockdown did not affect bone resorption but moderately inhibited osteoclast formation. Growth of osteoclasts in 3D gels reversed the effect of HIF-2α knockdown; HIF-2α siRNA increasing osteoclast formation in 3D. Glycolysis is the main HIF-regulated pathway that drives bone resorption. HIF knockdown only affected glucose uptake and bone resorption in hypoxic conditions. Inhibition of glycolysis with 2-deoxy-d-glucose (2-DG) reduced osteoclast formation and activity under both basal and hypoxic conditions, emphasising the importance of glycolytic metabolism in osteoclast biology. In summary, HIF-1α and HIF-2α play different but overlapping roles in osteoclast biology, highlighting the importance of the HIF pathway as a potential therapeutic target in osteolytic disease.
Collapse
|
42
|
Natural and Synthetic Biomaterials for Engineering Multicellular Tumor Spheroids. Polymers (Basel) 2020; 12:polym12112506. [PMID: 33126468 PMCID: PMC7692845 DOI: 10.3390/polym12112506] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023] Open
Abstract
The lack of in vitro models that represent the native tumor microenvironment is a significant challenge for cancer research. Two-dimensional (2D) monolayer culture has long been the standard for in vitro cell-based studies. However, differences between 2D culture and the in vivo environment have led to poor translation of cancer research from in vitro to in vivo models, slowing the progress of the field. Recent advances in three-dimensional (3D) culture have improved the ability of in vitro culture to replicate in vivo conditions. Although 3D cultures still cannot achieve the complexity of the in vivo environment, they can still better replicate the cell-cell and cell-matrix interactions of solid tumors. Multicellular tumor spheroids (MCTS) are three-dimensional (3D) clusters of cells with tumor-like features such as oxygen gradients and drug resistance, and represent an important translational tool for cancer research. Accordingly, natural and synthetic polymers, including collagen, hyaluronic acid, Matrigel®, polyethylene glycol (PEG), alginate and chitosan, have been used to form and study MCTS for improved clinical translatability. This review evaluates the current state of biomaterial-based MCTS formation, including advantages and disadvantages of the different biomaterials and their recent applications to the field of cancer research, with a focus on the past five years.
Collapse
|
43
|
Murakami S, Mukaisho KI, Iwasa T, Kawabe M, Yoshida S, Taniura N, Nakayama T, Noi M, Yamamoto G, Sugihara H. Application of "Tissueoid Cell Culture System" Using a Silicate Fiber Scaffold for Cancer Research. Pathobiology 2020; 87:291-301. [PMID: 32966983 DOI: 10.1159/000509133] [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: 12/16/2019] [Accepted: 06/03/2020] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND We developed a 3-dimensional (3D) culture system using a high-purity silica fiber scaffold of unwoven sheets called CellbedTM. METHODS We used adherent colon and esophagogastric junction adenocarcinoma cells, tongue squamous cell carcinoma (SqCC) cells, and nonadherent gastric cancer cells. These cells were subjected to staining with various substances and observed by electron microscopy. To evaluate the effects of extracellular matrix in carcinoma tissues, SqCC cells were cultured in Cellbed coated with collagens I, III, and IV. RESULTS Especially well-differentiated carcinoma cells cultured in this 3D system showed their own unique characteristics: luminal formation in adenocarcinoma cells and cell stratification and keratinization in SqCC cells. Scanning electron microscopy revealed the proliferation of cancer cells with cytoplasm entwined in Cellbed. Intercellular desmosomes in squamous epithelia were detected by transmission electron microscopy of vertical cross sections. SqCC cells cultured in Cellbed coated with collagen IV showed enhanced invasive and proliferative abilities. CONCLUSION Because the morphology of cancer cells cultured in this 3D culture system is similar to that in living organisms, we called the system a "tissueoid cell culture system." Coating with collagen IV enables the modification of cell-matrix interactions as well as recapitulation of the in vivo microenvironment.
Collapse
Affiliation(s)
- Shoko Murakami
- Division of Molecular and Diagnostic Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan.,Department of Oral and Maxillofacial Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Ken-Ichi Mukaisho
- Division of Molecular and Diagnostic Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan,
| | - Takuya Iwasa
- Central Research Laboratory, Japan Vilene Company, Ltd, Koga, Japan
| | - Masaaki Kawabe
- Central Research Laboratory, Japan Vilene Company, Ltd, Koga, Japan
| | - Saori Yoshida
- Division of Molecular and Diagnostic Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Naoko Taniura
- Division of Molecular and Diagnostic Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Takahisa Nakayama
- Division of Molecular and Diagnostic Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Masaharu Noi
- Division of Molecular and Diagnostic Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan.,Department of Oral and Maxillofacial Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Gaku Yamamoto
- Department of Oral and Maxillofacial Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Hiroyuki Sugihara
- Division of Molecular and Diagnostic Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
44
|
Development of Injectable Thermosensitive Chitosan-Based Hydrogels for Cell Encapsulation. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10186550] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The three-dimensional complexity of the native extracellular matrix (ECM) suggests switching from 2D to 3D culture systems for providing the cells with an architecture more similar to the physiological environment. Reproducing the three-dimensionality in vitro can guarantee beneficial effects in terms of cell growth, adhesion, proliferation, and/or their differentiation. Hydrogels have the same tailorable physico-chemical and biological characteristics as ECM materials. In this study, we propose a thermoresponsive chitosan-based hydrogel that gels thanks to the addition of organic and inorganic salt solutions (beta-glycerolphosphate and sodium hydrogen carbonate) and is suitable for cell encapsulation allowing obtaining 3D culture systems. Physico-chemical analyses showed that the hydrogel formulations jellify at physiological conditions (37 °C, pH 7.4), are stable in vitro up to three weeks, have high swelling ratios and mechanical stiffness suitable for cellular encapsulation. Moreover, preliminary biological tests underlined the pronounced biocompatibility of the system. Therefore, these chitosan-based hydrogels are proposed as valid biomaterials for cell encapsulation.
Collapse
|
45
|
Balaji P, Murugadas A, Ramkumar A, Thirumurugan R, Shanmugaapriya S, Akbarsha MA. Characterization of Hen's Egg White To Use It as a Novel Platform To Culture Three-Dimensional Multicellular Tumor Spheroids. ACS OMEGA 2020; 5:19760-19770. [PMID: 32803071 PMCID: PMC7424746 DOI: 10.1021/acsomega.0c02508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/23/2020] [Indexed: 06/11/2023]
Abstract
We are standardizing protocols to develop egg white (EW) as a cost-effective platform for culture of three-dimensional (3-D) multicellular tumor spheroids for application in understanding tumor microenvironments and drug screening. In this article, we describe several physical and physiological characteristics of EW to use it as 3-D cell culture platform. Field emission scanning electron microscopy revealed the presence of different microstructures. Hydrodynamic size distribution data indicated nano- and micron-sized particles. Rheological measurements revealed the viscosity and viscoelastic behavior appropriate for maintaining cell viability and supporting 3-D cell growth under high-sheer conditions. It was found that thereis no autofluorescence, a requirement for imparting transparency and for microscopic observations of the spheroids. The EW facilitated the development of 3-D tumor spheroids, with an emphasis of difference in cell proliferation and intercellular cytoskeletal organization between two-dimensional and 3-D spheroid cultures. Put together, EW proves to be a cost-affordable and simple platform for 3-D culture of tumor spheroids.
Collapse
Affiliation(s)
- Perumalsamy Balaji
- Department of Biomedical
Science, Bharathidasan University, Tiruchirapalli 620024, India
- National
Centre for Alternatives to Animal Experiments, Bharathidasan University, Tiruchirappalli 620 024, India
| | - Anbazhagan Murugadas
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia 30322, United States
- National
Centre for Alternatives to Animal Experiments, Bharathidasan University, Tiruchirappalli 620 024, India
| | - Arunachalam Ramkumar
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirapalli 620024, India
- National
Centre for Alternatives to Animal Experiments, Bharathidasan University, Tiruchirappalli 620 024, India
| | - Ramasamy Thirumurugan
- Department of Animal Science, Bharathidasan University, Tiruchirapalli 620024, India
- National
Centre for Alternatives to Animal Experiments, Bharathidasan University, Tiruchirappalli 620 024, India
| | | | - Mohammad Abdulkader Akbarsha
- National
College (Autonomous), Tiruchirappalli 620001, India
- Mahatma Gandhi-Doerenkamp
Centre for Alternatives, Bharathidasan University, Tiruchirappalli 620 024, India
| |
Collapse
|
46
|
Hulley PA, Papadimitriou-Olivgeri I, Knowles HJ. Osteoblast-Osteoclast Coculture Amplifies Inhibitory Effects of FG-4592 on Human Osteoclastogenesis and Reduces Bone Resorption. JBMR Plus 2020; 4:e10370. [PMID: 32666021 PMCID: PMC7340438 DOI: 10.1002/jbm4.10370] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/31/2020] [Accepted: 04/19/2020] [Indexed: 12/13/2022] Open
Abstract
The link between bone and blood vessels is regulated by hypoxia and the hypoxia‐inducible transcription factor, HIF, which drives both osteogenesis and angiogenesis. The recent clinical approval of PHD enzyme inhibitors, which stabilize HIF protein, introduces the potential for a new clinical strategy to treat osteolytic conditions such as osteoporosis, osteonecrosis, and skeletal fracture and nonunion. However, bone‐resorbing osteoclasts also play a central role in bone remodeling and pathological osteolysis, and HIF promotes osteoclast activation and bone loss in vitro. It is therefore likely that the result of PHD enzyme inhibition in vivo would be mediated by a balance between increased bone formation and increased bone resorption. It is essential that we improve our understanding of the effects of HIF on osteoclast formation and function and consider the potential contribution of inhibitory interactions with other musculoskeletal cells. The PHD enzyme inhibitor FG‐4592 stabilized HIF protein and stimulated osteoclast‐mediated bone resorption, but inhibited differentiation of human CD14+ monocytes into osteoclasts. Formation of osteoclasts in a more physiologically relevant 3D collagen gel did not affect the sensitivity of osteoclastogenesis to FG‐4592, but increased sensitivity to reduced concentrations of RANKL. Coculture with osteoblasts amplified inhibition of osteoclastogenesis by FG‐4592, whether the osteoblasts were proliferating, differentiating, or in the presence of exogenous M‐CSF and RANKL. Osteoblast coculture dampened the ability of high concentrations of FG‐4592 to increase bone resorption. These data provide support for the therapeutic use of PHD enzyme inhibitors to improve bone formation and/or reduce bone loss for the treatment of osteolytic pathologies and indicate that FG‐4592 might act in vivo to inhibit the formation and activity of the osteoclasts that drive osteolysis. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Philippa A Hulley
- Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences University of Oxford Oxford UK
| | - Ioanna Papadimitriou-Olivgeri
- Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences University of Oxford Oxford UK.,Department of Anatomy Histology & Embryology University of Patras Patras Greece
| | - Helen J Knowles
- Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences University of Oxford Oxford UK
| |
Collapse
|
47
|
Bayer IS. Hyaluronic Acid and Controlled Release: A Review. Molecules 2020; 25:molecules25112649. [PMID: 32517278 PMCID: PMC7321085 DOI: 10.3390/molecules25112649] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022] Open
Abstract
Hyaluronic acid (HA) also known as hyaluronan, is a natural polysaccharide—an anionic, non-sulfated glycosaminoglycan—commonly found in our bodies. It occurs in the highest concentrations in the eyes and joints. Today HA is used during certain eye surgeries and in the treatment of dry eye disease. It is a remarkable natural lubricant that can be injected into the knee for patients with knee osteoarthritis. HA has also excellent gelling properties due to its capability to bind water very quickly. As such, it is one the most attractive controlled drug release matrices and as such, it is frequently used in various biomedical applications. Due to its reactivity, HA can be cross-linked or conjugated with assorted bio-macromolecules and it can effectively encapsulate several different types of drugs, even at nanoscale. Moreover, the physiological significance of the interactions between HA and its main membrane receptor, CD44 (a cell-surface glycoprotein that modulates cell–cell interactions, cell adhesion and migration), in pathological processes, e.g., cancer, is well recognized and this has resulted in an extensive amount of studies on cancer drug delivery and tumor targeting. HA acts as a therapeutic but also as a tunable matrix for drug release. Thus, this review focuses on controlled or sustained drug release systems assembled from HA and its derivatives. More specifically, recent advances in controlled release of proteins, antiseptics, antibiotics and cancer targeting drugs from HA and its derivatives were reviewed. It was shown that controlled release from HA has many benefits such as optimum drug concentration maintenance, enhanced therapeutic effects, improved efficiency of treatment with less drug, very low or insignificant toxicity and prolonged in vivo release rates.
Collapse
Affiliation(s)
- Ilker S Bayer
- Smart Materials, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| |
Collapse
|
48
|
Hyaluronic acid binding to CD44S is indiscriminate of molecular weight. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183348. [PMID: 32428448 DOI: 10.1016/j.bbamem.2020.183348] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/09/2020] [Accepted: 05/07/2020] [Indexed: 01/06/2023]
Abstract
The ubiquitous presence of hyaluronic acid (HA) in the extracellular matrix (ECM) of both healthy and diseased tissues underscores its importance in human physiology. Previous studies suggest that HA can be used as a probe to qualitatively monitor cell surface levels of CD44 and other important HA receptors; however, these studies use mixtures of HA at various molecular weights. Using fluorescently labeled HA, we evaluated the apparent differences of low (25 kilodalton) and high (700 kilodalton) molecular weight HA interacting with breast cancer cell lines of varying levels of CD44. Our results confirm that CD44 expression and the apparent level of HA interaction correlates with molecular weight. Importantly, we show that HA only binds a small fraction of the major CD44 isoform, CD44S, on cell surfaces and that CD44S interactions account for <50% of the total HA bound to cell surfaces. Although increased fluorescence level correlates with higher molecular weight of HA, this appears to be an artifact of chain length and not a result of multivalent binding between HA and CD44S. Accordingly, we verify that HA binding characteristics of cell surfaces is similar to previous artificial membrane models which proposed that HA anchors to CD44S and forms a non-binding corona of HA that extends beyond the surface.
Collapse
|
49
|
Kaushik S, Gandhi S, Chauhan M, Ma S, Das S, Ghosh D, Chandrasekharan A, Alam MB, Parmar AS, Sharma A, Santhoshkumar TR, Suhag D. Water-Templated, Polysaccharide-rich Bioartificial 3D Microarchitectures as Extra-Cellular Matrix Bioautomatons. ACS APPLIED MATERIALS & INTERFACES 2020; 12:20912-20921. [PMID: 32255604 DOI: 10.1021/acsami.0c01012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
This is the first report of exploiting the "quasi-spherical" shape of water molecules for recapitulating a true human extracellular matrix (ECM). Herein, water behaved as a quasi-spherical porogen, for engineering polysaccharide-rich and chemically defined 3D-microarchitecture, with semi-interpenetrating networks (S-IPNs). Furthermore, their viscoelastic behavior along with a heterogeneous, fibroporous morphology, facilitated instructive, self-remodeling of the bioartificial scaffolds, thence effectively permitting and promoting the growth of 3D tumor spheroids of divergent origins. The hybrid composites displayed reproducible, uniform tumor spheroids with a Z-depth of ∼65 ± 2 μm in case of human adenocarcinoma (DLD-1) and ∼54 ± 3 μm for human glioblastoma cells (U-251) (vs. nonuniform spheroids, on Agarose matrix). Thereafter, their capacity for anticancer drug screening was examined using limited cancer drugs. The conflicting drug screening results for Etoposide's reduced efficacy on glioblastoma cells cultured on our 3D matrix could be ascribed to decreased drug access and thus lower ingression. Nonetheless, adenocarcinoma's resistance to Camptothecin was paralleled. Moreover, their potential for real-time, high-content, phenotypic precision oncology was affirmed by the exceptional transparency of the synthesized composite. Since this 3D microarchitecture typifies ECM bioautomaton, this matrix can also be wielded for precision oncology.
Collapse
Affiliation(s)
- Swati Kaushik
- Institute of Nano Science & Technology, Habitat Centre, Phase 10, Sector 64, Sahibzada Ajit Singh Nagar, Mohali-140307, Punjab, India
- Rajiv Gandhi Centre for Biotechnology, Poojapura, Thycaud, Thiruvananthapuram, Kerala-695014, India
| | - Sonu Gandhi
- DBT-National Institute of Animal Biotechnology, Hyderabad-500032, Telangana, India
| | - Mehak Chauhan
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Sector-125, Noida-201313, Uttar Pradesh, India
| | - Shaohua Ma
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, 518055, China
| | - Souvik Das
- Lab MP3CV, EA7517, University Center for Health Research (CURS), University of Picardie Jules Verne, Amiens 80054, France
| | - Deepa Ghosh
- Institute of Nano Science & Technology, Habitat Centre, Phase 10, Sector 64, Sahibzada Ajit Singh Nagar, Mohali-140307, Punjab, India
| | - Aneesh Chandrasekharan
- Rajiv Gandhi Centre for Biotechnology, Poojapura, Thycaud, Thiruvananthapuram, Kerala-695014, India
| | - Md Bayazeed Alam
- Department of Physics, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Avanish Singh Parmar
- Department of Physics, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Alpana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Sri Aurobindo Marg, Ansari Nagar, Ansari Nagar East, New Delhi-110029, India
| | - T R Santhoshkumar
- Rajiv Gandhi Centre for Biotechnology, Poojapura, Thycaud, Thiruvananthapuram, Kerala-695014, India
| | - Deepa Suhag
- Amity Institute of Biotechnology, Amity University Haryana, Amity Education Valley Gurugram, Manesar, Panchgaon, Haryana 122413, India
| |
Collapse
|
50
|
Guan S, Zhang Q, Bao J, Hu R, Czech T, Tang J. Recognition Sites for Cancer-targeting Drug Delivery Systems. Curr Drug Metab 2020; 20:815-834. [PMID: 31580248 DOI: 10.2174/1389200220666191003161114] [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: 06/19/2019] [Revised: 09/10/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Target-homing drug delivery systems are now gaining significant attention for use as novel therapeutic approaches in antitumor targeting for cancer therapy. Numerous targeted drug delivery systems have been designed to improve the targeting effects because these systems can display a range of favorable properties, thus, providing suitable characteristics for clinical applicability of anticancer drugs, such as increasing the solubility, and improving the drug distribution at target sites. The majority of these targeting systems are designed with respect to differences between cancerous and normal tissues, for instance, the low pH of tumor tissues or overexpressed receptors on tumor cell membranes. Due to the growing number of targeting possibilities, it is important to know the tumor-specific recognition strategies for designing novel, targeted, drug delivery systems. Herein, we identify and summarize literature pertaining to various recognition sites for optimizing the design of targeted drug delivery systems to augment current chemotherapeutic approaches. OBJECTIVE This review focuses on the identification of the recognition sites for developing targeted drug delivery systems for use in cancer therapeutics. METHODS We have reviewed and compiled cancer-specific recognition sites and their abnormal characteristics within tumor tissues (low pH, high glutathione, targetable receptors, etc.), tumor cells (receptor overexpression or tumor cell membrane changes) and tumor cell organelles (nuclear and endoplasmic reticular dysregulation) utilizing existing scientific literature. Moreover, we have highlighted the design of some targeted drug delivery systems that can be used as homing tools for these recognition sites. RESULTS AND CONCLUSION Targeted drug delivery systems are a promising therapeutic approach for tumor chemotherapy. Additional research focused on finding novel recognition sites, and subsequent development of targeting moieties for use with drug delivery systems will aid in the evaluation and clinical application of new and improved chemotherapeutics.
Collapse
Affiliation(s)
- Siyu Guan
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Qianqian Zhang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jianwei Bao
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Rongfeng Hu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Anhui "115" Xin'an Medicine Research & Development Innovation Team, Hefei 230038, China
| | - Tori Czech
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH 44272, United States
| | - Jihui Tang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| |
Collapse
|