301
|
Parra-Cantu C, Li W, Quiñones-Hinojosa A, Zhang YS. 3D bioprinting of glioblastoma models. ACTA ACUST UNITED AC 2020; 4:113-125. [PMID: 33200034 DOI: 10.2217/3dp-2019-0027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 10/12/2020] [Indexed: 12/14/2022]
Abstract
The most common and malignant primary brain tumor in adults is glioblastoma (GBM). In vitro 3D brain models are needed to better understand the pathological processes underlying GBM and ultimately develop more efficient antineoplastic agents. Here, we describe the bioprinting methods that have been used to fabricate volumetric GBM models. We explain several factors that should be considered for 3D bioprinting, including bioinks, cells and construct designs, in relation to GBM modeling. Although 3D-bioprinted brain models are still to be improved, they have the potential to become a powerful tool for drug screening.
Collapse
Affiliation(s)
- Carolina Parra-Cantu
- Division of Engineering in Medicine, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Wanlu Li
- Division of Engineering in Medicine, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | | | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
302
|
Sanicola HW, Stewart CE, Mueller M, Ahmadi F, Wang D, Powell SK, Sarkar K, Cutbush K, Woodruff MA, Brafman DA. Guidelines for establishing a 3-D printing biofabrication laboratory. Biotechnol Adv 2020; 45:107652. [PMID: 33122013 DOI: 10.1016/j.biotechadv.2020.107652] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/23/2022]
Abstract
Advanced manufacturing and 3D printing are transformative technologies currently undergoing rapid adoption in healthcare, a traditionally non-manufacturing sector. Recent development in this field, largely enabled by merging different disciplines, has led to important clinical applications from anatomical models to regenerative bioscaffolding and devices. Although much research to-date has focussed on materials, designs, processes, and products, little attention has been given to the design and requirements of facilities for enabling clinically relevant biofabrication solutions. These facilities are critical to overcoming the major hurdles to clinical translation, including solving important issues such as reproducibility, quality control, regulations, and commercialization. To improve process uniformity and ensure consistent development and production, large-scale manufacturing of engineered tissues and organs will require standardized facilities, equipment, qualification processes, automation, and information systems. This review presents current and forward-thinking guidelines to help design biofabrication laboratories engaged in engineering model and tissue constructs for therapeutic and non-therapeutic applications.
Collapse
Affiliation(s)
- Henry W Sanicola
- Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia
| | - Caleb E Stewart
- Department of Neurosurgery, Louisiana State Health Sciences Center, Shreveport, LA 71103, USA.
| | | | - Farzad Ahmadi
- Department of Electrical and Computer Engineering, Youngstown State University, Youngstown, OH 44555, USA
| | - Dadong Wang
- Quantitative Imaging Research Team, Data61, Commonwealth Scientific and Industrial Research Organization, Marsfield, NSW 2122, Australia
| | - Sean K Powell
- Science and Engineering Faculty, Queensland University of Technology, Brisbane 4029, Australia
| | - Korak Sarkar
- M3D Laboratory, Ochsner Health System, New Orleans, LA 70121, USA
| | - Kenneth Cutbush
- Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia
| | - Maria A Woodruff
- Science and Engineering Faculty, Queensland University of Technology, Brisbane 4029, Australia.
| | - David A Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA.
| |
Collapse
|
303
|
Kim J, Kong JS, Han W, Kim BS, Cho DW. 3D Cell Printing of Tissue/Organ-Mimicking Constructs for Therapeutic and Drug Testing Applications. Int J Mol Sci 2020; 21:E7757. [PMID: 33092184 PMCID: PMC7589604 DOI: 10.3390/ijms21207757] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/15/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
The development of artificial tissue/organs with the functional maturity of their native equivalents is one of the long-awaited panaceas for the medical and pharmaceutical industries. Advanced 3D cell-printing technology and various functional bioinks are promising technologies in the field of tissue engineering that have enabled the fabrication of complex 3D living tissue/organs. Various requirements for these tissues, including a complex and large-volume structure, tissue-specific microenvironments, and functional vasculatures, have been addressed to develop engineered tissue/organs with native relevance. Functional tissue/organ constructs have been developed that satisfy such criteria and may facilitate both in vivo replenishment of damaged tissue and the development of reliable in vitro testing platforms for drug development. This review describes key developments in technologies and materials for engineering 3D cell-printed constructs for therapeutic and drug testing applications.
Collapse
Affiliation(s)
- Jongmin Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Jeong Sik Kong
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Wonil Han
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Byoung Soo Kim
- Future IT Innovation Laboratory, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- Institute of Convergence Science, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
304
|
Ruiz-Garcia H, Alvarado-Estrada K, Schiapparelli P, Quinones-Hinojosa A, Trifiletti DM. Engineering Three-Dimensional Tumor Models to Study Glioma Cancer Stem Cells and Tumor Microenvironment. Front Cell Neurosci 2020; 14:558381. [PMID: 33177991 PMCID: PMC7596188 DOI: 10.3389/fncel.2020.558381] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM) is the most common and devastating primary brain tumor, leading to a uniform fatality after diagnosis. A major difficulty in eradicating GBM is the presence of microscopic residual infiltrating disease remaining after multimodality treatment. Glioma cancer stem cells (CSCs) have been pinpointed as the treatment-resistant tumor component that seeds ultimate tumor progression. Despite the key role of CSCs, the ideal preclinical model to study the genetic and epigenetic landmarks driving their malignant behavior while simulating an accurate interaction with the tumor microenvironment (TME) is still missing. The introduction of three-dimensional (3D) tumor platforms, such as organoids and 3D bioprinting, has allowed for a better representation of the pathophysiologic interactions between glioma CSCs and the TME. Thus, these technologies have enabled a more detailed study of glioma biology, tumor angiogenesis, treatment resistance, and even performing high-throughput screening assays of drug susceptibility. First, we will review the foundation of glioma biology and biomechanics of the TME, and then the most up-to-date insights about the applicability of these new tools in malignant glioma research.
Collapse
Affiliation(s)
- Henry Ruiz-Garcia
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, United States.,Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | | | - Paula Schiapparelli
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | | | - Daniel M Trifiletti
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, United States.,Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
305
|
Aggarwal V, Miranda O, Johnston PA, Sant S. Three dimensional engineered models to study hypoxia biology in breast cancer. Cancer Lett 2020; 490:124-142. [PMID: 32569616 PMCID: PMC7442747 DOI: 10.1016/j.canlet.2020.05.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/13/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022]
Abstract
Breast cancer is the second leading cause of mortality among women worldwide. Despite the available therapeutic regimes, variable treatment response is reported among different breast cancer subtypes. Recently, the effects of the tumor microenvironment on tumor progression as well as treatment responses have been widely recognized. Hypoxia and hypoxia inducible factors in the tumor microenvironment have long been known as major players in tumor progression and survival. However, the majority of our understanding of hypoxia biology has been derived from two dimensional (2D) models. Although many hypoxia-targeted therapies have elicited promising results in vitro and in vivo, these results have not been successfully translated into clinical trials. These limitations of 2D models underscore the need to develop and integrate three dimensional (3D) models that recapitulate the complex tumor-stroma interactions in vivo. This review summarizes role of hypoxia in various hallmarks of cancer progression. We then compare traditional 2D experimental systems with novel 3D tissue-engineered models giving accounts of different bioengineering platforms available to develop 3D models and how these 3D models are being exploited to understand the role of hypoxia in breast cancer progression.
Collapse
Affiliation(s)
- Vaishali Aggarwal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Oshin Miranda
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Paul A Johnston
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; UPMC-Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; UPMC-Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
306
|
Tang M, Xie Q, Gimple RC, Zhong Z, Tam T, Tian J, Kidwell RL, Wu Q, Prager BC, Qiu Z, Yu A, Zhu Z, Mesci P, Jing H, Schimelman J, Wang P, Lee D, Lorenzini MH, Dixit D, Zhao L, Bhargava S, Miller TE, Wan X, Tang J, Sun B, Cravatt BF, Muotri AR, Chen S, Rich JN. Three-dimensional bioprinted glioblastoma microenvironments model cellular dependencies and immune interactions. Cell Res 2020; 30:833-853. [PMID: 32499560 PMCID: PMC7608409 DOI: 10.1038/s41422-020-0338-1] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 04/29/2020] [Indexed: 12/15/2022] Open
Abstract
Brain tumors are dynamic complex ecosystems with multiple cell types. To model the brain tumor microenvironment in a reproducible and scalable system, we developed a rapid three-dimensional (3D) bioprinting method to construct clinically relevant biomimetic tissue models. In recurrent glioblastoma, macrophages/microglia prominently contribute to the tumor mass. To parse the function of macrophages in 3D, we compared the growth of glioblastoma stem cells (GSCs) alone or with astrocytes and neural precursor cells in a hyaluronic acid-rich hydrogel, with or without macrophage. Bioprinted constructs integrating macrophage recapitulate patient-derived transcriptional profiles predictive of patient survival, maintenance of stemness, invasion, and drug resistance. Whole-genome CRISPR screening with bioprinted complex systems identified unique molecular dependencies in GSCs, relative to sphere culture. Multicellular bioprinted models serve as a scalable and physiologic platform to interrogate drug sensitivity, cellular crosstalk, invasion, context-specific functional dependencies, as well as immunologic interactions in a species-matched neural environment.
Collapse
Affiliation(s)
- Min Tang
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Qi Xie
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, 92037, USA.
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA.
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China.
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China.
| | - Ryan C Gimple
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
- Department of Pathology, Case Western University, Cleveland, OH, USA
| | - Zheng Zhong
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Trevor Tam
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Jing Tian
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Reilly L Kidwell
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Qiulian Wu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Briana C Prager
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
- Department of Pathology, Case Western University, Cleveland, OH, USA
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Zhixin Qiu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Aaron Yu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Zhe Zhu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Pinar Mesci
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
- Department of Cellular & Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Pediatrics/Rady Children's Hospital San Diego, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Hui Jing
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jacob Schimelman
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Pengrui Wang
- Materials Science and Engineering Program, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Derrick Lee
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Michael H Lorenzini
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Deobrat Dixit
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Linjie Zhao
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Shruti Bhargava
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, 92037, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Tyler E Miller
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Xueyi Wan
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jing Tang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
- Key Laboratory of Growth Regulation and Translation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Bingjie Sun
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Benjamin F Cravatt
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Alysson R Muotri
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
- Department of Cellular & Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Pediatrics/Rady Children's Hospital San Diego, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Kavli Institute for Brain and Mind, University of California San Diego, La Jolla, CA, 92093, USA
- Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, CA, 92093, USA
| | - Shaochen Chen
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
- Materials Science and Engineering Program, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - Jeremy N Rich
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, 92037, USA.
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA.
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, 92037, USA.
| |
Collapse
|
307
|
Yong U, Lee S, Jung S, Jang J. Interdisciplinary approaches to advanced cardiovascular tissue engineering: ECM-based biomaterials, 3D bioprinting, and its assessment. ACTA ACUST UNITED AC 2020. [DOI: 10.1088/2516-1091/abb211] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
308
|
Xie F, Sun L, Pang Y, Xu G, Jin B, Xu H, Lu X, Xu Y, Du S, Wang Y, Feng S, Sang X, Zhong S, Wang X, Sun W, Zhao H, Zhang H, Yang H, Huang P, Mao Y. Three-dimensional bio-printing of primary human hepatocellular carcinoma for personalized medicine. Biomaterials 2020; 265:120416. [PMID: 33007612 DOI: 10.1016/j.biomaterials.2020.120416] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/30/2020] [Accepted: 09/19/2020] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal tumors worldwide. This study aims to address the lack of faithful and available in vitro models for patient-specific drug screening for HCC. We recently established a novel modeling system using three-dimensional (3D) bioprinting technology and constructed hepatorganoids with HepaRG cells, which retain the liver function and prolong the survival of mice with liver failure after abdominal transplantation. Here we extend this modeling system to establish individualized model for hepatocellular carcinoma. HCC specimens were obtained from six patients after surgery. Primary HCC cells were isolated and mixed with gelatin and sodium alginate to form the bioink for printing. Patient-derived three-dimensional bio-printed HCC (3DP-HCC) models were successfully established afterward and grew well during long-term culture. These models retained the features of parental HCCs, including stable expression of the biomarker, stable maintenances of the genetic alterations and expression profiles. 3DP-HCC models are capable of displaying the results of drug screening intuitively and quantitatively. In conclusion, 3DP-HCC models are faithful in vitro models that are reliable in long-term culture and able to predict patient-specific drugs for personalized treatment.
Collapse
Affiliation(s)
- Feihu Xie
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Lejia Sun
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Yuan Pang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China; Overseas Expertise Introduction Center for Discipline Innovation, Tsinghua University, Beijing, 100084, China
| | - Gang Xu
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Bao Jin
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Haifeng Xu
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Xin Lu
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Yiyao Xu
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Yanan Wang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, PUMC & CAMS, Beijing, 100005, China
| | - Shi Feng
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Xinting Sang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Shouxian Zhong
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Xin Wang
- Research Center for Laboratory Animal Science, Inner Mongolia University, Huhhot, 010021, China; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA; Hepatoscience Section, Cell Lab Tech Inc., Sunnyvale, CA, 94085, USA
| | - Wei Sun
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China; Overseas Expertise Introduction Center for Discipline Innovation, Tsinghua University, Beijing, 100084, China; Department of Mechanical Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China
| | - Hongbing Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, PUMC & CAMS, Beijing, 100005, China
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China.
| | - Pengyu Huang
- School of Life Science and Technology, Shanghai Tech University, Shanghai, 201210, China; CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences (CAMS), Beijing, 100730, China.
| |
Collapse
|
309
|
Bae M, Yi HG, Jang J, Cho DW. Microphysiological Systems for Neurodegenerative Diseases in Central Nervous System. MICROMACHINES 2020; 11:E855. [PMID: 32947879 PMCID: PMC7570039 DOI: 10.3390/mi11090855] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022]
Abstract
Neurodegenerative diseases are among the most severe problems in aging societies. Various conventional experimental models, including 2D and animal models, have been used to investigate the pathogenesis of (and therapeutic mechanisms for) neurodegenerative diseases. However, the physiological gap between humans and the current models remains a hurdle to determining the complexity of an irreversible dysfunction in a neurodegenerative disease. Therefore, preclinical research requires advanced experimental models, i.e., those more physiologically relevant to the native nervous system, to bridge the gap between preclinical stages and patients. The neural microphysiological system (neural MPS) has emerged as an approach to summarizing the anatomical, biochemical, and pathological physiology of the nervous system for investigation of neurodegenerative diseases. This review introduces the components (such as cells and materials) and fabrication methods for designing a neural MPS. Moreover, the review discusses future perspectives for improving the physiological relevance to native neural systems.
Collapse
Affiliation(s)
- Mihyeon Bae
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea;
| | - Hee-Gyeong Yi
- Department of Rural and Biosystems Engineering, College of Agricultural Sciences, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea;
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea
- Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Chungam-ro, Nam-gu, Pohang 37673, Korea;
- Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| |
Collapse
|
310
|
Huayamares SG, Song JY, Huang A, Crowl SR, Groer CE, Forrest ML, Berkland CJ. Constructing a Biomaterial to Simulate Extracellular Drug Transport in Solid Tumors. Macromol Biosci 2020; 20:e2000251. [PMID: 32924274 DOI: 10.1002/mabi.202000251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/24/2020] [Indexed: 12/25/2022]
Abstract
Designing an in vitro model of the tumor extracellular microenvironment to screen intratumoral drugs is an active challenge. As recent clinical successes of human intratumoral therapies stimulate research on intratumoral delivery, a need for a 3D tumor model to screen intratumoral therapies arises. When injecting the drug formulation directly into the tumor, the biophysics affecting intratumoral retention must be considered; especially for biologic therapies, which may be dominated by extracellular transport mechanisms. Fibrotic regions in solid tumors are typically rich in collagen I fibers. Using shear rheology, head and neck tumors with higher collagen density show a higher stiffness. Similarly, the stiffness of the hyaluronic acid (HA) hydrogel models is increased by adding collagen fibers to model the bulk biomechanical properties of solid tumors. HA hydrogels are then used as intratumoral injection site simulators to model in vitro the retention of glatiramer acetate (GA) and polyethylene glycol (PEG) administered intratumorally. Both compounds are also injected in murine tumors and retention is studied ex vivo for comparison. Retention of GA in the hydrogels is significantly longer than PEG, analogous to the solid tumors, suggesting the utility of HA hydrogels with collagen I fibers for screening extracellular drug transport after intratumoral administration.
Collapse
Affiliation(s)
| | - Jimmy Y Song
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66045, USA
| | - Aric Huang
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66045, USA
| | - Samuel R Crowl
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS, 66045, USA
| | | | - M Laird Forrest
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66045, USA.,HylaPharm, LLC, Lawrence, KS, 66045, USA
| | - Cory J Berkland
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, 66045, USA.,Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66045, USA.,Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS, 66045, USA
| |
Collapse
|
311
|
Cui X, Ma C, Vasudevaraja V, Serrano J, Tong J, Peng Y, Delorenzo M, Shen G, Frenster J, Morales RTT, Qian W, Tsirigos A, Chi AS, Jain R, Kurz SC, Sulman EP, Placantonakis DG, Snuderl M, Chen W. Dissecting the immunosuppressive tumor microenvironments in Glioblastoma-on-a-Chip for optimized PD-1 immunotherapy. eLife 2020; 9:52253. [PMID: 32909947 PMCID: PMC7556869 DOI: 10.7554/elife.52253] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Programmed cell death protein-1 (PD-1) checkpoint immunotherapy efficacy remains unpredictable in glioblastoma (GBM) patients due to the genetic heterogeneity and immunosuppressive tumor microenvironments. Here, we report a microfluidics-based, patient-specific 'GBM-on-a-Chip' microphysiological system to dissect the heterogeneity of immunosuppressive tumor microenvironments and optimize anti-PD-1 immunotherapy for different GBM subtypes. Our clinical and experimental analyses demonstrated that molecularly distinct GBM subtypes have distinct epigenetic and immune signatures that may lead to different immunosuppressive mechanisms. The real-time analysis in GBM-on-a-Chip showed that mesenchymal GBM niche attracted low number of allogeneic CD154+CD8+ T-cells but abundant CD163+ tumor-associated macrophages (TAMs), and expressed elevated PD-1/PD-L1 immune checkpoints and TGF-β1, IL-10, and CSF-1 cytokines compared to proneural GBM. To enhance PD-1 inhibitor nivolumab efficacy, we co-administered a CSF-1R inhibitor BLZ945 to ablate CD163+ M2-TAMs and strengthened CD154+CD8+ T-cell functionality and GBM apoptosis on-chip. Our ex vivo patient-specific GBM-on-a-Chip provides an avenue for a personalized screening of immunotherapies for GBM patients.
Collapse
Affiliation(s)
- Xin Cui
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, United States.,Department of Biomedical Engineering, New York University, Brooklyn, United States.,Department of Biomedical Engineering, Jinan University, Guangzhou, China
| | - Chao Ma
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, United States.,Department of Biomedical Engineering, New York University, Brooklyn, United States
| | | | - Jonathan Serrano
- Department of Pathology, NYU Langone Health, New York, United States
| | - Jie Tong
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, United States.,Department of Biomedical Engineering, New York University, Brooklyn, United States
| | - Yansong Peng
- Department of Biomedical Engineering, New York University, Brooklyn, United States
| | - Michael Delorenzo
- Department of Pathology, NYU Langone Health, New York, United States
| | - Guomiao Shen
- Department of Pathology, NYU Langone Health, New York, United States
| | - Joshua Frenster
- Stem Cell Biology Program, NYU School of Medicine, New York, United States
| | | | - Weiyi Qian
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, United States
| | | | - Andrew S Chi
- Perlmutter Cancer Center, NYU Langone Health, New York, United States.,Department of Neurology, NYU Langone Health, New York, United States
| | - Rajan Jain
- Department of Neuroradiology, NYU Langone Health, New York, United States.,Department of Neurosurgery, NYU Langone Health, New York, United States
| | - Sylvia C Kurz
- Perlmutter Cancer Center, NYU Langone Health, New York, United States.,Department of Neurosurgery, NYU Langone Health, New York, United States
| | - Erik P Sulman
- Perlmutter Cancer Center, NYU Langone Health, New York, United States.,Department of Radiation Oncology, NYU Langone Health, New York, United States
| | - Dimitris G Placantonakis
- Perlmutter Cancer Center, NYU Langone Health, New York, United States.,Department of Neurosurgery, NYU Langone Health, New York, United States
| | - Matija Snuderl
- Department of Pathology, NYU Langone Health, New York, United States.,Perlmutter Cancer Center, NYU Langone Health, New York, United States
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, United States.,Department of Biomedical Engineering, New York University, Brooklyn, United States.,Perlmutter Cancer Center, NYU Langone Health, New York, United States
| |
Collapse
|
312
|
Cha GD, Kang T, Baik S, Kim D, Choi SH, Hyeon T, Kim DH. Advances in drug delivery technology for the treatment of glioblastoma multiforme. J Control Release 2020; 328:350-367. [PMID: 32896613 DOI: 10.1016/j.jconrel.2020.09.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023]
Abstract
Glioblastoma multiforme (GBM) is a particularly aggressive and malignant type of brain tumor, notorious for its high recurrence rate and low survival rate. The treatment of GBM is challenging mainly because several issues associated with the GBM microenvironment have not yet been resolved. These obstacles originate from a variety of factors such as genetics, anatomy, and cytology, all of which collectively hinder the treatment of GBM. Recent advances in materials and device engineering have presented new perspectives with regard to unconventional drug administration methods for GBM treatment. Such novel drug delivery approaches, based on the clear understanding of the intrinsic properties of GBM, have shown promise in overcoming some of the obstacles. In this review, we first recapitulate the first-line therapy and clinical challenges in the current treatment of GBM. Afterwards, we introduce the latest technological advances in drug delivery strategies to improve the efficiency for GBM treatment, mainly focusing on materials and devices. We describe such efforts by classifying them into two categories, systemic and local drug delivery. Finally, we discuss unmet challenges and prospects for the clinical translation of these drug delivery technologies.
Collapse
Affiliation(s)
- Gi Doo Cha
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Taegyu Kang
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Seungmin Baik
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Dokyoon Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea; Department of Bionano Engineering and Bionanotechnology, Hanyang University, Ansan 15588, Republic of Korea
| | - Seung Hong Choi
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea; Department of Radiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea.
| | - Dae-Hyeong Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
313
|
Li J, Parra-Cantu C, Wang Z, Zhang YS. Improving Bioprinted Volumetric Tumor Microenvironments In Vitro. Trends Cancer 2020; 6:745-756. [PMID: 32680649 PMCID: PMC7483398 DOI: 10.1016/j.trecan.2020.06.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/03/2020] [Accepted: 06/09/2020] [Indexed: 01/09/2023]
Abstract
Despite the great breakthroughs in the past few decades in illuminating the pathological mechanisms of cancer and in developing new anticancer drugs, it remains extremely challenging to cure most cancers. Therefore, it is imperative to develop more sophisticated and more biomimetic preclinical cancer models. 3D models combined with dynamic culture techniques show great potential to accurately emulate the volumetric tumor microenvironment (TME). Here we introduce advances in bioprinting technologies for in vitro cancer modeling and their applications. Finally, we look ahead to the remaining challenges associated with current bioprinting strategies for achieving faithful cancer modeling.
Collapse
Affiliation(s)
- Jun Li
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Carolina Parra-Cantu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Zongyi Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
| |
Collapse
|
314
|
Wang X, Li X, Ding J, Long X, Zhang H, Zhang X, Jiang X, Xu T. 3D bioprinted glioma microenvironment for glioma vascularization. J Biomed Mater Res A 2020; 109:915-925. [PMID: 32779363 DOI: 10.1002/jbm.a.37082] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 07/28/2020] [Accepted: 08/03/2020] [Indexed: 12/16/2022]
Abstract
Glioblastoma is the most frequently diagnosed primary malignant brain tumor with unfavourable prognosis and high mortality. One of its key features is the extensive abnormal vascular network. Up to now, the mechanism of angiogenesis and the origin of tumor vascularization remain controversial. It is essential to establish an ideal preclinical tumor model to elucidate the mechanism of tumor vascularization, and the role of tumor cells in this process. In this study, both U118 cell and GSC23 cell exhibited good printability and cell proliferation. Compared with 3D-U118, 3D-GSC23 had a greater ability to form cell spheroids, to secrete vascular endothelial growth factor (VEGFA), and to form tubule-like structures in vitro. More importantly, 3D-glioma stem cells (GSC)23 cells had a greater power to transdifferentiate into functional endothelial cells, and blood vessels composed of tumor cells with an abnormal endothelial phenotype was observed in vivo. In summary, 3D bioprinted hydrogel scaffold provided a suitable tumor microenvironment (TME) for glioma cells and GSCs. This bioprinted model supported a novel TME for the research of glioma cells, especially GSCs in glioma vascularization and therapeutic targeting of tumor angiogenesis.
Collapse
Affiliation(s)
- Xuanzhi Wang
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Xinda Li
- Department of Mechanical Engineering, Biomanufacturing Center, Tsinghua University, Beijing, China
| | - Jinju Ding
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, China
| | - Xiaoyan Long
- Department of research and development, East China Institute of Digital Medical Engineering, Shangrao, People's Republic of China
| | - Haitao Zhang
- Department of research and development, East China Institute of Digital Medical Engineering, Shangrao, People's Republic of China
| | - Xinzhi Zhang
- Department of research and development, Medprin Regenerative Medical Technologies Co., Ltd, Shenzhen, People's Republic of China
| | - Xiaochun Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Tao Xu
- Department of Mechanical Engineering, Biomanufacturing Center, Tsinghua University, Beijing, China.,Department of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, China
| |
Collapse
|
315
|
Deal HE, Brown AC, Daniele MA. Microphysiological systems for the modeling of wound healing and evaluation of pro-healing therapies. J Mater Chem B 2020; 8:7062-7075. [PMID: 32756718 PMCID: PMC7460719 DOI: 10.1039/d0tb00544d] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Wound healing is a multivariate process involving the coordinated response of numerous proteins and cell types. Accordingly, biomedical research has seen an increased adoption of the use of in vitro wound healing assays with complexity beyond that offered by traditional well-plate constructs. These microphysiological systems (MPS) seek to recapitulate one or more physiological features of the in vivo microenvironment, while retaining the analytical capacity of more reductionist assays. Design efforts to achieve relevant wound healing physiology include the use of dynamic perfusion over static culture, the incorporation of multiple cell types, the arrangement of cells in three dimensions, the addition of biomechanically and biochemically relevant hydrogels, and combinations thereof. This review provides a brief overview of the wound healing process and in vivo assays, and we critically review the current state of MPS and supporting technologies for modelling and studying wound healing. We distinguish between MPS that seek to inform a particular phase of wound healing, and constructs that have the potential to inform multiple phases of wound healing. This distinction is a product of whether analysis of a particular process is prioritized, or a particular physiology is prioritized, during design. Material selection is emphasized throughout, and relevant fabrication techniques discussed.
Collapse
Affiliation(s)
- Halston E Deal
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA. and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA
| | - Ashley C Brown
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA. and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA
| | - Michael A Daniele
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA. and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA and Department of Electrical & Computer Engineering, North Carolina State University, 890 Oval Dr., Raleigh, NC 27695, USA
| |
Collapse
|
316
|
Soubéran A, Tchoghandjian A. Practical Review on Preclinical Human 3D Glioblastoma Models: Advances and Challenges for Clinical Translation. Cancers (Basel) 2020; 12:cancers12092347. [PMID: 32825103 PMCID: PMC7563542 DOI: 10.3390/cancers12092347] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/07/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023] Open
Abstract
Fifteen years after the establishment of the Stupp protocol as the standard of care to treat glioblastomas, no major clinical advances have been achieved and increasing patient’s overall survival remains a challenge. Nevertheless, crucial molecular and cellular findings revealed the intra-tumoral and inter-tumoral complexities of these incurable brain tumors, and the essential role played by cells of the microenvironment in the lack of treatment efficacy. Taking this knowledge into account, fulfilling gaps between preclinical models and clinical samples is necessary to improve the successful rate of clinical trials. Since the beginning of the characterization of brain tumors initiated by Bailey and Cushing in the 1920s, several glioblastoma models have been developed and improved. In this review, we focused on the most widely used 3D human glioblastoma models, including spheroids, tumorospheres, organotypic slices, explants, tumoroids and glioblastoma-derived from cerebral organoids. We discuss their history, development and especially their usefulness.
Collapse
|
317
|
Abstract
Purpose of review This review focuses on the development and progression of glioblastoma through the brain and glioma microenvironment. Specifically we highlight how the tumor microenvironment contributes to the hallmarks of cancer in hopes of offering novel therapeutic options and tools to target this microenvironment. Recent findings The hallmarks of cancer, which represent elements of cancers that contribute to the disease's malignancy, yet elements within the brain tumor microenvironment, such as other cellular types as well as biochemical and biophysical cues that can each uniquely affect tumor cells, have not been well-described in this context and serve as potential targets for modulation. Summary Here, we highlight how the brain tumor microenvironment contributes to the progression and therapeutic response of tumor cells. Specifically, we examine these contributions through the lens of Hanahan & Weinberg's Hallmarks of Cancer in order to identify potential novel targets within the brain that may offer a means to treat brain cancers, including the deadliest brain cancer, glioblastoma.
Collapse
|
318
|
Kang Y, Datta P, Shanmughapriya S, Ozbolat IT. 3D Bioprinting of Tumor Models for Cancer Research. ACS APPLIED BIO MATERIALS 2020; 3:5552-5573. [DOI: 10.1021/acsabm.0c00791] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Youngnam Kang
- The Huck Institutes of the Life Sciences, Penn State University, University Park, Pennsylvania 16802, United States
- Engineering Science and Mechanics Department, Penn State University, University Park, Pennsylvania 16802, United States
| | - Pallab Datta
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology Shibpur, Howrah, West Bengal 711103, India
| | - Santhanam Shanmughapriya
- Department of Medicine, Penn State University, College of Medicine, Hershey, Pennsylvania 17033, United States
- Department of Cellular and Molecular Physiology, Penn State University, College of Medicine, Hershey, Pennsylvania 17033, United States
- Heart and Vascular Institute, Penn State University, College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Ibrahim T. Ozbolat
- The Huck Institutes of the Life Sciences, Penn State University, University Park, Pennsylvania 16802, United States
- Engineering Science and Mechanics Department, Penn State University, University Park, Pennsylvania 16802, United States
- Biomedical Engineering Department, Penn State University, University Park, Pennsylvania 16802, United States
- Materials Research Institute, Penn State University, University Park, Pennsylvania 16802, United States
- Department of Neurosurgery, Penn State University, Hershey, Pennsylvania 17033, United States
| |
Collapse
|
319
|
Correia Carreira S, Begum R, Perriman AW. 3D Bioprinting: The Emergence of Programmable Biodesign. Adv Healthc Mater 2020; 9:e1900554. [PMID: 31407502 DOI: 10.1002/adhm.201900554] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/04/2019] [Indexed: 11/10/2022]
Abstract
Until recently, bioprinting was largely limited to highly interdisciplinary research teams, as the process requires significant input from specialists in the fields of materials science, engineering, and cell biology. With the advent of commercially available high-performance bioprinters, the field has become accessible to a wider range of research groups, who can now buy the hardware off the shelf instead of having to build it from scratch. As a result, bioprinting has rapidly expanded to address a wide array of research foci, which include organotypic in vitro models, complex engineered tissues, and even bioprinted microbial systems. Moreover, in the early days, the range of suitable bioinks was limited. Now, there is a plethora of viable options to suit many cell phenotypes. This rapidly evolving dynamic environment creates endless opportunities for scientists to design and construct highly complex biological systems. However, this scientific diversity presents its own set of challenges, such as defining standardized protocols for characterizing bioprinted structures, which is essential for eventual organ replacement. In this progress report, the current state-of-the-art in the field of bioprinting is discussed, with a special emphasis on recent hardware developments, bioprinting for regenerative medicine, and late-breaking nontraditional topics.
Collapse
Affiliation(s)
- Sara Correia Carreira
- School of Cellular and Molecular MedicineUniversity of Bristol University Walk Bristol BS8 1TD UK
| | - Runa Begum
- School of Cellular and Molecular MedicineUniversity of Bristol University Walk Bristol BS8 1TD UK
| | - Adam W. Perriman
- School of Cellular and Molecular MedicineUniversity of Bristol University Walk Bristol BS8 1TD UK
| |
Collapse
|
320
|
Kim BS, Das S, Jang J, Cho DW. Decellularized Extracellular Matrix-based Bioinks for Engineering Tissue- and Organ-specific Microenvironments. Chem Rev 2020; 120:10608-10661. [PMID: 32786425 DOI: 10.1021/acs.chemrev.9b00808] [Citation(s) in RCA: 226] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Biomaterials-based biofabrication methods have gained much attention in recent years. Among them, 3D cell printing is a pioneering technology to facilitate the recapitulation of unique features of complex human tissues and organs with high process flexibility and versatility. Bioinks, combinations of printable hydrogel and cells, can be utilized to create 3D cell-printed constructs. The bioactive cues of bioinks directly trigger cells to induce tissue morphogenesis. Among the various printable hydrogels, the tissue- and organ-specific decellularized extracellular matrix (dECM) can exert synergistic effects in supporting various cells at any component by facilitating specific physiological properties. In this review, we aim to discuss a new paradigm of dECM-based bioinks able to recapitulate the inherent microenvironmental niche in 3D cell-printed constructs. This review can serve as a toolbox for biomedical engineers who want to understand the beneficial characteristics of the dECM-based bioinks and a basic set of fundamental criteria for printing functional human tissues and organs.
Collapse
Affiliation(s)
- Byoung Soo Kim
- Future IT Innovation Laboratory, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu,, Pohang, Kyungbuk 37673, Republic of Korea.,POSTECH-Catholic Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea
| | - Sanskrita Das
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea
| | - Jinah Jang
- Future IT Innovation Laboratory, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu,, Pohang, Kyungbuk 37673, Republic of Korea.,Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,POSTECH-Catholic Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,POSTECH-Catholic Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea.,Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
321
|
Zhao F, Cheng J, Sun M, Yu H, Wu N, Li Z, Zhang J, Li Q, Yang P, Liu Q, Hu X, Ao Y. Digestion degree is a key factor to regulate the printability of pure tendon decellularized extracellular matrix bio-ink in extrusion-based 3D cell printing. Biofabrication 2020; 12:045011. [DOI: 10.1088/1758-5090/aba411] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
322
|
Mao S, He J, Zhao Y, Liu T, Xie F, Yang H, Mao Y, Pang Y, Sun W. Bioprinting of patient-derived in vitro intrahepatic cholangiocarcinoma tumor model: establishment, evaluation and anti-cancer drug testing. Biofabrication 2020; 12:045014. [PMID: 32599574 DOI: 10.1088/1758-5090/aba0c3] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Towards the development of in vivo-mimicking tumor model for extensive study of tumorigenesis and establishment of personalized therapy, patient-derived primary tumor cells were employed in this work for three-dimensional (3D) bioprinting. Intrahepatic cholangiocarcinoma cells isolated from patient were bioprinted using a composite hydrogel system of gelatin-alginate-MatrigelTM into pre-designed grid architecture. ICC cells were observed to process a colony forming ability with high survival rate and active proliferation. Expression levels of tumor markers, cancer stem cell markers, matrix metalloproteinase protein, index of tumor fibrosis, index of liver function, and epithelial-mesenchymal transition regulatory proteins confirmed the development of the invasive and metastatic phenotype of the intrahepatic cholangiocarcinoma cells in the 3D printed tumor microenvironment. Similar results were obtained in anti-cancer drug resistance of the intrahepatic cholangiocarcinoma cells in the 3D bioprinted construct that demonstrated stem-like properties, which suggested the promising potential of current 3D printed tumor model in the development of personalized therapy, especially for discovery of more conducive targeted drugs.
Collapse
Affiliation(s)
- Shuangshuang Mao
- Department of Mechanical Engineering, Biomanufacturing Center, Tsinghua University, Beijing, People's Republic of China. Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, People's Republic of China. 'Biomanufacturing and Engineering Living Systems' 111 - Innovation International Talents Base, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
323
|
Datta P, Dey M, Ataie Z, Unutmaz D, Ozbolat IT. 3D bioprinting for reconstituting the cancer microenvironment. NPJ Precis Oncol 2020; 4:18. [PMID: 32793806 PMCID: PMC7385083 DOI: 10.1038/s41698-020-0121-2] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 05/13/2020] [Indexed: 12/30/2022] Open
Abstract
The cancer microenvironment is known for its complexity, both in its content as well as its dynamic nature, which is difficult to study using two-dimensional (2D) cell culture models. Several advances in tissue engineering have allowed more physiologically relevant three-dimensional (3D) in vitro cancer models, such as spheroid cultures, biopolymer scaffolds, and cancer-on-a-chip devices. Although these models serve as powerful tools for dissecting the roles of various biochemical and biophysical cues in carcinoma initiation and progression, they lack the ability to control the organization of multiple cell types in a complex dynamic 3D architecture. By virtue of its ability to precisely define perfusable networks and position of various cell types in a high-throughput manner, 3D bioprinting has the potential to more closely recapitulate the cancer microenvironment, relative to current methods. In this review, we discuss the applications of 3D bioprinting in mimicking cancer microenvironment, their use in immunotherapy as prescreening tools, and overview of current bioprinted cancer models.
Collapse
Affiliation(s)
- Pallab Datta
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology Shibpur, Howrah, India
| | - Madhuri Dey
- Department of Chemistry, Penn State University, University Park, PA USA
| | - Zaman Ataie
- Engineering Science and Mechanics Department, Penn State University, University Park, PA USA
| | - Derya Unutmaz
- The Jackson Laboratory of Genomics Medicine, Farmington, CT USA
| | - Ibrahim T. Ozbolat
- Engineering Science and Mechanics Department, Penn State University, University Park, PA USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA USA
- Biomedical Engineering Department, Penn State University, University Park, PA USA
- Materials Research Institute, Penn State University, University Park, PA USA
| |
Collapse
|
324
|
Lin KC, Sun Y, Torga G, Sherpa P, Zhao Y, Qu J, Amend SR, Pienta KJ, Sturm JC, Austin RH. An in vitro tumor swamp model of heterogeneous cellular and chemotherapeutic landscapes. LAB ON A CHIP 2020; 20:2453-2464. [PMID: 32555901 DOI: 10.1039/d0lc00131g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The heterogenous, highly metabolic stressed, poorly irrigated, solid tumor microenvironment - the tumor swamp - is widely recognized to play an important role in cancer progression as well as the development of therapeutic resistance. It is thus important to create realistic in vitro models within the therapeutic pipeline that can recapitulate the fundamental stress features of the tumor swamp. Here we describe a microfluidic system which generates a chemical gradient within connected microenvironments achieved through a static diffusion mechanism rather than active pumping. We show that the gradient can be stably maintained for over a week. Due to the accessibility and simplicity of the experimental platform, the system allows for not only well-controlled continuous studies of the interactions among various cell types at single-cell resolution, but also parallel experimentation for time-resolved downstream cellular assays on the time scale of weeks. This approach enables simple, compact implementation and is compatible with existing 6-well imaging technology for simultaneous experiments. As a proof-of-concept, we report the co-culture of a human bone marrow stromal cell line and a bone-metastatic prostate cancer cell line using the presented device, revealing on the same chip a transition in cancer cell survival as a function of drug concentration on the population level while exhibiting an enrichment of poly-aneuploid cancer cells (PACCs) as an evolutionary consequence of high stress. The device allows for the quantitative study of cancer cell dynamics on a stress landscape by real-time monitoring of various cell types with considerable experimental throughput.
Collapse
Affiliation(s)
- Ke-Chih Lin
- Department of Physics, Princeton University, Princeton, NJ 08544, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
325
|
Chadwick M, Yang C, Liu L, Gamboa CM, Jara K, Lee H, Sabaawy HE. Rapid Processing and Drug Evaluation in Glioblastoma Patient-Derived Organoid Models with 4D Bioprinted Arrays. iScience 2020; 23:101365. [PMID: 32731171 PMCID: PMC7393526 DOI: 10.1016/j.isci.2020.101365] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/29/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma is the most common and deadly primary brain malignancy. Despite advances in precision medicine oncology (PMO) allowing the identification of molecular vulnerabilities in glioblastoma, treatment options remain limited, and molecular assays guided by genomic and expression profiling to inform patient enrollment in life-saving trials are lacking. Here, we generate four-dimensional (4D) cell-culture arrays for rapid assessment of drug responses in glioblastoma patient-derived models. The arrays are 3D printed with thermo-responsive shape memory polymer (SMP). Upon heating, the SMP arrays self-transform in time from 3D cell-culture inserts into histological cassettes. We assess the utility of these arrays with glioblastoma cells, gliospheres, and patient derived organoid-like (PDO) models and demonstrate their use with glioblastoma PDOs for assessing drug sensitivity, on-target activity, and synergy in drug combinations. When including genomic and drug testing assays, this platform is poised to offer rapid functional drug assessments for future selection of therapies in PMO.
Collapse
Affiliation(s)
- Michelle Chadwick
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| | - Chen Yang
- Department of Mechanical and Aerospace Engineering, Rutgers University - New Brunswick, New Brunswick, NJ 08901, USA
| | - Liqiong Liu
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| | - Christian Moya Gamboa
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| | - Kelly Jara
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| | - Howon Lee
- Department of Mechanical and Aerospace Engineering, Rutgers University - New Brunswick, New Brunswick, NJ 08901, USA.
| | - Hatem E Sabaawy
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA; Departments of Medicine and Pathology and Laboratory Medicine, RBHS-Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA.
| |
Collapse
|
326
|
Mao S, Pang Y, Liu T, Shao Y, He J, Yang H, Mao Y, Sun W. Bioprinting of in vitro tumor models for personalized cancer treatment: a review. Biofabrication 2020; 12:042001. [PMID: 32470967 DOI: 10.1088/1758-5090/ab97c0] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Studying biological characteristics of tumors and evaluating the treatment effects require appropriate in vitro tumor models. However, the occurrence, progression, and migration of tumors involve spatiotemporal changes, cell-microenvironment and cell-cell interactions, and signal transmission in cells, which makes the construction of in vitro tumor models extremely challenging. In the past few years, advances in biomaterials and tissue engineering methods, especially development of the bioprinting technology, have paved the way for innovative platform technologies for in vitro cancer research. Bioprinting can accurately control the distribution of cells, active molecules, and biomaterials. Furthermore, this technology recapitulates the key characteristics of the tumor microenvironment and constructs in vitro tumor models with bionic structures and physiological systems. These models can be used as robust platforms to study tumor initiation, interaction with the microenvironment, angiogenesis, motility and invasion, as well as intra- and extravasation. Bioprinted tumor models can also be used for high-throughput drug screening and validation and provide the possibility for personalized cancer treatment research. This review describes the basic characteristics of the tumor and its microenvironment and focuses on the importance and relevance of bioprinting technology in the construction of tumor models. Research progress in the bioprinting of monocellular, multicellular, and personalized tumor models is discussed, and comprehensive application of bioprinting in preclinical drug screening and innovative therapy is reviewed. Finally, we offer our perspective on the shortcomings of the existing models and explore new technologies to outline the direction of future development and application prospects of next-generation tumor models.
Collapse
Affiliation(s)
- Shuangshuang Mao
- Department of Mechanical Engineering, Biomanufacturing Center, Tsinghua University, Beijing, People's Republic of China. Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, People's Republic of China. 'Biomanufacturing and Engineering Living Systems' 111 -Innovation International Talents Base, Beijing, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
327
|
Browning JR, Derr P, Derr K, Doudican N, Michael S, Lish SR, Taylor NA, Krueger JG, Ferrer M, Carucci JA, Gareau DS. A 3D biofabricated cutaneous squamous cell carcinoma tissue model with multi-channel confocal microscopy imaging biomarkers to quantify antitumor effects of chemotherapeutics in tissue. Oncotarget 2020; 11:2587-2596. [PMID: 32676161 PMCID: PMC7343636 DOI: 10.18632/oncotarget.27570] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 04/03/2020] [Indexed: 11/25/2022] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) causes approximately 10,000 deaths annually in the U. S. Current therapies are largely ineffective against metastatic and locally advanced cSCC. There is a need to identify novel, effective, and less toxic small molecule cSCC therapeutics. We developed a 3-dimensional bioprinted skin (3DBPS) model of cSCC tumors together with a microscopy assay to test chemotherapeutic effects in tissue. The full thickness SCC tissue model was validated using hematoxylin and eosin (H&E) and immunohistochemical histological staining, confocal microscopy, and cDNA microarray analysis. A nondestructive, 3D fluorescence confocal imaging assay with tdTomato-labeled A431 SCC and ZsGreen-labeled keratinocytes was developed to test efficacy and general toxicity of chemotherapeutics. Fluorescence-derived imaging biomarkers indicated that 50% of cancer cells were killed in the tissue after 1μM 5-Fluorouracil 48-hour treatment, compared to a baseline of 12% for untreated controls. The imaging biomarkers also showed that normal keratinocytes were less affected by treatment (11% killed) than the untreated tissue, which had no significant killing effect. Data showed that 5-Fluorouracil selectively killed cSCC cells more than keratinocytes. Our 3DBPS assay platform provides cellular-level measurement of cell viability and can be adapted to achieve nondestructive high-throughput screening (HTS) in bio-fabricated tissues.
Collapse
Affiliation(s)
- James R Browning
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA
| | - Paige Derr
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Kristy Derr
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Nicole Doudican
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York, USA
| | - Sam Michael
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Samantha R Lish
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA
| | - Nicholas A Taylor
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York, USA
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA
| | - Marc Ferrer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - John A Carucci
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York, USA
| | - Daniel S Gareau
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA
| |
Collapse
|
328
|
|
329
|
Wang K, Man K, Liu J, Liu Y, Chen Q, Zhou Y, Yang Y. Microphysiological Systems: Design, Fabrication, and Applications. ACS Biomater Sci Eng 2020; 6:3231-3257. [PMID: 33204830 PMCID: PMC7668566 DOI: 10.1021/acsbiomaterials.9b01667] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Microphysiological systems, including organoids, 3-D printed tissue constructs and organ-on-a-chips (organ chips), are physiologically relevant in vitro models and have experienced explosive growth in the past decades. Different from conventional, tissue culture plastic-based in vitro models or animal models, microphysiological systems recapitulate key microenvironmental characteristics of human organs and mimic their primary functions. The advent of microphysiological systems is attributed to evolving biomaterials, micro-/nanotechnologies and stem cell biology, which enable the precise control over the matrix properties and the interactions between cells, tissues and organs in physiological conditions. As such, microphysiological systems have been developed to model a broad spectrum of organs from microvasculature, eye, to lung and many others to understand human organ development and disease pathology and facilitate drug discovery. Multiorgans-on-a-chip systems have also been developed by integrating multiple associated organ chips in a single platform, which allows to study and employ the organ function in a systematic approach. Here we first discuss the design principles of microphysiological systems with a focus on the anatomy and physiology of organs, and then review the commonly used fabrication techniques and biomaterials for microphysiological systems. Subsequently, we discuss the recent development of microphysiological systems, and provide our perspectives on advancing microphysiological systems for preclinical investigation and drug discovery of human disease.
Collapse
Affiliation(s)
- Kai Wang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Kun Man
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Jiafeng Liu
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Yang Liu
- North Texas Eye Research Institute, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas 76107, United States
| | - Qi Chen
- The Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Yong Zhou
- Department of Emergency, Xinqiao Hospital, Chongqing 400037, China
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| |
Collapse
|
330
|
Cao Y, Zhao H, Hu Z, Ma S. Cascade Pumping Overcomes Hydraulic Resistance and Moderates Shear Conditions for Slow Gelatin Fiber Shaping in Narrow Tubes. iScience 2020; 23:101228. [PMID: 32540773 PMCID: PMC7298654 DOI: 10.1016/j.isci.2020.101228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/06/2020] [Accepted: 05/28/2020] [Indexed: 11/30/2022] Open
Abstract
In microextrusion-based 3D bioprinting, shaping gel fibers online, i.e., in narrow tubes, benefits the structural maintenance after extrusion, but it is challenging for materials possessing slow sol-gel transition dynamics. Gelatin, for example, transforms into thermostable fibers via transglutaminase (TG) reaction in as much as 10 min. It causes dramatic flow resistance accumulation and shear stress increase in fluids moving along narrow tubes, resulting in channel clogging and cell detriments. In this study, we overcome the limitations by adopting cascade pumping and performing in a single peristaltic pump that comprises multi-channel pumping units. The pressure and shear stress reduction by over 1-fold are verified by finite element simulation; continuous gelatin fiber production and patterning in a substrate-free manner are achieved via slow online enzymatic cross-linking. The online fiber shaping can be scaled up by numbering up the pumping units and provides another paradigm for biomanufacturing. Cascade pumping overcomes hydraulic resistance and reduces shear stress significantly Cascade pumping suits online gelatin fiber shaping via slow enzymatic cross-linking Online fiber shaping enables substrate-free 3D printing Gelatin fibers are stronger when gelled via thermal and enzymatic dual cross-linking
Collapse
Affiliation(s)
- Yuanxiong Cao
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen, 518055, China; Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, 518055, China
| | - Haoran Zhao
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen, 518055, China; Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, 518055, China
| | - Zhiwei Hu
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen, 518055, China; Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, 518055, China
| | - Shaohua Ma
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen, 518055, China; Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, 518055, China.
| |
Collapse
|
331
|
Sun H, Jia Y, Dong H, Dong D, Zheng J. Combining additive manufacturing with microfluidics: an emerging method for developing novel organs-on-chips. Curr Opin Chem Eng 2020. [DOI: 10.1016/j.coche.2019.10.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
332
|
3D bioprinted hyaluronic acid-based cell-laden scaffold for brain microenvironment simulation. Biodes Manuf 2020. [DOI: 10.1007/s42242-020-00076-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
333
|
Ferreira LP, Gaspar VM, Mano JF. Decellularized Extracellular Matrix for Bioengineering Physiomimetic 3D in Vitro Tumor Models. Trends Biotechnol 2020; 38:1397-1414. [PMID: 32416940 DOI: 10.1016/j.tibtech.2020.04.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/10/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023]
Abstract
Recent advances in the extraction and purification of decellularized extracellular matrix (dECM) obtained from healthy or malignant tissues open new avenues for engineering physiomimetic 3D in vitro tumor models, which closely recapitulate key biomolecular hallmarks and the dynamic cancer cell-ECM interactions in the tumor microenvironment. We review current and upcoming methodologies for chemical modification of dECM-based biomaterials and advanced bioprocessing into organotypic 3D solid tumor models. A comprehensive review of disruptive advances and shortcomings of exploring dECM-based biomaterials for recapitulating the native tumor-supporting matrix is also provided. We hope to drive the discussion on how 3D dECM testing platforms can be leveraged for generating microphysiological tumor surrogates that generate more robust and predictive data on therapeutic bioperformance.
Collapse
Affiliation(s)
- Luís P Ferreira
- Department of Chemistry, CICECO, Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Vítor M Gaspar
- Department of Chemistry, CICECO, Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO, Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
334
|
Zhao Y, Demirci U, Chen Y, Chen P. Multiscale brain research on a microfluidic chip. LAB ON A CHIP 2020; 20:1531-1543. [PMID: 32150176 DOI: 10.1039/c9lc01010f] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
One major challenge in current brain research is generating an integrative understanding of the brain's functions and disorders from its multiscale neuronal architectures and connectivity. Thus, innovative neurotechnology tools are urgently required for deciphering the multiscale functional and structural organizations of the brain at hierarchical scales from the molecular to the organismal level by multiple brain research initiatives launched by the European Union, United States, Australia, Canada, China, Korea, and Japan. To meet this demand, microfluidic chips (μFCs) have rapidly evolved as a trans-scale neurotechnological toolset to enable multiscale studies of the brain due to their unique advantages in flexible microstructure design, multifunctional integration, accurate microenvironment control, and capacity for automatic sample processing. Here, we review the recent progress in applying innovative μFC-based neuro-technologies to promote multiscale brain research and uniquely focus on representative applications of μFCs to address challenges in brain research at each hierarchical level. We discuss the current trend of combinational applications of μFCs with other neuro- and biotechnologies, including optogenetics, brain organoids, and 3D bioprinting, for better multiscale brain research. In addition, we offer our insights into the existing outstanding questions at each hierarchical level of brain research that could potentially be addressed by advancing microfluidic techniques. This review will serve as a timely guide for bioengineers and neuroscientists to develop and apply μFC-based neuro-technologies for promoting basic and translational brain research.
Collapse
Affiliation(s)
- Yanan Zhao
- Department of Biomedical Engineering, Wuhan University School of Basic Medical Sciences, 115 Donghu Road, Wuhan 430071, China.
| | | | | | | |
Collapse
|
335
|
Bhattacharya S, Calar K, de la Puente P. Mimicking tumor hypoxia and tumor-immune interactions employing three-dimensional in vitro models. J Exp Clin Cancer Res 2020; 39:75. [PMID: 32357910 PMCID: PMC7195738 DOI: 10.1186/s13046-020-01583-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023] Open
Abstract
The heterogeneous tumor microenvironment (TME) is highly complex and not entirely understood. These complex configurations lead to the generation of oxygen-deprived conditions within the tumor niche, which modulate several intrinsic TME elements to promote immunosuppressive outcomes. Decoding these communications is necessary for designing effective therapeutic strategies that can effectively reduce tumor-associated chemotherapy resistance by employing the inherent potential of the immune system.While classic two-dimensional in vitro research models reveal critical hypoxia-driven biochemical cues, three-dimensional (3D) cell culture models more accurately replicate the TME-immune manifestations. In this study, we review various 3D cell culture models currently being utilized to foster an oxygen-deprived TME, those that assess the dynamics associated with TME-immune cell penetrability within the tumor-like spatial structure, and discuss state of the art 3D systems that attempt recreating hypoxia-driven TME-immune outcomes. We also highlight the importance of integrating various hallmarks, which collectively might influence the functionality of these 3D models.This review strives to supplement perspectives to the quickly-evolving discipline that endeavors to mimic tumor hypoxia and tumor-immune interactions using 3D in vitro models.
Collapse
Affiliation(s)
- Somshuvra Bhattacharya
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 E 60th Street N, Sioux Falls, SD, 57104, USA
| | - Kristin Calar
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 E 60th Street N, Sioux Falls, SD, 57104, USA
| | - Pilar de la Puente
- Cancer Biology and Immunotherapies Group, Sanford Research, 2301 E 60th Street N, Sioux Falls, SD, 57104, USA.
- Department of Surgery, University of South Dakota Sanford School of Medicine, Sioux Falls, SD, USA.
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, USA.
| |
Collapse
|
336
|
Decellularized matrices for tumor cell modeling. Methods Cell Biol 2020. [PMID: 32334714 DOI: 10.1016/bs.mcb.2019.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Collagen is the main component of the extracellular matrix and it plays a key role in tumor progression. Commercial collagen solutions are derived from animals, such as rat-tail and bovine or porcine skin. Their cost is quite high and the product is stable only at low temperature, with the disadvantage of a short expiring date. Most importantly, lot-to-lot variability can occur and the reconstituted collagen gels differ significantly from native tissues in terms of both structure and stiffness. In this chapter, we describe a straightforward method to use native, collagen rich skin samples derived from by-products of the tanning industry. The protocol proposed preserves the microstructure of the ovine skin collagen network, offering structurally competent and more relevant model to investigate cell behavior in vitro. Other advantages of the proposed procedure consist in the cost-effectiveness of the process and an increased level of reproducibility. The decellularized ovine skin samples support the adhesion and growth of different cancer cell lines (pancreatic, breast and melanoma cells). The proposed decellularized skin scaffolds are meant as future low-cost competitors for conventional porous scaffold derived by biomaterials, since they offer a biomimetic environment for the cells.
Collapse
|
337
|
|
338
|
Azzarelli R. Organoid Models of Glioblastoma to Study Brain Tumor Stem Cells. Front Cell Dev Biol 2020; 8:220. [PMID: 32373607 PMCID: PMC7176979 DOI: 10.3389/fcell.2020.00220] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/16/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma represents an aggressive form of brain cancer characterized by poor prognosis and a 5-year survival rate of only 3–7%. Despite remarkable advances in brain tumor research in the past decades, very little has changed for patients, due in part to the recurrent nature of the disease and to the lack of suitable models to perform genotype-phenotype association studies and personalized drug screening. In vitro culture of cancer cells derived from patient biopsies has been fundamental in understanding tumor biology and for testing the effect of various drugs. These cultures emphasize the role of in vitro cancer stem cells (CSCs), which fuel tumor growth and are thought to be the cause of relapse after treatment. However, it has become clear over the years that a 2D monolayer culture of these CSCs has certain disadvantages, including the lack of heterogeneous cell-cell and cell-environment interactions, which can now be partially overcome by the introduction of 3D organoid cultures. This is a novel and expanding field of research and in this review, I describe the emerging 3D models of glioblastoma. I also discuss their potential to advance our knowledge of tumor biology and CSC heterogeneity, while debating their current limitations.
Collapse
Affiliation(s)
- Roberta Azzarelli
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| |
Collapse
|
339
|
Nie J, Gao Q, Fu J, He Y. Grafting of 3D Bioprinting to In Vitro Drug Screening: A Review. Adv Healthc Mater 2020; 9:e1901773. [PMID: 32125787 DOI: 10.1002/adhm.201901773] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/13/2020] [Accepted: 02/17/2020] [Indexed: 01/09/2023]
Abstract
The inadequacy of conventional cell-monolayer planar cultures and animal experiments in predicting the toxicity and clinical efficacy of drug candidates has led to an imminent need for in vitro methods with the ability to better represent in vivo conditions and facilitate the systematic investigation of drug candidates. Recent advances in 3D bioprinting have prompted the precise manipulation of cells and biomaterials, rendering it a promising technology for the construction of in vitro tissue/organ models and drug screening devices. This review presents state-of-the-art in vitro methods used for preclinical drug screening and discusses the limitations of these methods. In particular, the significance of constructing 3D in vitro tissue/organ models and microfluidic analysis devices for drug screening is emphasized, and a focus is placed on the grafting process of 3D bioprinting technology to the construction of such models and devices. The in vitro methods for drug screening are generalized into three types: mini-tissue, organ-on-a-chip, and tissue/organ construct. The revolutionary process of the in vitro methods is demonstrated in detail, and relevant studies are listed as examples. Specifically, the tumor model is adopted as a precedent to illustrate the possible grafting of 3D bioprinting to antitumor drug screening.
Collapse
Affiliation(s)
- Jing Nie
- State Key Laboratory of Fluid Power and Mechatronic SystemsSchool of Mechanical EngineeringZhejiang University Hangzhou 310027 China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang ProvinceSchool of Mechanical EngineeringZhejiang University Hangzhou 310027 China
| | - Qing Gao
- State Key Laboratory of Fluid Power and Mechatronic SystemsSchool of Mechanical EngineeringZhejiang University Hangzhou 310027 China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang ProvinceSchool of Mechanical EngineeringZhejiang University Hangzhou 310027 China
| | - Jianzhong Fu
- State Key Laboratory of Fluid Power and Mechatronic SystemsSchool of Mechanical EngineeringZhejiang University Hangzhou 310027 China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang ProvinceSchool of Mechanical EngineeringZhejiang University Hangzhou 310027 China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic SystemsSchool of Mechanical EngineeringZhejiang University Hangzhou 310027 China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang ProvinceSchool of Mechanical EngineeringZhejiang University Hangzhou 310027 China
| |
Collapse
|
340
|
Kang D, Hong G, An S, Jang I, Yun WS, Shim JH, Jin S. Bioprinting of Multiscaled Hepatic Lobules within a Highly Vascularized Construct. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1905505. [PMID: 32078240 DOI: 10.1002/smll.201905505] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/25/2020] [Indexed: 06/10/2023]
Abstract
Highly vascularized complex liver tissue is generally divided into lobes, lobules, hepatocytes, and sinusoids, which can be viewed under different types of lens from the micro- to macro-scale. To engineer multiscaled heterogeneous tissues, a sophisticated and rapid tissue engineering approach is required, such as advanced 3D bioprinting. In this study, a preset extrusion bioprinting technique, which can create heterogeneous, multicellular, and multimaterial structures simultaneously, is utilized for creating a hepatic lobule (≈1 mm) array. The fabricated hepatic lobules include hepatic cells, endothelial cells, and a lumen. The endothelial cells surround the hepatic cells, the exterior of the lobules, the lumen, and finally, become interconnected with each other. Compared to hepatic cell/endothelial cell mixtures, the fabricated hepatic lobule shows higher albumin secretion, urea production, and albumin, MRP2, and CD31 protein levels, as well as, cytochrome P450 enzyme activity. It is found that each cell type with spatial cell patterning in bioink accelerates cellular organization, which could preserve structural integrity and improve cellular functions. In conclusion, preset extruded hepatic lobules within a highly vascularized construct are successfully constructed, enabling both micro- and macro-scale tissue fabrication, which can support the creation of large 3D tissue constructs for multiscale tissue engineering.
Collapse
Affiliation(s)
- Donggu Kang
- Research Institute, T&R Biofab. Co. Ltd., 242 Pangyo-ro, Seongnam-si, 13487, Republic of Korea
| | - Gyusik Hong
- Department of Mechanical Engineering, Korea Polytechnic University, 237 Sangidaehak-ro, Siheung-si, 15073, Republic of Korea
| | - Seongmin An
- Department of Mechanical Engineering, Korea Polytechnic University, 237 Sangidaehak-ro, Siheung-si, 15073, Republic of Korea
| | - Ilho Jang
- Research Institute, T&R Biofab. Co. Ltd., 242 Pangyo-ro, Seongnam-si, 13487, Republic of Korea
| | - Won-Soo Yun
- Department of Mechanical Engineering, Korea Polytechnic University, 237 Sangidaehak-ro, Siheung-si, 15073, Republic of Korea
| | - Jin-Hyung Shim
- Department of Mechanical Engineering, Korea Polytechnic University, 237 Sangidaehak-ro, Siheung-si, 15073, Republic of Korea
| | - Songwan Jin
- Department of Mechanical Engineering, Korea Polytechnic University, 237 Sangidaehak-ro, Siheung-si, 15073, Republic of Korea
| |
Collapse
|
341
|
|
342
|
Liu Y, Zou Y, Feng C, Lee A, Yin J, Chung R, Park JB, Rizos H, Tao W, Zheng M, Farokhzad OC, Shi B. Charge Conversional Biomimetic Nanocomplexes as a Multifunctional Platform for Boosting Orthotopic Glioblastoma RNAi Therapy. NANO LETTERS 2020; 20:1637-1646. [PMID: 32013452 DOI: 10.1021/acs.nanolett.9b04683] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Nanotechnology-based RNA interference (RNAi) has shown great promise in overcoming the limitations of traditional clinical treatments for glioblastoma (GBM). However, because of the complexity of brain physiology, simple blood-brain barrier (BBB) penetration or tumor-targeting strategies cannot entirely meet the demanding requirements of different therapeutic delivery stages. Herein, we developed a charge conversional biomimetic nanoplatform with a three-layer core-shell structure to programmatically overcome persistent obstacles in siRNA delivery to GBM. The resulting nanocomplex presents good biocompatibility, prolonged blood circulation, high BBB transcytosis, effective tumor accumulation, and specific uptake by tumor cells in the brain. Moreover, red blood cell membrane (RBCm) disruption and effective siRNA release can be further triggered elegantly by charge conversion from negative to positive in the endo/lysosome (pH 5.0-6.5) of tumor cells, leading to highly potent target-gene silencing with a strong anti-GBM effect. Our study provides an intelligent biomimetic nanoplatform tailored for systemically siRNA delivery to GBM, leveraging Angiopep-2 peptide-modified, immune-free RBCm and charge conversional components. Improved therapeutic efficacy, higher survival rates, and minimized systemic side effects were achieved in orthotopic U87MG-luc human glioblastoma tumor-bearing nude mice.
Collapse
Affiliation(s)
- Yanjie Liu
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yan Zou
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Chan Feng
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Albert Lee
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Jinlong Yin
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, South Korea
| | - Roger Chung
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Jong Bea Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, South Korea
| | - Helen Rizos
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Meng Zheng
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Bingyang Shi
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
343
|
Huang NF, Chaudhuri O, Cahan P, Wang A, Engler AJ, Wang Y, Kumar S, Khademhosseini A, Li S. Multi-scale cellular engineering: From molecules to organ-on-a-chip. APL Bioeng 2020; 4:010906. [PMID: 32161833 PMCID: PMC7054123 DOI: 10.1063/1.5129788] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 01/28/2020] [Indexed: 12/11/2022] Open
Abstract
Recent technological advances in cellular and molecular engineering have provided new
insights into biology and enabled the design, manufacturing, and manipulation of complex
living systems. Here, we summarize the state of advances at the molecular, cellular, and
multi-cellular levels using experimental and computational tools. The areas of focus
include intrinsically disordered proteins, synthetic proteins, spatiotemporally dynamic
extracellular matrices, organ-on-a-chip approaches, and computational modeling, which all
have tremendous potential for advancing fundamental and translational science.
Perspectives on the current limitations and future directions are also described, with the
goal of stimulating interest to overcome these hurdles using multi-disciplinary
approaches.
Collapse
Affiliation(s)
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, California 94305, USA
| | - Patrick Cahan
- Department of Biomedical Engineering, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | - Adam J Engler
- Department of Bioengineering, Jacob School of Engineering, University of California San Diego, La Jolla, California 92093, USA
| | - Yingxiao Wang
- Department of Bioengineering, Jacob School of Engineering, University of California San Diego, La Jolla, California 92093, USA
| | | | | | - Song Li
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
344
|
Levato R, Jungst T, Scheuring RG, Blunk T, Groll J, Malda J. From Shape to Function: The Next Step in Bioprinting. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1906423. [PMID: 32045053 PMCID: PMC7116209 DOI: 10.1002/adma.201906423] [Citation(s) in RCA: 226] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/08/2019] [Indexed: 05/04/2023]
Abstract
In 2013, the "biofabrication window" was introduced to reflect the processing challenge for the fields of biofabrication and bioprinting. At that time, the lack of printable materials that could serve as cell-laden bioinks, as well as the limitations of printing and assembly methods, presented a major constraint. However, recent developments have now resulted in the availability of a plethora of bioinks, new printing approaches, and the technological advancement of established techniques. Nevertheless, it remains largely unknown which materials and technical parameters are essential for the fabrication of intrinsically hierarchical cell-material constructs that truly mimic biologically functional tissue. In order to achieve this, it is urged that the field now shift its focus from materials and technologies toward the biological development of the resulting constructs. Therefore, herein, the recent material and technological advances since the introduction of the biofabrication window are briefly summarized, i.e., approaches how to generate shape, to then focus the discussion on how to acquire the biological function within this context. In particular, a vision of how biological function can evolve from the possibility to determine shape is outlined.
Collapse
Affiliation(s)
- Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - Tomasz Jungst
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Ruben G Scheuring
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Torsten Blunk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Juergen Groll
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CX, Utrecht, The Netherlands
| |
Collapse
|
345
|
Papadimitriou L, Manganas P, Ranella A, Stratakis E. Biofabrication for neural tissue engineering applications. Mater Today Bio 2020; 6:100043. [PMID: 32190832 PMCID: PMC7068131 DOI: 10.1016/j.mtbio.2020.100043] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/28/2022] Open
Abstract
Unlike other tissue types, the nervous tissue extends to a wide and complex environment that provides a plurality of different biochemical and topological stimuli, which in turn defines the advanced functions of that tissue. As a consequence of such complexity, the traditional transplantation therapeutic methods are quite ineffective; therefore, the restoration of peripheral and central nervous system injuries has been a continuous scientific challenge. Tissue engineering and regenerative medicine in the nervous system have provided new alternative medical approaches. These methods use external biomaterial supports, known as scaffolds, to create platforms for the cells to migrate to the injury site and repair the tissue. The challenge in neural tissue engineering (NTE) remains the fabrication of scaffolds with precisely controlled, tunable topography, biochemical cues, and surface energy, capable of directing and controlling the function of neuronal cells toward the recovery from neurological disorders and injuries. At the same time, it has been shown that NTE provides the potential to model neurological diseases in vitro, mainly via lab-on-a-chip systems, especially in cases for which it is difficult to obtain suitable animal models. As a consequence of the intense research activity in the field, a variety of synthetic approaches and 3D fabrication methods have been developed for the fabrication of NTE scaffolds, including soft lithography and self-assembly, as well as subtractive (top-down) and additive (bottom-up) manufacturing. This article aims at reviewing the existing research effort in the rapidly growing field related to the development of biomaterial scaffolds and lab-on-a-chip systems for NTE applications. Besides presenting recent advances achieved by NTE strategies, this work also delineates existing limitations and highlights emerging possibilities and future prospects in this field.
Collapse
Affiliation(s)
- L. Papadimitriou
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
| | - P. Manganas
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
| | - A. Ranella
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
| | - E. Stratakis
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
- Physics Department, University of Crete, Heraklion, 71003, Crete, Greece
| |
Collapse
|
346
|
Sheykhzadeh S, Luo M, Peng B, White J, Abdalla Y, Tang T, Mäkilä E, Voelcker NH, Tong WY. Transferrin-targeted porous silicon nanoparticles reduce glioblastoma cell migration across tight extracellular space. Sci Rep 2020; 10:2320. [PMID: 32047170 PMCID: PMC7012928 DOI: 10.1038/s41598-020-59146-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 01/24/2020] [Indexed: 02/07/2023] Open
Abstract
Mortality of glioblastoma multiforme (GBM) has not improved over the last two decades despite medical breakthroughs in the treatment of other types of cancers. Nanoparticles hold tremendous promise to overcome the pharmacokinetic challenges and off-target adverse effects. However, an inhibitory effect of nanoparticles by themselves on metastasis has not been explored. In this study, we developed transferrin-conjugated porous silicon nanoparticles (Tf@pSiNP) and studied their effect on inhibiting GBM migration by means of a microfluidic-based migration chip. This platform, designed to mimic the tight extracellular migration tracts in brain parenchyma, allowed high-content time-resolved imaging of cell migration. Tf@pSiNP were colloidally stable, biocompatible, and their uptake into GBM cells was enhanced by receptor-mediated internalisation. The migration of Tf@pSiNP-exposed cells across the confined microchannels was suppressed, but unconfined migration was unaffected. The pSiNP-induced destabilisation of focal adhesions at the leading front may partially explain the migration inhibition. More corroborating evidence suggests that pSiNP uptake reduced the plasticity of GBM cells in reducing cell volume, an effect that proved crucial in facilitating migration across the tight confined tracts. We believe that the inhibitory effect of Tf@pSiNP on cell migration, together with the drug-delivery capability of pSiNP, could potentially offer a disruptive strategy to treat GBM.
Collapse
Affiliation(s)
- Sana Sheykhzadeh
- Department of Pharmaceutical and Biological Chemistry, UCL School of Pharmacy, University College London, Brunswick Square, London, United Kingdom
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutics Science, Monash University, Parkville, Victoria, Australia
| | - Meihua Luo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutics Science, Monash University, Parkville, Victoria, Australia
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Bo Peng
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutics Science, Monash University, Parkville, Victoria, Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO), Clayton, Victoria, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia
| | - Jacinta White
- Commonwealth Scientific and Industrial Research Organization (CSIRO), Clayton, Victoria, Australia
| | - Youssef Abdalla
- Department of Pharmaceutical and Biological Chemistry, UCL School of Pharmacy, University College London, Brunswick Square, London, United Kingdom
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutics Science, Monash University, Parkville, Victoria, Australia
| | - Tweety Tang
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutics Science, Monash University, Parkville, Victoria, Australia
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong
| | - Ermei Mäkilä
- Industrial Physics Laboratory, Department of Physics and Astronomy, University of Turku, Turku, Finland
| | - Nicolas H Voelcker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutics Science, Monash University, Parkville, Victoria, Australia.
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
- Commonwealth Scientific and Industrial Research Organization (CSIRO), Clayton, Victoria, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia.
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia.
| | - Wing Yin Tong
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutics Science, Monash University, Parkville, Victoria, Australia.
- Commonwealth Scientific and Industrial Research Organization (CSIRO), Clayton, Victoria, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, Australia.
| |
Collapse
|
347
|
Lin L, Yi L, Zhao F, Wu Z, Zheng Y, Li N, Lin JM, Sun J. ATP-responsive mitochondrial probes for monitoring metabolic processes of glioma stem cells in a 3D model. Chem Sci 2020; 11:2744-2749. [PMID: 34084333 PMCID: PMC8157640 DOI: 10.1039/c9sc06185a] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/03/2020] [Indexed: 02/05/2023] Open
Abstract
The metastatic cascade of cancer stem cells (CSCs) is always accompanied by elevated levels of adenosine triphosphate (ATP) as well as the alterntion of energy metabolism to support their differentiation and migration. Here we propose a 3D microfluidic tumor model coupled with an ATP-responsive mitochondrial probe (AMP) for investigation of metabolic processes of glioma stem cells (GSCs). The 3D tumor model has a middle matrix gel microchannel mimicking the extracellular matrix (ECM), which is sandwiched between a GSC culture chamber and a stimulation chamber. The AMPs consist of structure-switching ATP aptamers and triphenylphosphonium (TPP)-conjugated peptide nucleic acids (PNAs). Under TGF-β stimulation, invasive migration of GSCs accompanied by a high ATP level and spindle mesenchymal morphologies is observed due to the epithelial-to-mesenchymal transition (EMT). Moreover, acidic stress can keep GSCs in a low-energy state, while long-term low pH stimulation screens out more malignant glioma cells. This AMP-assisted 3D microfluidic tumor model provides a tremendous opportunity for studying the biological properties of CSCs.
Collapse
Affiliation(s)
- Ling Lin
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 China
| | - Linglu Yi
- State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, West China Medical School, Sichuan University, Collaborative Innovation Center of Biotherapy Chengdu Sichuan 610064 China
| | - Fanghao Zhao
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 China
| | - Zengnan Wu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 China
| | - Yajing Zheng
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 China
| | - Nan Li
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 China
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University Beijing 100084 China
| | - Jin-Ming Lin
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University Beijing 100084 China
| | - Jiashu Sun
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing 100190 China
| |
Collapse
|
348
|
Raimondi I, Izzo L, Tunesi M, Comar M, Albani D, Giordano C. Organ-On-A-Chip in vitro Models of the Brain and the Blood-Brain Barrier and Their Value to Study the Microbiota-Gut-Brain Axis in Neurodegeneration. Front Bioeng Biotechnol 2020; 7:435. [PMID: 31998702 PMCID: PMC6965718 DOI: 10.3389/fbioe.2019.00435] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 12/05/2019] [Indexed: 12/21/2022] Open
Abstract
We are accumulating evidence that intestinal microflora, collectively named gut microbiota, can alter brain pathophysiology, but researchers have just begun to discover the mechanisms of this bidirectional connection (often referred to as microbiota-gut-brain axis, MGBA). The most noticeable hypothesis for a pathological action of gut microbiota on the brain is based on microbial release of soluble neurotransmitters, hormones, immune molecules and neuroactive metabolites, but this complex scenario requires reliable and controllable tools for its causal demonstration. Thanks to three-dimensional (3D) cultures and microfluidics, engineered in vitro models could improve the scientific knowledge in this field, also from a therapeutic perspective. This review briefly retraces the main discoveries linking the activity of gut microbiota to prevalent brain neurodegenerative disorders, and then provides a deep insight into the state-of-the-art for in vitro modeling of the brain and the blood-brain barrier (BBB), two key players of the MGBA. Several brain and BBB microfluidic devices have already been developed to implement organ-on-a-chip solutions, but some limitations still exist. Future developments of organ-on-a-chip tools to model the MGBA will require an interdisciplinary approach and the synergy with cutting-edge technologies (for instance, bioprinting) to achieve multi-organ platforms and support basic research, also for the development of new therapies against neurodegenerative diseases.
Collapse
Affiliation(s)
- Ilaria Raimondi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Luca Izzo
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Marta Tunesi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Manola Comar
- SSD of Advanced Translational Microbiology, IRCCS “Burlo Garofolo”, Department of Medical Sciences (DMS), University of Trieste, Trieste, Italy
| | - Diego Albani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| |
Collapse
|
349
|
Peck RW, Hinojosa CD, Hamilton GA. Organs-on-Chips in Clinical Pharmacology: Putting the Patient Into the Center of Treatment Selection and Drug Development. Clin Pharmacol Ther 2020; 107:181-185. [PMID: 31758803 PMCID: PMC6977308 DOI: 10.1002/cpt.1688] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022]
Abstract
There have been rapid advances since Organs-on-Chips were first developed. Organ-Chips are now available beyond academic laboratories with the initial emphasis to reduce animal experimentation and improve predictability of drug development through better prediction of safety and efficacy. There is now a huge opportunity to use chips to understand efficacy and disease variability. We propose that by 2030, Organs-on-Chips will play a key role in clinical pharmacology as part of the diagnostic and treatment workflow for some diseases by informing the right drug and dose regimen for each patient.
Collapse
Affiliation(s)
- Richard W. Peck
- Pharma Research & Early Development (pRED)Roche Innovation Center BaselBaselSwitzerland
| | | | | |
Collapse
|
350
|
Brancato V, Oliveira JM, Correlo VM, Reis RL, Kundu SC. Could 3D models of cancer enhance drug screening? Biomaterials 2019; 232:119744. [PMID: 31918229 DOI: 10.1016/j.biomaterials.2019.119744] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/29/2019] [Accepted: 12/25/2019] [Indexed: 02/06/2023]
Abstract
Cancer is a multifaceted pathology, where cellular and acellular players interact to drive cancer progression and, in the worst-case, metastasis. The current methods to investigate the heterogeneous nature of cancer are inadequate, since they rely on 2D cell cultures and animal models. The cell line-based drug efficacy and toxicity assays are not able to predict the tumor response to anti-cancer agents and it is already widely discussed how molecular pathway are not recapitulated in vitro so called flat biology. On the other side, animal models often fail to detect the side-effects of drugs, mimic the metastatic progression or the interaction between cancer and immune system, due to biologic difference in human and animals. Moreover, ethical and regulatory issues limit animal experimentation. Every year pharma/biotech companies lose resources in drug discovery and testing processes that are successful only in 5% of the cases. There is an urgent need to validate accurate and predictive platforms in order to enhance drug-testing process taking into account the physiopathology of the tumor microenvironment. Three dimensional in vitro tumor models could enhance drug manufactures in developing effective drugs for cancer diseases. The 3D in vitro cancer models can improve the predictability of toxicity and drug sensitivity in cancer. Despite the demonstrated advantages of 3D in vitro disease systems when compared to 2D culture and animal models, they still do not reach the standardization required for preclinical trials. This review highlights in vitro models that may be used as preclinical models, accelerating the drug development process towards more precise and personalized standard of care for cancer patients. We describe the state-of-the art of 3D in vitro culture systems, with a focus on how these different approaches could be coupled in order to achieve a compromise between standardization and reliability in recapitulating tumor microenvironment and drug response.
Collapse
Affiliation(s)
- Virginia Brancato
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| | - Joaquim Miguel Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Vitor Manuel Correlo
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Rui Luis Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Subhas C Kundu
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|