1
|
Habibi S, Bahramian S, Saeedeh ZJ, Mehri S, Ababzadeh S, Kavianpour M. Novel strategies in breast cancer management: From treatment to long-term remission. Crit Rev Oncol Hematol 2025; 211:104715. [PMID: 40187709 DOI: 10.1016/j.critrevonc.2025.104715] [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: 03/02/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Breast cancer (BC) is the most common malignancy among women and a leading cause of cancer-related mortality worldwide. Although improvements in early detection and therapy have been made, metastatic breast cancer (mBC) continues to be an incurable disease. Although existing treatments can prolong survival and enhance quality of life, they do not provide a definitive cure. Targeted therapies have significantly improved outcomes, particularly for subtypes such as human epidermal growth factor receptor 2 (HER2)-positive and hormone receptor (HR)-positive (HR+) BC. Key innovations include antibodydrug conjugates (ADCs) and next-generation endocrine therapies. ADCs combine monoclonal antibodies with cytotoxic agents, allowing targeted delivery to tumor cells while minimizing systemic toxicity. Immunotherapy is emerging as a promising approach for aggressive subtypes, such as triple-negative breast cancer (TNBC). Strategies under investigation include chimeric antigen receptor T-cell (CAR-T) therapy, tumor-infiltrating lymphocyte (TIL) therapies, and natural killer (NK) cell treatments, all aimed at enhancing the ability of the immune system to target and eliminate resistant tumor cells. Tissue engineering, particularly hydrogel-based delivery systems, offers the potential for localized treatment. These systems enable the controlled release of therapeutic agents or immune cells directly to the tumor site, supporting tissue regeneration and enhancing immune surveillance to reduce recurrence. Despite these advancements, challenges remain, including treatment resistance, the immunosuppressive tumor microenvironment, and high costs. Overcoming these barriers requires further innovation in drug delivery systems and a deeper understanding of tumor biology.
Collapse
Affiliation(s)
- Sina Habibi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shabbou Bahramian
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Zare Jalise Saeedeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Sara Mehri
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Guilan, Iran
| | - Shima Ababzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran; Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Maria Kavianpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran; Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| |
Collapse
|
2
|
Amini MA, Khodadadi I, Tavilani H, Abbasalipourkabir R, Azizi M, Rashidi K, Samadian H, Karimi J. Fabrication, characterization, and application of gelatin/alginate-based hydrogels incorporating selenium-doped deferoxamine-derived carbon quantum dots: In vitro and in vivo studies. Int J Biol Macromol 2025; 303:140569. [PMID: 39909275 DOI: 10.1016/j.ijbiomac.2025.140569] [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: 10/08/2024] [Revised: 01/16/2025] [Accepted: 01/31/2025] [Indexed: 02/07/2025]
Abstract
This study developed a gelatin/alginate-based nanocomposite hydrogel (NC gel), incorporating selenium-doped deferoxamine-derived carbon quantum dots (Se.DFO-CQDs). Initially, Se.DFO-CQDs were synthesized and characterized through several tests, and subsequently, NC gels were created using an dual crosslinking method and analyzed through characterization tests such as SEM, EDX, FT-IR, XRD, tensile strength, water uptake, water vapor transmission rate, weight loss, porosity, blood compatibility, microbial penetration, and DPPH. In vivo studies revealed that NC gels containing Se.DFO-CQDs at 50 % and 0 % exhibited higher wound closure percentages than the control group. The highest wound closure percentage was observed in NC gels with Se.DFO-CQDs at 50 %, reaching 85.7 ± 3.98 % on the 7th day and 98.1 ± 3.95 % on the 14th day. Histological examinations demonstrated that NC gels with Se.DFO-CQDs at 50 % promoted more significant neovascularization, re-epithelialization, and collagen synthesis. Additionally, RT-qPCR results indicated that NC gels with Se.DFO-CQDs at 50 % significantly upregulated the mRNA expression of VEGF-A, bFGF, PDGF-b, and lncRNA GAS5 on the 7th day and COL1A1 on the 14th day. In conclusion, our findings suggest that the NC gels with Se.DFO-CQDs at 50 % show promise for enhancing wound healing and skin regeneration, potentially offering clinical applications.
Collapse
Affiliation(s)
- Mohammad Amin Amini
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Khodadadi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Heidar Tavilani
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Roghayeh Abbasalipourkabir
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehdi Azizi
- Cancer Research Center, Institute of Cancer, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Khodabakhsh Rashidi
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hadi Samadian
- Research Center for Molecular Medicine, Institute of Cancer, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Jamshid Karimi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
3
|
Feng K, Yi Z, Xu B. Artificial Intelligence and Breast Cancer Management: From Data to the Clinic. CANCER INNOVATION 2025; 4:e159. [PMID: 39981497 PMCID: PMC11840326 DOI: 10.1002/cai2.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 02/22/2025]
Abstract
Breast cancer (BC) remains a significant threat to women's health worldwide. The oncology field had an exponential growth in the abundance of medical images, clinical information, and genomic data. With its continuous advancement and refinement, artificial intelligence (AI) has demonstrated exceptional capabilities in processing intricate multidimensional BC-related data. AI has proven advantageous in various facets of BC management, encompassing efficient screening and diagnosis, precise prognosis assessment, and personalized treatment planning. However, the implementation of AI into precision medicine and clinical practice presents ongoing challenges that necessitate enhanced regulation, transparency, fairness, and integration of multiple clinical pathways. In this review, we provide a comprehensive overview of the current research related to AI in BC, highlighting its extensive applications throughout the whole BC cycle management and its potential for innovative impact. Furthermore, this article emphasizes the significance of constructing patient-oriented AI algorithms. Additionally, we explore the opportunities and potential research directions within this burgeoning field.
Collapse
Affiliation(s)
- Kaixiang Feng
- Department of Breast and Thyroid Surgery, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Zongbi Yi
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
4
|
Santhamoorthy M, Kim SC. A Review of the Development of Biopolymer Hydrogel-Based Scaffold Materials for Drug Delivery and Tissue Engineering Applications. Gels 2025; 11:178. [PMID: 40136883 PMCID: PMC11942562 DOI: 10.3390/gels11030178] [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: 01/10/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Biopolymer hydrogel-based scaffold materials have received a lot of interest in tissue engineering and regenerative medicine because of their unique characteristics, which include biocompatibility, biodegradability, and the ability to replicate the natural extracellular matrix (ECM). These hydrogels are three-dimensional biopolymer networks that are highly hydrated and provide a supportive, wet environment conducive to cell growth, migration, and differentiation. They are especially useful in applications involving wound healing, cartilage, bone, and soft tissue regeneration. Natural biopolymers such as collagen, chitosan, hyaluronic acid, and alginate are frequently employed as the foundation for hydrogel fabrication, providing benefits such as low toxicity and improved cell adherence. Despite their potential, biopolymer hydrogel scaffolds have various difficulties that prevent broad clinical implementation. Key difficulties include the challenge of balancing mechanical strength and flexibility to meet the needs of various tissues, managing degradation rates to line up with tissue regeneration, and assuring large-scale manufacturing while retaining scaffold uniformity and quality. Furthermore, fostering appropriate vascularization and cell infiltration in larger tissues remains a significant challenge for optimal tissue integration and function. Future developments in biopolymer hydrogel-based scaffolds are likely to concentrate on addressing these obstacles. Strategies such as the creation of hybrid hydrogels that combine natural and synthetic materials, smart hydrogels with stimulus-responsive features, and 3D bioprinting technologies for accurate scaffold production show significant potential. Furthermore, integrating bioactive compounds and growth factors into hydrogel matrices to promote tissue regeneration is critical for enhancing therapeutic results.
Collapse
Affiliation(s)
| | - Seong-Cheol Kim
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea
| |
Collapse
|
5
|
Balahura (Stămat) LR, Dinu AI, Lungu A, Herman H, Balta C, Hermenean A, Șerban AI, Dinescu S. Implantable Polymer Scaffolds Loaded with Paclitaxel-Cyclodextrin Complexes for Post-Breast Cancer Tissue Reconstruction. Polymers (Basel) 2025; 17:402. [PMID: 39940603 PMCID: PMC11819909 DOI: 10.3390/polym17030402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
The side effects associated with the chemotherapy of triple-negative breast cancer (TNBC), such as nucleotide-binding oligomerization domain (NOD)-like receptor family (NLR), pyrin domain containing 3 (NLRP3) inflammasome activity, are responsible for the treatment failure and high mortality rates. Therefore, advanced delivery systems have been developed to improve the transport and targeted administration of anti-tumor agents at the tumor sites using tissue engineering approaches. Implantable delivery systems based on biodegradable polymers are an effective alternative due high biocompatibility, porosity, and mechanical strength. Moreover, the use of paclitaxel (PTX)-cyclodextrin complexes increases the solubility and permeability of PTX, enhancing the bioavailability and efficacy of the drug. All of these properties contribute to the efficient encapsulation and controlled release of drugs, preventing the damage of healthy tissues. In the current study, we detailed the synthesis process and evaluation of 3D scaffolds based on gelatin functionalized with methacryloyl groups (GelMA) and pectin loaded with PTX-cyclodextrin inclusion complexes on TNBC pathogenesis in vitro and in vivo. Bio-physio-chemical analysis of the proposed scaffolds revealed favorable mechanical and biological properties for the cellular component. To improve the drug solubility, a host-guest interaction was performed by the complexation of PTX with a cyclodextrin derivative prior to scaffold synthesis. The presence of PTX suppressed the growth of breast tumor cells and promoted caspase-1 activity, the release of interleukin (IL)-1β, and the production of reactive oxygen species (ROS), conditioning the expression levels of the genes and proteins associated with breast tumorigenesis and NLRP3 inflammasome. The in vivo experiments suggested the activation of pyroptosis tumor cell death, confirming the in vitro experiments. In conclusion, the bio-mechanical properties of the GelMA and pectin-based scaffolds as well as the addition of the PTX-cyclodextrin complexes allow for the targeted and efficient delivery of PTX, suppressing the viability of the breast tumor cells via pyroptosis cell death initiation.
Collapse
Affiliation(s)
| | - Andreea Ioana Dinu
- Advanced Polymer Materials Group, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (A.I.D.); (A.L.)
| | - Adriana Lungu
- Advanced Polymer Materials Group, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (A.I.D.); (A.L.)
| | - Hildegard Herman
- “Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 310414 Arad, Romania; (H.H.); (C.B.); (A.H.)
| | - Cornel Balta
- “Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 310414 Arad, Romania; (H.H.); (C.B.); (A.H.)
| | - Anca Hermenean
- “Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 310414 Arad, Romania; (H.H.); (C.B.); (A.H.)
| | - Andreea Iren Șerban
- Department Preclinical Sciences, Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 050097 Bucharest, Romania;
| | - Sorina Dinescu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania;
- Research Institute of the University of Bucharest, 050663 Bucharest, Romania
| |
Collapse
|
6
|
Zhu S, Shou X, Kuang G, Kong X, Sun W, Zhang Q, Xia J. Stimuli-responsive hydrogel microspheres encapsulated with tumor-cell-derived microparticles for malignant ascites treatment. Acta Biomater 2025; 192:328-339. [PMID: 39586349 DOI: 10.1016/j.actbio.2024.11.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/04/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
Tumor-cell-derived microparticles (TMPs) have been recognized as chemotherapeutic drug carriers and immunomodulators for anti-tumor therapy. Research in the clinical application of TMPs has been devoted to developing an effective delivery formulation that could enhance their therapeutic effects. Here, we propose thermal-responsive agarose hydrogel microspheres (MTX-TMPs@MSs) with encapsulation of Methotrexate (MTX)-packaging TMPs (MTX-TMPs) and black phosphorus quantum dots (BPQDs) by microfluidic technology for synergistic treatment of malignant ascites. The laden MTX-TMPs, separated from apoptotic tumor cells, could target tumor cells for the delivery of chemotherapy drugs and modulate the tumor immune microenvironment. Under near-infrared (NIR) induced thermal stimulation, MTX-TMPs could be controllably released from the low-melting-point agarose matrix hydrogel microspheres for chemotherapy (CHT) and immunotherapy (IMT). In addition, benefiting from photothermal therapy (PTT)-induced tumor immunogenic death, the anti-tumor immune response triggered by MTX-TMPs was further enhanced. Based on these features, the MTX-TMPs@MSs could remarkably eliminate tumor cells in vitro and obviously suppress tumor growth in vivo through synergistic PTT, CHT, and IMT. Therefore, it is envisaged that this TMPs-integrated microcarrier will have promising applications in clinical tumor therapy. STATEMENT OF SIGNIFICANCE: Primary liver cancer ranks third among the causes of cancer deaths globally, with hepatocellular carcinoma (HCC) being the most common type. In particular, patients with advanced HCC accompanied by malignant ascites, a common complication, indicate tumor metastasis and a poor prognosis. In this paper, we developed stimuli-responsive hydrogel microspheres from microfluidics for the delivery of methotrexate (MTX)-loaded tumor-cell-derived microparticles (MTX-TMPs) for synergistic chemotherapy, photothermal therapy, and immunotherapy. The release of MTX-TMPs from hydrogel microspheres could be on-demand controlled through BPQDs-mediated photothermal stimulus. On the other hand, BPQDs-mediated mild hyperthermia cooperatesss with MTX-TMPs-induced chemotherapy could participate in remodeling the tumor immunosuppressive microenvironment. Thus, the prepared microcarrier system holds great promise for tumor therapy.
Collapse
Affiliation(s)
- Shishi Zhu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Xin Shou
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Gaizhen Kuang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Xiuyan Kong
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Weijian Sun
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | - Qingfei Zhang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China.
| | - Jinglin Xia
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; National Medical Center & National Clinical Research Center for Interventional Medicine. Liver Cancer Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| |
Collapse
|
7
|
Yang J, Wang L, Wu R, He Y, Zhao Y, Wang W, Gao X, Wang D, Zhao L, Li W. 3D Bioprinting in Cancer Modeling and Biomedicine: From Print Categories to Biological Applications. ACS OMEGA 2024; 9:44076-44100. [PMID: 39524656 PMCID: PMC11541486 DOI: 10.1021/acsomega.4c06051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
The continuous interaction between tumor cells and the local microenvironment plays a decisive role in tumor development. Selecting effective models to simulate the tumor microenvironment to study the physiological processes of tumorigenesis and progression is extremely important and challenging. Currently, three-dimensional (3D) bioprinting technology makes it possible to replicate a physiologically relevant tumor microenvironment and induce genomic and proteomic expression to better mimic tumors in vivo. Meanwhile, it plays a crucial role in the prevention and treatment of human diseases, contributing to drug delivery and drug screening, tissue development and regenerative medicine. This paper provides an overview of the categories of 3D bioprinting technology, and the recent advances in the bioinks required for printing. In addition, we summarize the current tumor models based on 3D bioprinting and provide an assessment of possible future biological applications.
Collapse
Affiliation(s)
- Jianye Yang
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, China
| | - Le Wang
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, China
| | - Ruimei Wu
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, China
| | - Yanan He
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, China
| | - Yu Zhao
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, China
| | - Wenchi Wang
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, China
| | - Xiaochen Gao
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, China
| | - Dan Wang
- Department
of Physical Education, School of Foundation Medical, Shandong Second Medical University, Weifang 261053, China
| | - Lidan Zhao
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, China
| | - Wenfang Li
- School
of Life Science and Technology, Shandong
Second Medical University, Weifang 261053, China
| |
Collapse
|
8
|
Zhao J, Zhang H, Liu Y, Lu G, Wang Z, Mo Q, Wang G, Shen Y, Jiao L. HIF-1α knockdown suppresses breast cancer metastasis via epithelial mesenchymal transition Abrogation. Heliyon 2024; 10:e37900. [PMID: 39386828 PMCID: PMC11462230 DOI: 10.1016/j.heliyon.2024.e37900] [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: 06/24/2024] [Revised: 09/12/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
Lung metastasis, a leading cause of breast cancer mortality, lacks effective therapeutic options. Hypoxia-inducible factor 1-alpha (HIF-1α) plays important roles in breast cancer progression, but its direct impact on lung metastasis remains unclear. Herein, in this study, we investigated the role of HIF-1α in breast cancer lung metastasis and the potential of targeting it for therapeutic benefit. HIF-1α expression was knocked down in the 4T1 mouse mammary carcinoma cell line using a lentiviral vector. HIF-1α knockdown significantly reduced the migratory ability of 4T1 cells in vitro and lung metastasis in a mouse model. Mechanistically, HIF-1α knockdown decreased the expression of matrix metalloproteinases (MMP-2 and MMP-9) that degrade the extracellular matrix and suppressed the epithelial-to-mesenchymal transition (EMT) by increasing E-cadherin and decreasing vimentin expression. The findings of this study demonstrate that HIF-1α knockdown effectively inhibits lung metastasis of 4T1 cells both in vitro and in vivo by suppressing EMT. These results underscore a promising new approach for managing breast cancer metastasis.
Collapse
Affiliation(s)
- Jinjin Zhao
- Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453100, China
- Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453100, China
| | - Haiguang Zhang
- Department of Gynecology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453100, China
| | - Yaqian Liu
- Otorhinolaryngology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453100, China
- Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453100, China
| | - Guangjian Lu
- Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453100, China
| | - Zhaohui Wang
- Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453100, China
| | - Qingjiang Mo
- Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453100, China
| | - Guoqiang Wang
- Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453100, China
| | - Yanfei Shen
- Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453100, China
- Medical School, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Luyang Jiao
- Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453100, China
- Department of Blood Transfusion, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453100, China
| |
Collapse
|
9
|
Fan J, Wang M, Wang X. The potential of three-dimensional printed stents in post-operative treatment of breast cancer. BIOMATERIALS TRANSLATIONAL 2024; 5:331-333. [PMID: 39734702 PMCID: PMC11681180 DOI: 10.12336/biomatertransl.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/06/2024] [Accepted: 09/24/2024] [Indexed: 12/31/2024]
Affiliation(s)
- Junjuan Fan
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Min Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, Anhui Province, China
| | - Xianwen Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
10
|
Shu Y, Li B, Ma H, Liu J, Cheng YY, Li X, Liu T, Yang C, Ma X, Song K. Three-dimensional breast cancer tumor models based on natural hydrogels: a review. J Zhejiang Univ Sci B 2024; 25:736-755. [PMID: 39308065 PMCID: PMC11422793 DOI: 10.1631/jzus.b2300840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Breast cancer is the most common cancer in women and one of the deadliest cancers worldwide. According to the distribution of tumor tissue, breast cancer can be divided into invasive and non-invasive forms. The cancer cells in invasive breast cancer pass through the breast and through the immune system or systemic circulation to different parts of the body, forming metastatic breast cancer. Drug resistance and distant metastasis are the main causes of death from breast cancer. Research on breast cancer has attracted extensive attention from researchers. In vitro construction of tumor models by tissue engineering methods is a common tool for studying cancer mechanisms and anticancer drug screening. The tumor microenvironment consists of cancer cells and various types of stromal cells, including fibroblasts, endothelial cells, mesenchymal cells, and immune cells embedded in the extracellular matrix. The extracellular matrix contains fibrin proteins (such as types I, II, III, IV, VI, and X collagen and elastin) and glycoproteins (such as proteoglycan, laminin, and fibronectin), which are involved in cell signaling and binding of growth factors. The current traditional two-dimensional (2D) tumor models are limited by the growth environment and often cannot accurately reproduce the heterogeneity and complexity of tumor tissues in vivo. Therefore, in recent years, research on three-dimensional (3D) tumor models has gradually increased, especially 3D bioprinting models with high precision and repeatability. Compared with a 2D model, the 3D environment can better simulate the complex extracellular matrix components and structures in the tumor microenvironment. Three-dimensional models are often used as a bridge between 2D cellular level experiments and animal experiments. Acellular matrix, gelatin, sodium alginate, and other natural materials are widely used in the construction of tumor models because of their excellent biocompatibility and non-immune rejection. Here, we review various natural scaffold materials and construction methods involved in 3D tissue-engineered tumor models, as a reference for research in the field of breast cancer.
Collapse
Affiliation(s)
- Yan Shu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Bing Li
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Hailin Ma
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Jiaqi Liu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, NSW 2007, Australia
| | - Xiangqin Li
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Tianqing Liu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Chuwei Yang
- Emergency Center, the Second Hospital of Dalian Medical University, Dalian 116023, China. ,
| | - Xiao Ma
- Department of Anesthesia, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China. ,
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China.
| |
Collapse
|
11
|
Liu S, Wang J, Jiang Y, Wang Y, Yang B, Li H, Zhou G. One Stone Several Birds: Self-Localizing Submicrocages With Dual Loading Points for Multifunctional Drug Delivery. Macromol Biosci 2024; 24:e2400033. [PMID: 38642330 DOI: 10.1002/mabi.202400033] [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: 02/01/2024] [Revised: 03/25/2024] [Indexed: 04/22/2024]
Abstract
As the core index, how to improve bioavailability of loaded cargoes is a hot topic of drug carriers. In this study, aminated β-cyclodextrin (β-CD) as a cross-linking points is first integrated into 3D poly(acrylamide-co-acrylonitrile) (P(AAm-co-AN)) network to build up a unique submicrocage (466.2 ± 47.6 nm), featuring upper critical solution temperature (UCST; ≈40 °C), high volume expansion coefficient, and excellent biocompatibility. Hereinto, hydrophobic β-elemene (ELE) is locally loaded in β-CD with high loading efficiency (8.72%) and encapsulation efficiency (78.60%) through hydrophobic desolvation and host-guest interaction. Above UCST, the release of the loaded ELE is accelerated to 72.87% in 24 h, together with the enhanced sensitization effect of synergized radiotherapy. Given spontaneous long-lasting delivery, targeted embolization, and post-treatment removal of such UCST-type submicrocage, it is anticipated to provide a novel, facile, efficient, and versatile strategy of comprehensive anticancer treatments for high drug bioavailability.
Collapse
Affiliation(s)
- Shuxuan Liu
- Guangdong Provincial Key Laboratory of Optical Information Materials and Technology, Institute of Electronic Paper Displays, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou, 510006, P. R. China
- National Center for International Research on Green Optoelectronics, South China Normal University, Guangzhou, 510006, P. R. China
| | - Jifei Wang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Yong Jiang
- The Fourth Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Yao Wang
- Guangdong Provincial Key Laboratory of Optical Information Materials and Technology, Institute of Electronic Paper Displays, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou, 510006, P. R. China
- National Center for International Research on Green Optoelectronics, South China Normal University, Guangzhou, 510006, P. R. China
| | - Bin Yang
- The Fourth Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Hao Li
- Guangdong Provincial Key Laboratory of Optical Information Materials and Technology, Institute of Electronic Paper Displays, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou, 510006, P. R. China
- National Center for International Research on Green Optoelectronics, South China Normal University, Guangzhou, 510006, P. R. China
| | - Guofu Zhou
- Guangdong Provincial Key Laboratory of Optical Information Materials and Technology, Institute of Electronic Paper Displays, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou, 510006, P. R. China
- National Center for International Research on Green Optoelectronics, South China Normal University, Guangzhou, 510006, P. R. China
| |
Collapse
|
12
|
Gao X, Caruso BR, Li W. Advanced Hydrogels in Breast Cancer Therapy. Gels 2024; 10:479. [PMID: 39057502 PMCID: PMC11276203 DOI: 10.3390/gels10070479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Breast cancer is the most common malignancy among women and is the second leading cause of cancer-related death for women. Depending on the tumor grade and stage, breast cancer is primarily treated with surgery and antineoplastic therapy. Direct or indirect side effects, emotional trauma, and unpredictable outcomes accompany these traditional therapies, calling for therapies that could improve the overall treatment and recovery experiences of patients. Hydrogels, biomimetic materials with 3D network structures, have shown great promise for augmenting breast cancer therapy. Hydrogel implants can be made with adipogenic and angiogenic properties for tissue integration. 3D organoids of malignant breast tumors grown in hydrogels retain the physical and genetic characteristics of the native tumors, allowing for post-surgery recapitulation of the diseased tissues for precision medicine assessment of the responsiveness of patient-specific cancers to antineoplastic treatment. Hydrogels can also be used as carrier matrices for delivering chemotherapeutics and immunotherapeutics or as post-surgery prosthetic scaffolds. The hydrogel delivery systems could achieve localized and controlled medication release targeting the tumor site, enhancing efficacy and minimizing the adverse effects of therapeutic agents delivered by traditional procedures. This review aims to summarize the most recent advancements in hydrogel utilization for breast cancer post-surgery tissue reconstruction, tumor modeling, and therapy and discuss their limitations in clinical translation.
Collapse
Affiliation(s)
- Xiangyu Gao
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
- Doctor of Medicine Program, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA;
| | - Benjamin R. Caruso
- Doctor of Medicine Program, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA;
| | - Weimin Li
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| |
Collapse
|
13
|
Angelopoulou A. Nanostructured Biomaterials in 3D Tumor Tissue Engineering Scaffolds: Regenerative Medicine and Immunotherapies. Int J Mol Sci 2024; 25:5414. [PMID: 38791452 PMCID: PMC11121067 DOI: 10.3390/ijms25105414] [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: 04/28/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
The evaluation of nanostructured biomaterials and medicines is associated with 2D cultures that provide insight into biological mechanisms at the molecular level, while critical aspects of the tumor microenvironment (TME) are provided by the study of animal xenograft models. More realistic models that can histologically reproduce human tumors are provided by tissue engineering methods of co-culturing cells of varied phenotypes to provide 3D tumor spheroids that recapitulate the dynamic TME in 3D matrices. The novel approaches of creating 3D tumor models are combined with tumor tissue engineering (TTE) scaffolds including hydrogels, bioprinted materials, decellularized tissues, fibrous and nanostructured matrices. This review focuses on the use of nanostructured materials in cancer therapy and regeneration, and the development of realistic models for studying TME molecular and immune characteristics. Tissue regeneration is an important aspect of TTE scaffolds used for restoring the normal function of the tissues, while providing cancer treatment. Thus, this article reports recent advancements in the development of 3D TTE models for antitumor drug screening, studying tumor metastasis, and tissue regeneration. Also, this review identifies the significant opportunities of using 3D TTE scaffolds in the evaluation of the immunological mechanisms and processes involved in the application of immunotherapies.
Collapse
Affiliation(s)
- Athina Angelopoulou
- Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece
| |
Collapse
|
14
|
Zhou M, Lin X, Wang L, Yang C, Yu Y, Zhang Q. Preparation and Application of Hemostatic Hydrogels. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309485. [PMID: 38102098 DOI: 10.1002/smll.202309485] [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: 10/19/2023] [Revised: 11/28/2023] [Indexed: 12/17/2023]
Abstract
Hemorrhage remains a critical challenge in various medical settings, necessitating the development of advanced hemostatic materials. Hemostatic hydrogels have emerged as promising solutions to address uncontrolled bleeding due to their unique properties, including biocompatibility, tunable physical characteristics, and exceptional hemostatic capabilities. In this review, a comprehensive overview of the preparation and biomedical applications of hemostatic hydrogels is provided. Particularly, hemostatic hydrogels with various materials and forms are introduced. Additionally, the applications of hemostatic hydrogels in trauma management, surgical procedures, wound care, etc. are summarized. Finally, the limitations and future prospects of hemostatic hydrogels are discussed and evaluated. This review aims to highlight the biomedical applications of hydrogels in hemorrhage management and offer insights into the development of clinically relevant hemostatic materials.
Collapse
Affiliation(s)
- Minyu Zhou
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiang Lin
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Li Wang
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Chaoyu Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Yunru Yu
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Qingfei Zhang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| |
Collapse
|
15
|
Mozafari N, Jahanbekam S, Ashrafi H, Shahbazi MA, Azadi A. Recent Biomaterial-Assisted Approaches for Immunotherapeutic Inhibition of Cancer Recurrence. ACS Biomater Sci Eng 2024; 10:1207-1234. [PMID: 38416058 DOI: 10.1021/acsbiomaterials.3c01347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Biomaterials possess distinctive properties, notably their ability to encapsulate active biological products while providing biocompatible support. The immune system plays a vital role in preventing cancer recurrence, and there is considerable demand for an effective strategy to prevent cancer recurrence, necessitating effective strategies to address this concern. This review elucidates crucial cellular signaling pathways in cancer recurrence. Furthermore, it underscores the potential of biomaterial-based tools in averting or inhibiting cancer recurrence by modulating the immune system. Diverse biomaterials, including hydrogels, particles, films, microneedles, etc., exhibit promising capabilities in mitigating cancer recurrence. These materials are compelling candidates for cancer immunotherapy, offering in situ immunostimulatory activity through transdermal, implantable, and injectable devices. They function by reshaping the tumor microenvironment and impeding tumor growth by reducing immunosuppression. Biomaterials facilitate alterations in biodistribution, release kinetics, and colocalization of immunostimulatory agents, enhancing the safety and efficacy of therapy. Additionally, how the method addresses the limitations of other therapeutic approaches is discussed.
Collapse
Affiliation(s)
- Negin Mozafari
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| | - Sheida Jahanbekam
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| | - Hajar Ashrafi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Amir Azadi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| |
Collapse
|
16
|
Myung N, Kang HW. Local dose-dense chemotherapy for triple-negative breast cancer via minimally invasive implantation of 3D printed devices. Asian J Pharm Sci 2024; 19:100884. [PMID: 38357526 PMCID: PMC10861843 DOI: 10.1016/j.ajps.2024.100884] [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: 07/18/2023] [Revised: 10/18/2023] [Accepted: 11/18/2023] [Indexed: 02/16/2024] Open
Abstract
Dose-dense chemotherapy is the preferred first-line therapy for triple-negative breast cancer (TNBC), a highly aggressive disease with a poor prognosis. This treatment uses the same drug doses as conventional chemotherapy but with shorter dosing intervals, allowing for promising clinical outcomes with intensive treatment. However, the frequent systemic administration used for this treatment results in systemic toxicity and low patient compliance, limiting therapeutic efficacy and clinical benefit. Here, we report local dose-dense chemotherapy to treat TNBC by implanting 3D printed devices with time-programmed pulsatile release profiles. The implantable device can control the time between drug releases based on its internal microstructure design, which can be used to control dose density. The device is made of biodegradable materials for clinical convenience and designed for minimally invasive implantation via a trocar. Dose density variation of local chemotherapy using programmable release enhances anti-cancer effects in vitro and in vivo. Under the same dose density conditions, device-based chemotherapy shows a higher anti-cancer effect and less toxic response than intratumoral injection. We demonstrate local chemotherapy utilizing the implantable device that simulates the drug dose, number of releases, and treatment duration of the dose-dense AC (doxorubicin and cyclophosphamide) regimen preferred for TNBC treatment. Dose density modulation inhibits tumor growth, metastasis, and the expression of drug resistance-related proteins, including p-glycoprotein and breast cancer resistance protein. To the best of our knowledge, local dose-dense chemotherapy has not been reported, and our strategy can be expected to be utilized as a novel alternative to conventional therapies and improve anti-cancer efficiency.
Collapse
Affiliation(s)
- Noehyun Myung
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulju-gun 44919, South Korea
| | - Hyun-Wook Kang
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulju-gun 44919, South Korea
| |
Collapse
|
17
|
Su Y, Liu Y, Hu X, Lu Y, Zhang J, Jin W, Liu W, Shu Y, Cheng YY, Li W, Nie Y, Pan B, Song K. Caffeic acid-grafted chitosan/sodium alginate/nanoclay-based multifunctional 3D-printed hybrid scaffolds for local drug release therapy after breast cancer surgery. Carbohydr Polym 2024; 324:121441. [PMID: 37985071 DOI: 10.1016/j.carbpol.2023.121441] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/02/2023] [Accepted: 09/26/2023] [Indexed: 11/22/2023]
Abstract
Breast cancer is one of the most common malignant tumors in women all over the world. Mastectomy is the most effective treatment, but there are serious problems such as high tumor recurrence rate and side effects of chemotherapy. Therefore, there is an urgent need for a therapeutic strategy that can effectively promote postoperative wound healing and inhibit local tumor recurrence. In this study, a 3D printing scaffold based on carbon dots-curcumin nano-drug release (CCNPs) was developed as a local drug delivery platform (named CCNACA using CCNPs, Sodium alginate, Nanoclay and Caffeic Acid grafted Chitosan as raw materials), which has the ability to visualize drug release. The 14-day drug release test in vitro showed that the tumor inhibition rate of CCNACA scaffolds on breast cancer cells (MCF-7) was 73.77 ± 1.68 %. And the CCNACA scaffolds had good long-term antibacterial (Escherichia coli and Staphylococcus aureus) activity. Animal experiments have shown that implanting CCNACA scaffolds into surgical defects can inhibit postoperative residual cancer cells, reduce inflammation, promote angiogenesis, and repair tissue defects caused by surgery. In summary, the local drug delivery system of this manuscript has great potential in wound healing and prevention of tumor recurrence after breast cancer surgery.
Collapse
Affiliation(s)
- Ya Su
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yaqian Liu
- Department of Breast Surgery, The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Shahekou District, Dalian, Liaoning 116023, China
| | - Xueyan Hu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yueqi Lu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China; Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou 450000, China
| | - Jinyuan Zhang
- Department of Breast Surgery, The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Shahekou District, Dalian, Liaoning 116023, China
| | - Wenbo Jin
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Wang Liu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yan Shu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, NSW 2007, Australia
| | - Wenfang Li
- School of Life Science and Technology, Weifang Medical University, Weifang 261053, China.
| | - Yi Nie
- Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou 450000, China.
| | - Bo Pan
- Department of Breast Surgery, The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Shahekou District, Dalian, Liaoning 116023, China.
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China; Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou 450000, China.
| |
Collapse
|
18
|
Eluu SC, Obayemi JD, Salifu AA, Yiporo D, Oko AO, Aina T, Oparah JC, Ezeala CC, Etinosa PO, Ugwu CM, Esimone CO, Soboyejo WO. In-vivo studies of targeted and localized cancer drug release from microporous poly-di-methyl-siloxane (PDMS) devices for the treatment of triple negative breast cancer. Sci Rep 2024; 14:31. [PMID: 38167999 PMCID: PMC10761815 DOI: 10.1038/s41598-023-50656-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024] Open
Abstract
Triple-negative breast cancer (TNBC) treatment is challenging and frequently characterized by an aggressive phenotype and low prognosis in comparison to other subtypes. This paper presents fabricated implantable drug-loaded microporous poly-di-methyl-siloxane (PDMS) devices for the delivery of targeted therapeutic agents [Luteinizing Hormone-Releasing Hormone conjugated paclitaxel (PTX-LHRH) and Luteinizing Hormone-Releasing Hormone conjugated prodigiosin (PG-LHRH)] for the treatment and possible prevention of triple-negative cancer recurrence. In vitro assessment using the Alamar blue assay demonstrated a significant reduction (p < 0.05) in percentage of cell growth in a time-dependent manner in the groups treated with PG, PG-LHRH, PTX, and PTX-LHRH. Subcutaneous triple-negative xenograft breast tumors were then induced in athymic female nude mice that were four weeks old. Two weeks later, the tumors were surgically but partially removed, and the device implanted. Mice were observed for tumor regrowth and organ toxicity. The animal study revealed that there was no tumor regrowth, six weeks post-treatment, when the LHRH targeted drugs (LHRH-PTX and LHRH-PGS) were used for the treatment. The possible cytotoxic effects of the released drugs on the liver, kidney, and lung are assessed using quantitative biochemical assay from blood samples of the treatment groups. Ex vivo histopathological results from organ tissues showed that the targeted cancer drugs released from the implantable drug-loaded device did not induce any adverse effect on the liver, kidneys, or lungs, based on the results of qualitative toxicity studies. The implications of the results are discussed for the targeted and localized treatment of triple negative breast cancer.
Collapse
Affiliation(s)
- S C Eluu
- Department of Pharmaceutical Microbiology and Biotechnology, Nnamdi Azikiwe University, Ifite Awka, 420110, Anambra State, Nigeria
| | - J D Obayemi
- Department of Mechanical Engineering, Higgins Lab, Worcester Polytechnic Institute (WPI), 100 Institute Road, Worcester, MA, 01609, USA
- Department of Biomedical Engineering, Gateway Park Life Sciences and Bioengineering Centre, Worcester Polytechnic Institute, 60 Prescott Street, Worcester, MA, 01609, USA
| | - A A Salifu
- Department of Engineering, Morrissey College of Arts and Science, Boston College, Boston, USA
| | - D Yiporo
- Department of Mechanical Engineering, Ashesi University, Berekuso, Ghana
| | - A O Oko
- Department of Biology and Biotechnology, David Umahi Federal, University of Health Sciences, Uburu, Nigeria
| | - T Aina
- Department of Material Science, African University of Science and Technology, Km 10 Airport Road, Abuja, Nigeria
| | - J C Oparah
- Department of Material Science, African University of Science and Technology, Km 10 Airport Road, Abuja, Nigeria
| | - C C Ezeala
- Department of Material Science, African University of Science and Technology, Km 10 Airport Road, Abuja, Nigeria
| | - P O Etinosa
- Department of Mechanical Engineering, Higgins Lab, Worcester Polytechnic Institute (WPI), 100 Institute Road, Worcester, MA, 01609, USA
| | - C M Ugwu
- Department of Pharmaceutical Microbiology and Biotechnology, Nnamdi Azikiwe University, Ifite Awka, 420110, Anambra State, Nigeria
| | - C O Esimone
- Department of Pharmaceutical Microbiology and Biotechnology, Nnamdi Azikiwe University, Ifite Awka, 420110, Anambra State, Nigeria
| | - W O Soboyejo
- Department of Mechanical Engineering, Higgins Lab, Worcester Polytechnic Institute (WPI), 100 Institute Road, Worcester, MA, 01609, USA.
- Department of Biomedical Engineering, Gateway Park Life Sciences and Bioengineering Centre, Worcester Polytechnic Institute, 60 Prescott Street, Worcester, MA, 01609, USA.
- Department of Engineering, SUNY Polytechnic Institute, 100 Seymour Rd, Utica, NY, 13502, USA.
| |
Collapse
|
19
|
Orbach SM, DeVaull CY, Bealer EJ, Ross BC, Jeruss JS, Shea LD. An engineered niche delineates metastatic potential of breast cancer. Bioeng Transl Med 2024; 9:e10606. [PMID: 38193115 PMCID: PMC10771563 DOI: 10.1002/btm2.10606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/29/2023] [Accepted: 09/20/2023] [Indexed: 01/10/2024] Open
Abstract
Metastatic breast cancer is often not diagnosed until secondary tumors have become macroscopically visible and millions of tumor cells have invaded distant tissues. Yet, metastasis is initiated by a cascade of events leading to formation of the pre-metastatic niche, which can precede tumor formation by a matter of years. We aimed to distinguish the potential for metastatic disease from nonmetastatic disease at early times in triple-negative breast cancer using sister cell lines 4T1 (metastatic), 4T07 (invasive, nonmetastatic), and 67NR (nonmetastatic). We used a porous, polycaprolactone scaffold, that serves as an engineered metastatic niche, to identify metastatic disease through the characteristics of the microenvironment. Analysis of the immune cell composition at the scaffold was able to distinguish noninvasive 67NR tumor-bearing mice from 4T07 and 4T1 tumor-bearing mice but could not delineate metastatic potential between the two invasive cell lines. Gene expression in the scaffolds correlated with the up-regulation of cancer hallmarks (e.g., angiogenesis, hypoxia) in the 4T1 mice relative to 4T07 mice. We developed a 9-gene signature (Dhx9, Dusp12, Fth1, Ifitm1, Ndufs1, Pja2, Slc1a3, Soga1, Spon2) that successfully distinguished 4T1 disease from 67NR or 4T07 disease throughout metastatic progression. Furthermore, this signature proved highly effective at distinguishing diseased lungs in publicly available datasets of mouse models of metastatic breast cancer and in human models of lung cancer. The early and accurate detection of metastatic disease that could lead to early treatment has the potential to improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Sophia M. Orbach
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | | | - Elizabeth J. Bealer
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Brian C. Ross
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Jacqueline S. Jeruss
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of PathologyUniversity of MichiganAnn ArborMichiganUSA
- Department of SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Lonnie D. Shea
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of Chemical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
20
|
Lu Z, Miao X, Song Q, Ding H, Rajan SAP, Skardal A, Votanopoulos KI, Dai K, Zhao W, Lu B, Atala A. Detection of lineage-reprogramming efficiency of tumor cells in a 3D-printed liver-on-a-chip model. Theranostics 2023; 13:4905-4918. [PMID: 37771785 PMCID: PMC10526656 DOI: 10.7150/thno.86921] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/04/2023] [Indexed: 09/30/2023] Open
Abstract
Background: The liver metastasis accompanied with the loss of liver function is one of the most common complications in patients with triple-negative breast cancers (TNBC). Lineage reprogramming, as a technique direct inducing the functional cell types from one lineage to another lineage without passing through an intermediate pluripotent stage, is promising in changing cell fates and overcoming the limitations of primary cells. However, most reprogramming techniques are derived from human fibroblasts, and whether cancer cells can be reversed into hepatocytes remains elusive. Methods: Herein, we simplify preparation of reprogramming reagents by expressing six transcriptional factors (HNF4A, FOXA2, FOXA3, ATF5, PROX1, and HNF1) from two lentiviral vectors, each expressing three factors. Then the virus was transduced into MDA-MB-231 cells to generated human induced hepatocyte-like cells (hiHeps) and single-cell sequencing was used to analyze the fate for the cells after reprogramming. Furthermore, we constructed a Liver-on-a-chip (LOC) model by bioprinting the Gelatin Methacryloyl hydrogel loaded with hepatocyte extracellular vesicles (GelMA-EV) bioink onto the microfluidic chip to assess the metastasis behavior of the reprogrammed TNBC cells under the 3D liver microenvironment in vitro. Results: The combination of the genes HNF4A, FOXA2, FOXA3, ATF5, PROX1 and HNF1A could reprogram MDA-MB-231 tumor cells into human-induced hepatocytes (hiHeps), limiting metastasis of these cells. Single-cell sequencing analysis showed that the oncogenes were significantly inhibited while the liver-specific genes were activated after lineage reprogramming. Finally, the constructed LOC model showed that the hepatic phenotypes of the reprogrammed cells could be observed, and the metastasis of embedded cancer cells could be inhibited under the liver microenvironment. Conclusion: Our findings demonstrate that reprogramming could be a promising method to produce hepatocytes and treat TNBC liver metastasis. And the LOC model could intimate the 3D liver microenvironment and assess the behavior of the reprogrammed TNBC cells.
Collapse
Affiliation(s)
- Zuyan Lu
- Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Xiangwan Miao
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Qianqian Song
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Huifen Ding
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Shiny Amala Priya Rajan
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Aleksander Skardal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | | | - Kerong Dai
- Department of Orthopaedic Surgery, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Weixin Zhao
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Baisong Lu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| |
Collapse
|
21
|
Dai CL, Zhang RJ, An P, Deng YQ, Rahman K, Zhang H. Cinobufagin: a promising therapeutic agent for cancer. J Pharm Pharmacol 2023; 75:1141-1153. [PMID: 37390473 DOI: 10.1093/jpp/rgad059] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/12/2023] [Indexed: 07/02/2023]
Abstract
OBJECTIVES Cinobufagin is a natural active ingredient isolated from the traditional Chinese medicine Venenum Bufonis (Chinese: Chansu), which is the dried secretion of the postauricular gland or skin gland of the Bufo gargarizans Cantor or Bufo melanostictus Schneider. There is increasing evidence indicating that cinobufagin plays an important role in the treatment of cancer. This article is to review and discuss the antitumor pharmacological effects and mechanisms of cinobufagin, along with a description of its toxicity and pharmacokinetics. METHODS The public databases including PubMed, China National Knowledge Infrastructure and Elsevier were referenced, and 'cinobufagin', 'Chansu', 'Venenum Bufonis', 'anticancer', 'cancer', 'carcinoma', and 'apoptosis' were used as keywords to summarize the comprehensive research and applications of cinobufagin published up to date. KEY FINDINGS Cinobufagin can induce tumour cell apoptosis and cycle arrest, inhibit tumour cell proliferation, migration, invasion and autophagy, reduce angiogenesis and reverse tumour cell multidrug resistance, through triggering DNA damage and activating the mitochondrial pathway and the death receptor pathway. CONCLUSIONS Cinobufagin has the potential to be further developed as a new drug against cancer.
Collapse
Affiliation(s)
- Chun-Lan Dai
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Run-Jing Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pei An
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi-Qing Deng
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Khalid Rahman
- School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, UK
| | - Hong Zhang
- College of Life Sciences, Huaibei Normal University, Huaibei, Anhui, China
| |
Collapse
|
22
|
Jia X, Hua C, Yang F, Li X, Zhao P, Zhou F, Lu Y, Liang H, Xing M, Lyu G. Hydrophobic aerogel-modified hemostatic gauze with thermal management performance. Bioact Mater 2023; 26:142-158. [PMID: 36911208 PMCID: PMC9996136 DOI: 10.1016/j.bioactmat.2023.02.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Current hemostatic agents or dressings are not efficient under extremely hot and cold environments due to deterioration of active ingredients, water evaporation and ice crystal growth. To address these challenges, we engineered a biocompatible hemostatic system with thermoregulatory properties for harsh conditions by combining the asymmetric wetting nano-silica aerogel coated-gauze (AWNSA@G) with a layer-by-layer (LBL) structure. Our AWNSA@G was a dressing with a tunable wettability prepared by spraying the hydrophobic nano-silica aerogel onto the gauze from different distances. The hemostatic time and blood loss of the AWNSA@G were 5.1 and 6.9 times lower than normal gauze in rat's injured femoral artery model. Moreover, the modified gauze was torn off after hemostasis without rebleeding, approximately 23.8 times of peak peeling force lower than normal gauze. For the LBL structure, consisting of the nano-silica aerogel layer and a n-octadecane phase change material layer, in both hot (70 °C) and cold (-27 °C) environments, exhibited dual-functional thermal management and maintained a stable internal temperature. We further verified our composite presented superior blood coagulation effect in extreme environments due to the LBL structure, the pro-coagulant properties of nano-silica aerogel and unidirectional fluid pumping of AWNSA@G. Our work, therefore, shows great hemostasis potential under normal and extreme temperature environments.
Collapse
Affiliation(s)
- Xiaoli Jia
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China.,Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China.,Department of Mechanical Engineering, University of Manitoba, Winnipeg, R3T 2N2, Canada
| | - Chao Hua
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China.,Medical School of Nantong University, Nantong, 226019, China
| | - Fengbo Yang
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China.,Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Xiaoxiao Li
- Nanjing University of Traditional Chinese Medicine, Nanjing, 210023, China
| | - Peng Zhao
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China
| | - Feifan Zhou
- Medical School of Nantong University, Nantong, 226019, China
| | - Yichi Lu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Hao Liang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, R3T 2N2, Canada
| | - Guozhong Lyu
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China.,Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China.,Medical School of Nantong University, Nantong, 226019, China.,Nanjing University of Traditional Chinese Medicine, Nanjing, 210023, China
| |
Collapse
|
23
|
Chinnakorn A, Nuansing W, Bodaghi M, Rolfe B, Zolfagharian A. Recent progress of 4D printing in cancer therapeutics studies. SLAS Technol 2023; 28:127-141. [PMID: 36804175 DOI: 10.1016/j.slast.2023.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023]
Abstract
Cancer is a critical cause of global human death. Not only are complex approaches to cancer prognosis, accurate diagnosis, and efficient therapeutics concerned, but post-treatments like postsurgical or chemotherapeutical effects are also followed up. The four-dimensional (4D) printing technique has gained attention for its potential applications in cancer therapeutics. It is the next generation of the three-dimensional (3D) printing technique, which facilitates the advanced fabrication of dynamic constructs like programmable shapes, controllable locomotion, and on-demand functions. As is well-known, it is still in the initial stage of cancer applications and requires the insight study of 4D printing. Herein, we present the first effort to report on 4D printing technology in cancer therapeutics. This review will illustrate the mechanisms used to induce the dynamic constructs of 4D printing in cancer management. The recent potential applications of 4D printing in cancer therapeutics will be further detailed, and future perspectives and conclusions will finally be proposed.
Collapse
Affiliation(s)
- Atchara Chinnakorn
- School of Physics, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Wiwat Nuansing
- School of Physics, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Mahdi Bodaghi
- Department of Engineering, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom
| | - Bernard Rolfe
- School of Engineering, Deakin University, Geelong, Victoria 3216, Australia
| | - Ali Zolfagharian
- School of Engineering, Deakin University, Geelong, Victoria 3216, Australia.
| |
Collapse
|
24
|
Belluomo R, Khodaei A, Amin Yavari S. Additively manufactured Bi-functionalized bioceramics for reconstruction of bone tumor defects. Acta Biomater 2023; 156:234-249. [PMID: 36028198 DOI: 10.1016/j.actbio.2022.08.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 02/08/2023]
Abstract
Bone tissue exhibits critical factors for metastatic cancer cells and represents an extremely pleasant spot for further growth of tumors. The number of metastatic bone lesions and primary tumors that arise directly from cells comprised in the bone milieu is constantly increasing. Bioceramics have recently received significant attention in bone tissue engineering and local drug delivery applications. Additionally, additive manufacturing of bioceramics offers unprecedented advantages including the possibilities to fill irregular voids after the resection and fabricate patient-specific implants. Herein, we investigated the recent advances in additively manufactured bioceramics and ceramic-based composites that were used in the local bone tumor treatment and reconstruction of bone tumor defects. Furthermore, it has been extensively explained how to bi-functionalize ceramics-based biomaterials and what current limitations impede their clinical application. We have also discussed the importance of further development into ceramic-based biomaterials and molecular biology of bone tumors to: (1) discover new potential therapeutic targets to enhance conventional therapies, (2) local delivering of bio-molecular agents in a customized and "smart" way, and (3) accomplish a complete elimination of tumor cells in order to prevent tumor recurrence formation. We emphasized that by developing the research focus on the introduction of novel 3D-printed bioceramics with unique properties such as stimuli responsiveness, it will be possible to fabricate smart bioceramics that promote bone regeneration while minimizing the side-effects and effectively eradicate bone tumors while promoting bone regeneration. In fact, by combining all these therapeutic strategies and additive manufacturing, it is likely to provide personalized tumor-targeting therapies for cancer patients in the foreseeable future. STATEMENT OF SIGNIFICANCE: To increase the survival rates of cancer patients, different strategies such as surgery, reconstruction, chemotherapy, radiotherapy, etc have proven to be essential. Nonetheless, these therapeutic protocols have reached a plateau in their effectiveness due to limitations including drug resistance, tumor recurrence after surgery, toxic side-effects, and impaired bone regeneration following tumor resection. Hence, novel approaches to specifically and locally attack cancer cells, while also regenerating the damaged bony tissue, have being developed in the past years. This review sheds light to the novel approaches that enhance local bone tumor therapy and reconstruction procedures by combining additive manufacturing of ceramic biomaterials and other polymers, bioactive molecules, nanoparticles to affect bone tumor functions, metabolism, and microenvironment.
Collapse
Affiliation(s)
- Ruggero Belluomo
- Department of Orthopedics, University Medical Center Utrecht, Utrecht 3508GA, the Netherlands
| | - Azin Khodaei
- Department of Orthopedics, University Medical Center Utrecht, Utrecht 3508GA, the Netherlands
| | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, Utrecht 3508GA, the Netherlands; Regenerative Medicine Utrecht, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
25
|
Pei J, Zhang S, Yang X, Han C, Pan Y, Li J, Wang Z, Sun C, Zhang J. Epigenetic regulator KDM4A activates Notch1-NICD-dependent signaling to drive tumorigenesis and metastasis in breast cancer. Transl Oncol 2022; 28:101615. [PMID: 36592610 PMCID: PMC9816809 DOI: 10.1016/j.tranon.2022.101615] [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: 04/15/2022] [Revised: 10/18/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Altered epigenetic reprogramming and events contribute to breast cancer (Bca) progression and metastasis. How the epigenetic histone demethylases modulate breast cancer progression remains poorly defined. We aimed to elucidate the biological roles of KDM4A in driving Notch1 activation and Bca progression. METHODS The KDM4A expression in Bca specimens was analyzed using quantitative PCR and immunohistochemical assays. The biological roles of KDM4A were evaluated using wound-healing assays and an in vivo metastasis model. The Chromatin Immunoprecipitation (ChIP)-qPCR assay was used to determine the role of KDM4A in Notch1 regulation. RESULTS Here, we screened that targeting KDM4A could induce notable cell growth suppression. KDM4A is required for the growth and progression of Bca cells. High KDM4A enhances tumor migration abilities and in vivo lung metastasis. Bioinformatic analysis suggested that KDM4A was highly expressed in tumors and high KDM4A correlates with poor survival outcomes. KDM4A activates Notch1 expressions via directly binding to the promoters and demethylating H3K9me3 modifications. KDM4A inhibition reduces expressions of a list of Notch1 downstream targets, and ectopic expressions of ICN1 could restore the corresponding levels. KDM4A relies on Notch1 signaling to maintain cell growth, migration and self-renewal capacities. Lastly, we divided a panel of cell lines into KDM4Ahigh and KDM4Alow groups. Targeting Notch1 using specific LY3039478 could efficiently suppress cell growth and colony formation abilities of KDM4Ahigh Bca. CONCLUSION Taken together, KDM4A could drive Bca progression via triggering the activation of Notch1 pathway by decreasing H3K9me3 levels, highlighting a promising therapeutic target for Bca.
Collapse
Affiliation(s)
- Jing Pei
- Department of Breast Surgery, First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, Anhui 230022, PR China,Corresponding authors.
| | - ShengQuan Zhang
- Department of Biochemistry and Molecular Biology, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, PR China
| | - Xiaowei Yang
- Department of Breast Surgery, First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, Anhui 230022, PR China
| | - Chunguang Han
- Department of Breast Surgery, First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, Anhui 230022, PR China
| | - Yubo Pan
- Department of Breast Surgery, First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, Anhui 230022, PR China
| | - Jun Li
- Department of Breast Surgery, First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, Anhui 230022, PR China
| | - Zhaorui Wang
- Department of Breast Surgery, First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, Anhui 230022, PR China
| | - Chenyu Sun
- AMITA Health Saint Joseph Hospital Chicago, 2900 N. Lake Shore Drive, Chicago, Illinois 60657, USA
| | - Jing Zhang
- The Department of Breast Surgery, The Tumor Hospital of XuZhou, 131 HuanCheng Road, XuZhou, Jiangsu 221003, PR China,Corresponding authors.
| |
Collapse
|
26
|
Zhang Q, Wang X, Kuang G, Zhao Y. Pt(IV) prodrug initiated microparticles from microfluidics for tumor chemo-, photothermal and photodynamic combination therapy. Bioact Mater 2022; 24:185-196. [PMID: 36606251 PMCID: PMC9804016 DOI: 10.1016/j.bioactmat.2022.12.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/09/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Multimodal treatment modalities hold great potential for cancer therapy, thus current efforts are focusing on the development of more effective and practical synergistic therapeutic platforms. Herein, we present a novel trans, trans,trans-[Pt(N3)2(OH)2(py)2] (Pt(IV)) prodrug-initiated hydrogel microparticles (MICG-Pt) with indocyanine green (ICG) encapsulation by microfluidics for efficiently synergistic chemo-, photothermal (PTT) and photodynamic therapy (PDT). The employed Pt(IV) could not only serves as an initiator to generate azidyl radical (N3 •) for photo-polymerization of methacrylate gelatin (GelMA) matrix, but also be reduced to high cytotoxic platinum(II) (Pt(II)) species for tumor chemotherapy. The laden ICG with highly photothermal heating ability and intrinsic reactive oxygen species (ROS) productivity endows the MICG-Pt with effective PTT/PDT performances upon near-infrared (NIR) light irradiation. In addition, benefiting from the production of oxygen during the photo-activation process of Pt(IV), the PDT efficacy of ICG-laden MICG-Pt could be further enhanced. Based on these advantages, we have demonstrated that the MICG-Pt could significantly eliminate cancer cells in vitro, and remarkably suppressed the tumor growth in vivo via synergistic chemotherapy, PTT, and PDT. These results indicate that such Pt(IV)-initiated hydrogel microparticles are ideal candidates of multimodal treatment platforms, holding great prospects for cancer therapy.
Collapse
Affiliation(s)
- Qingfei Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Xiaocheng Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Gaizhen Kuang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China,Corresponding author. Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
27
|
Mahmudi H, Adili-Aghdam MA, Shahpouri M, Jaymand M, Amoozgar Z, Jahanban-Esfahlan R. Tumor microenvironment penetrating chitosan nanoparticles for elimination of cancer relapse and minimal residual disease. Front Oncol 2022; 12:1054029. [PMID: 36531004 PMCID: PMC9751059 DOI: 10.3389/fonc.2022.1054029] [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: 09/26/2022] [Accepted: 11/09/2022] [Indexed: 10/17/2023] Open
Abstract
Chitosan and its derivatives are among biomaterials with numerous medical applications, especially in cancer. Chitosan is amenable to forming innumerable shapes such as micelles, niosomes, hydrogels, nanoparticles, and scaffolds, among others. Chitosan derivatives can also bring unprecedented potential to cross numerous biological barriers. Combined with other biomaterials, hybrid and multitasking chitosan-based systems can be realized for many applications. These include controlled drug release, targeted drug delivery, post-surgery implants (immunovaccines), theranostics, biosensing of tumor-derived circulating materials, multimodal systems, and combination therapy platforms with the potential to eliminate bulk tumors as well as lingering tumor cells to treat minimal residual disease (MRD) and recurrent cancer. We first introduce different formats, derivatives, and properties of chitosan. Next, given the barriers to therapeutic efficacy in solid tumors, we review advanced formulations of chitosan modules as efficient drug delivery systems to overcome tumor heterogeneity, multi-drug resistance, MRD, and metastasis. Finally, we discuss chitosan NPs for clinical translation and treatment of recurrent cancer and their future perspective.
Collapse
Affiliation(s)
- Hossein Mahmudi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amin Adili-Aghdam
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Shahpouri
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Jaymand
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
28
|
Enzyme-Enhanced Codelivery of Doxorubicin and Bcl-2 Inhibitor by Electrospun Nanofibers for Synergistic Inhibition of Prostate Cancer Recurrence. Pharmaceuticals (Basel) 2022; 15:ph15101244. [PMID: 36297356 PMCID: PMC9610395 DOI: 10.3390/ph15101244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/01/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022] Open
Abstract
One of the great challenges of postoperative prostate cancer management is tumor recurrence. Although postoperative chemotherapy presents benefits to inhibit unexpected recurrence, it is still limited due to the drug resistance or intolerable complications of some patients. Electrospun nanofiber, as a promising drug carrier, demonstrating sustained drug release behavior, can be implanted into the tumor resection site during surgery and is conductive to tumor inhibition. Herein, we fabricated electrospun nanofibers loaded with doxorubicin (DOX) and ABT199 to synergistically prevent postoperative tumor recurrence. Enzymatic degradation of the biodegradable electrospun nanofibers facilitated the release of the two drugs. The primarily released DOX from the electrospun nanofibers effectively inhibited tumor recurrence. However, the sustained release of DOX led to drug resistance of the tumor cells, yielding unsatisfactory eradication of the residual tumor. Remarkably, the combined administration of DOX and ABT199, simultaneously released from the nanofibers, not only prolonged the chemotherapy by DOX but also overcame the drug resistance via inhibiting the Bcl-2 activation and thereby enhancing the apoptosis of tumor cells by ABT199. This dual-drug-loaded implant system, combining efficient chemotherapy and anti-drug resistance, offers a prospective strategy for the potent inhibition of postoperative tumor recurrence.
Collapse
|
29
|
Zhang Q, Wang X, Kuang G, Yu Y, Zhao Y. Photopolymerized 3D Printing Scaffolds with Pt(IV) Prodrug Initiator for Postsurgical Tumor Treatment. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9784510. [PMID: 36111316 PMCID: PMC9448443 DOI: 10.34133/2022/9784510] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/01/2022] [Indexed: 12/20/2022]
Abstract
Biomedical scaffolds have shown great success in postsurgical tumor treatment; their current efforts are focusing on eradicating residual tumor cells and circulating tumor cells and simultaneously repairing postoperative tissue defects. Herein, we report a novel photopolymerized 3D scaffold with Pt(IV) prodrug initiator to achieve the desired features for tumor comprehensive therapy. The Pt-GelMA scaffold was fabricated from the microfluidic 3D printing of methacrylate gelatin (GelMA) bioinks through a Pt(IV)-induced photocrosslinked process without any other additional photoinitiator and chemotherapeutic drug. Thus, the resultant scaffold displayed efficient cell killing ability against breast cancer cells in vitro and significantly inhibited the local tumor growth and distant metastases on an orthotopic postoperative breast cancer model in vivo. Besides, benefiting from their ordered porous structures and favorable biocompatibility, the scaffolds supported the cell attachment, spreading, and proliferation of normal cells in vitro; could facilitate the nutrient transportation; and induced new tissue ingrowth for repairing tissue defects caused by surgery. These properties indicate that such 3D printing scaffold is a promising candidate for efficient postoperative tumor treatment in the practical application.
Collapse
Affiliation(s)
- Qingfei Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Xiaocheng Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Gaizhen Kuang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Yunru Yu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| |
Collapse
|
30
|
Genta S, Coburn B, Cescon DW, Spreafico A. Patient-derived cancer models: Valuable platforms for anticancer drug testing. Front Oncol 2022; 12:976065. [PMID: 36033445 PMCID: PMC9413077 DOI: 10.3389/fonc.2022.976065] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Molecularly targeted treatments and immunotherapy are cornerstones in oncology, with demonstrated efficacy across different tumor types. Nevertheless, the overwhelming majority metastatic disease is incurable due to the onset of drug resistance. Preclinical models including genetically engineered mouse models, patient-derived xenografts and two- and three-dimensional cell cultures have emerged as a useful resource to study mechanisms of cancer progression and predict efficacy of anticancer drugs. However, variables including tumor heterogeneity and the complexities of the microenvironment can impair the faithfulness of these platforms. Here, we will discuss advantages and limitations of these preclinical models, their applicability for drug testing and in co-clinical trials and potential strategies to increase their reliability in predicting responsiveness to anticancer medications.
Collapse
Affiliation(s)
- Sofia Genta
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Bryan Coburn
- Division of Infectious Diseases, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - David W. Cescon
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Anna Spreafico
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
31
|
Moroni S, Casettari L, Lamprou DA. 3D and 4D Printing in the Fight against Breast Cancer. BIOSENSORS 2022; 12:568. [PMID: 35892465 PMCID: PMC9394292 DOI: 10.3390/bios12080568] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 06/15/2023]
Abstract
Breast cancer is the second most common cancer worldwide, characterized by a high incidence and mortality rate. Despite the advances achieved in cancer management, improvements in the quality of life of breast cancer survivors are urgent. Moreover, considering the heterogeneity that characterizes tumors and patients, focusing on individuality is fundamental. In this context, 3D printing (3DP) and 4D printing (4DP) techniques allow for a patient-centered approach. At present, 3DP applications against breast cancer are focused on three main aspects: treatment, tissue regeneration, and recovery of the physical appearance. Scaffolds, drug-loaded implants, and prosthetics have been successfully manufactured; however, some challenges must be overcome to shift to clinical practice. The introduction of the fourth dimension has led to an increase in the degree of complexity and customization possibilities. However, 4DP is still in the early stages; thus, research is needed to prove its feasibility in healthcare applications. This review article provides an overview of current approaches for breast cancer management, including standard treatments and breast reconstruction strategies. The benefits and limitations of 3DP and 4DP technologies are discussed, as well as their application in the fight against breast cancer. Future perspectives and challenges are outlined to encourage and promote AM technologies in real-world practice.
Collapse
Affiliation(s)
- Sofia Moroni
- School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK;
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy;
| | - Luca Casettari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy;
| | | |
Collapse
|
32
|
Fang Y, Liu Z, Wang H, Luo X, Xu Y, Chan HF, Lv S, Tao Y, Li M. Implantable Sandwich-like Scaffold/Fiber Composite Spatiotemporally Releasing Combretastatin A4 and Doxorubicin for Efficient Inhibition of Postoperative Tumor Recurrence. ACS APPLIED MATERIALS & INTERFACES 2022; 14:27525-27537. [PMID: 35687834 DOI: 10.1021/acsami.2c02103] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Tumor recurrence is a critical conundrum in the postoperative therapy, on account of severe bleeding with disseminated tumor cells, residual tumor cells, and the rich nutrient and oxygen supply transported to tumors by the abundant blood vessels. Biodegradable drug-loaded implants, inserted in the resection cavity right away upon the surgery, possess bleeding prevention and efficient chemotherapeutic capabilities, considered to be a promising strategy to efficiently inhibit the recurrence of the solid tumor. Here, we developed a sandwich-like composite consisting of the combretastatin A4 (CA4)-loaded 3D-printed scaffold and doxorubicin (DOX)-loaded electrospun fiber (Scaffold-CA4@Fiber-DOX), presenting hemostatic, chemotherapeutic, and antibacterial potencies. The lyophilized 3D-printed scaffold with a porous structure rapidly absorbed and clotted the blood cells and disseminated tumor cells to prevent bleeding and tumor metastasis. Subsequently, the preferentially released CA4 from the scaffold disrupted the microtubules of the vascular endothelial cell, resulting in vascular deformation and consequent insufficient nutrient supply to the solid tumor. The sustained release of DOX from the sandwiched electrospun fiber dramatically inhibited the peripheral tumor cell proliferation. This all-in-one multifunctional implant system, combining efficient vascular disruption and chemotherapy, provides a promising strategy for postoperative tumor therapy.
Collapse
Affiliation(s)
- Youqiang Fang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zheng Liu
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xing Luo
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yanteng Xu
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Shixian Lv
- School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease, Guangzhou 510630, China
| |
Collapse
|
33
|
Three-Dimensional (3D) Printing in Cancer Therapy and Diagnostics: Current Status and Future Perspectives. Pharmaceuticals (Basel) 2022; 15:ph15060678. [PMID: 35745597 PMCID: PMC9229198 DOI: 10.3390/ph15060678] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 12/10/2022] Open
Abstract
Three-dimensional (3D) printing is a technique where the products are printed layer-by-layer via a series of cross-sectional slices with the exact deposition of different cell types and biomaterials based on computer-aided design software. Three-dimensional printing can be divided into several approaches, such as extrusion-based printing, laser-induced forward transfer-based printing systems, and so on. Bio-ink is a crucial tool necessary for the fabrication of the 3D construct of living tissue in order to mimic the native tissue/cells using 3D printing technology. The formation of 3D software helps in the development of novel drug delivery systems with drug screening potential, as well as 3D constructs of tumor models. Additionally, several complex structures of inner tissues like stroma and channels of different sizes are printed through 3D printing techniques. Three-dimensional printing technology could also be used to develop therapy training simulators for educational purposes so that learners can practice complex surgical procedures. The fabrication of implantable medical devices using 3D printing technology with less risk of infections is receiving increased attention recently. A Cancer-on-a-chip is a microfluidic device that recreates tumor physiology and allows for a continuous supply of nutrients or therapeutic compounds. In this review, based on the recent literature, we have discussed various printing methods for 3D printing and types of bio-inks, and provided information on how 3D printing plays a crucial role in cancer management.
Collapse
|
34
|
Li S, Wang X, Chen J, Guo J, Yuan M, Wan G, Yan C, Li W, Machens HG, Rinkevich Y, Yang X, Song H, Chen Z. Calcium ion cross-linked sodium alginate hydrogels containing deferoxamine and copper nanoparticles for diabetic wound healing. Int J Biol Macromol 2022; 202:657-670. [PMID: 35066024 DOI: 10.1016/j.ijbiomac.2022.01.080] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/13/2021] [Accepted: 01/12/2022] [Indexed: 11/28/2022]
Abstract
Chronic non-healing diabetic wounds and ulcers can be fatal, lead to amputations, and remain a major challenge to medical, and health care sectors. Susceptibility to infection and impaired angiogenesis are two central reasons for the clinical consequences associated with chronic non-healing diabetic wounds. Herein, we successfully developed calcium ion (Ca2+) cross-linked sodium alginate (SA) hydrogels with both pro-angiogenesis and antibacterial properties. Our results demonstrated that deferoxamine (DFO) and copper nanoparticles (Cu-NPs) worked synergistically to enhance the proliferation, migration, and angiogenesis of human umbilical venous endothelial cells in vitro. Results of colony formation assay indicated Cu-NPs were effective against E. coli and S. aureus in a dose-dependent manner in vitro. An SA hydrogel containing both DFO and Cu-NPs (SA-DFO/Cu) was prepared using a Ca2+ cross-linking method. Cytotoxicity assay and colony formation assay indicated that the hydrogel exhibited beneficial biocompatible and antibacterial properties in vitro. Furthermore, SA-DFO/Cu significantly accelerated diabetic wound healing, improved angiogenesis and reduced long-lasting inflammation in a mouse model of diabetic wound. Mechanistically, DFO and Cu-NPs synergistically stimulated the levels of hypoxia-inducible factor 1α and vascular endothelial growth factor in vivo. Given the pro-angiogenesis, antibacterial and healing properties, the hydrogel possesses high potential for clinical application in refractory wounds.
Collapse
Affiliation(s)
- Shengbo Li
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xuemei Wang
- College of Chemistry & Molecular Science, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan 430072, China
| | - Jing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiahe Guo
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Meng Yuan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Gui Wan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chengqi Yan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenqing Li
- Department of Hand and Foot Surgery, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Hans-Günther Machens
- Department of Plastic and Hand Surgery, Technical University of Munich, Munich 81675, Germany
| | - Yuval Rinkevich
- Institute of Lung Biology and Disease, Helmholtz Zentrum München, Max-Lebsche-Platz 31, 81377 Munich, Germany; Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Max-Lebsche-Platz 31, 81377 Munich, Germany
| | - Xiaofan Yang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Heng Song
- College of Chemistry & Molecular Science, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan 430072, China.
| | - Zhenbing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
35
|
Pial MMH, Tomitaka A, Pala N, Roy U. Implantable Devices for the Treatment of Breast Cancer. JOURNAL OF NANOTHERANOSTICS 2022; 3:19-38. [PMID: 37600442 PMCID: PMC10438892 DOI: 10.3390/jnt3010003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023] Open
Abstract
Breast cancer is one of the leading causes of death in the female population worldwide. Standard treatments such as chemotherapy show noticeable results. However, along with killing cancer cells, it causes systemic toxicity and apoptosis of the nearby healthy cells, therefore patients must endure side effects during the treatment process. Implantable drug delivery devices that enhance therapeutic efficacy by allowing localized therapy with programmed or controlled drug release can overcome the shortcomings of conventional treatments. An implantable device can be composed of biopolymer materials, nanocomposite materials, or a combination of both. This review summarizes the recent research and current state-of-the art in these types of implantable devices and gives perspective for future directions.
Collapse
Affiliation(s)
| | - Asahi Tomitaka
- Department of Electrical and Computer Engineering, Florida International University, Miami, FL 33174, USA
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
- Department of Computer Science, University of Houston-Victoria, Victoria, TX 77901, USA
| | - Nezih Pala
- Department of Electrical and Computer Engineering, Florida International University, Miami, FL 33174, USA
| | - Upal Roy
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| |
Collapse
|
36
|
Zamboulis A, Michailidou G, Koumentakou I, Bikiaris DN. Polysaccharide 3D Printing for Drug Delivery Applications. Pharmaceutics 2022; 14:145. [PMID: 35057041 PMCID: PMC8778081 DOI: 10.3390/pharmaceutics14010145] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/19/2021] [Accepted: 12/24/2021] [Indexed: 12/27/2022] Open
Abstract
3D printing, or additive manufacturing, has gained considerable interest due to its versatility regarding design as well as in the large choice of materials. It is a powerful tool in the field of personalized pharmaceutical treatment, particularly crucial for pediatric and geriatric patients. Polysaccharides are abundant and inexpensive natural polymers, that are already widely used in the food industry and as excipients in pharmaceutical and cosmetic formulations. Due to their intrinsic properties, such as biocompatibility, biodegradability, non-immunogenicity, etc., polysaccharides are largely investigated as matrices for drug delivery. Although an increasing number of interesting reviews on additive manufacturing and drug delivery are being published, there is a gap concerning the printing of polysaccharides. In this article, we will review recent advances in the 3D printing of polysaccharides focused on drug delivery applications. Among the large family of polysaccharides, the present review will particularly focus on cellulose and cellulose derivatives, chitosan and sodium alginate, printed by fused deposition modeling and extrusion-based printing.
Collapse
Affiliation(s)
- Alexandra Zamboulis
- Laboratory of Chemistry and Technology of Polymers and Dyes, Department of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece; (G.M.); (I.K.)
| | | | | | - Dimitrios N. Bikiaris
- Laboratory of Chemistry and Technology of Polymers and Dyes, Department of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece; (G.M.); (I.K.)
| |
Collapse
|
37
|
Deshmane S, Kendre P, Mahajan H, Jain S. Stereolithography 3D printing technology in pharmaceuticals: a review. Drug Dev Ind Pharm 2021; 47:1362-1372. [PMID: 34663145 DOI: 10.1080/03639045.2021.1994990] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Three-dimensional printing (3DP) technology is an innovative tool used in manufacturing medical devices, producing alloys, replacing biological tissues, producing customized dosage forms and so on. Stereolithography (SLA), a 3D printing technique, is very rapid and highly accurate and produces finished products of uniform quality. 3D formulations have been optimized with a perfect tool of artificial intelligence learning techniques. Complex designs/shapes can be fabricated through SLA using the photopolymerization principle. Different 3DP technologies are introduced and the most promising of these, SLA, and its commercial applications, are focused on. The high speed and effectiveness of SLA are highlighted. The working principle of SLA, the materials used and applications of the technique in a wide range of different sectors are highlighted in this review. An innovative idea of 3D printing customized pharmaceutical dosage forms is also presented. SLA compromises several advantages over other methods, such as cost effectiveness, controlled integrity of materials and greater speed. The development of SLA has allowed the development of printed pharmaceutical devices. Considering the present trends, it is expected that SLA will be used along with conventional methods of manufacturing of 3D model. This 3D printing technology may be utilized as a novel tool for delivering drugs on demand. This review will be useful for researchers working on 3D printing technologies.
Collapse
Affiliation(s)
- Subhash Deshmane
- Department of Pharmaceutics, Rajarshi Shahu College of Pharmacy, Malvihir, India
| | - Prakash Kendre
- Department of Pharmaceutics, Rajarshi Shahu College of Pharmacy, Malvihir, India
| | - Hitendra Mahajan
- Department of Pharmaceutics, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Shirish Jain
- Department of Pharmaceutics, Rajarshi Shahu College of Pharmacy, Malvihir, India
| |
Collapse
|
38
|
Biopolymer Hydrogel Scaffolds Containing Doxorubicin as A Localized Drug Delivery System for Inhibiting Lung Cancer Cell Proliferation. Polymers (Basel) 2021; 13:polym13203580. [PMID: 34685337 PMCID: PMC8540863 DOI: 10.3390/polym13203580] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 12/02/2022] Open
Abstract
A hydrogel scaffold is a localized drug delivery system that can maintain the therapeutic level of drug concentration at the tumor site. In this study, the biopolymer hydrogel scaffold encapsulating doxorubicin was fabricated from gelatin, sodium carboxymethyl cellulose, and gelatin/sodium carboxymethyl cellulose mixture using a lyophilization technique. The effects of a crosslinker on scaffold morphology and pore size were determined using scanning electron microscopy. The encapsulation efficiency and the release profile of doxorubicin from the hydrogel scaffolds were determined using UV-Vis spectrophotometry. The anti-proliferative effect of the scaffolds against the lung cancer cell line was investigated using an MTT assay. The results showed that scaffolds made from different types of natural polymer had different pore configurations and pore sizes. All scaffolds had high encapsulation efficiency and drug-controlled release profiles. The viability and proliferation of A549 cells, treated with gelatin, gelatin/SCMC, and SCMC scaffolds containing doxorubicin significantly decreased compared with control. These hydrogel scaffolds might provide a promising approach for developing a superior localized drug delivery system to kill lung cancer cells.
Collapse
|
39
|
Guo B, Dong R, Liang Y, Li M. Haemostatic materials for wound healing applications. Nat Rev Chem 2021; 5:773-791. [PMID: 37117664 DOI: 10.1038/s41570-021-00323-z] [Citation(s) in RCA: 448] [Impact Index Per Article: 112.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2021] [Indexed: 12/12/2022]
Abstract
Wounds are one of the most common health issues, and the cost of wound care and healing has continued to increase over the past decade. The first step in wound healing is haemostasis, and the development of haemostatic materials that aid wound healing has accelerated in the past 5 years. Numerous haemostatic materials have been fabricated, composed of different active components (including natural polymers, synthetic polymers, silicon-based materials and metal-containing materials) and in various forms (including sponges, hydrogels, nanofibres and particles). In this Review, we provide an overview of haemostatic materials in wound healing, focusing on their chemical design and operation. We describe the physiological process of haemostasis to elucidate the principles that underpin the design of haemostatic wound dressings. We also highlight the advantages and limitations of the different active components and forms of haemostatic materials. The main challenges and future directions in the development of haemostatic materials for wound healing are proposed.
Collapse
|
40
|
Salerno A, Netti PA. Review on Computer-Aided Design and Manufacturing of Drug Delivery Scaffolds for Cell Guidance and Tissue Regeneration. Front Bioeng Biotechnol 2021; 9:682133. [PMID: 34249885 PMCID: PMC8264554 DOI: 10.3389/fbioe.2021.682133] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
In the last decade, additive manufacturing (AM) processes have updated the fields of biomaterials science and drug delivery as they promise to realize bioengineered multifunctional devices and implantable tissue engineering (TE) scaffolds virtually designed by using computer-aided design (CAD) models. However, the current technological gap between virtual scaffold design and practical AM processes makes it still challenging to realize scaffolds capable of encoding all structural and cell regulatory functions of the native extracellular matrix (ECM) of health and diseased tissues. Indeed, engineering porous scaffolds capable of sequestering and presenting even a complex array of biochemical and biophysical signals in a time- and space-regulated manner, require advanced automated platforms suitable of processing simultaneously biomaterials, cells, and biomolecules at nanometric-size scale. The aim of this work was to review the recent scientific literature about AM fabrication of drug delivery scaffolds for TE. This review focused on bioactive molecule loading into three-dimensional (3D) porous scaffolds, and their release effects on cell fate and tissue growth. We reviewed CAD-based strategies, such as bioprinting, to achieve passive and stimuli-responsive drug delivery scaffolds for TE and cancer precision medicine. Finally, we describe the authors' perspective regarding the next generation of CAD techniques and the advantages of AM, microfluidic, and soft lithography integration for enhancing 3D porous scaffold bioactivation toward functional bioengineered tissues and organs.
Collapse
Affiliation(s)
| | - Paolo A. Netti
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Interdisciplinary Research Center on Biomaterials, University of Naples Federico II, Naples, Italy
| |
Collapse
|