151
|
Krentz NAJ, Shea LD, Huising MO, Shaw JAM. Restoring normal islet mass and function in type 1 diabetes through regenerative medicine and tissue engineering. Lancet Diabetes Endocrinol 2021; 9:708-724. [PMID: 34480875 PMCID: PMC10881068 DOI: 10.1016/s2213-8587(21)00170-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/17/2021] [Accepted: 06/08/2021] [Indexed: 02/09/2023]
Abstract
Type 1 diabetes is characterised by autoimmune-mediated destruction of pancreatic β-cell mass. With the advent of insulin therapy a century ago, type 1 diabetes changed from a progressive, fatal disease to one that requires lifelong complex self-management. Replacing the lost β-cell mass through transplantation has proven successful, but limited donor supply and need for lifelong immunosuppression restricts widespread use. In this Review, we highlight incremental advances over the past 20 years and remaining challenges in regenerative medicine approaches to restoring β-cell mass and function in type 1 diabetes. We begin by summarising the role of endocrine islets in glucose homoeostasis and how this is altered in disease. We then discuss the potential regenerative capacity of the remaining islet cells and the utility of stem cell-derived β-like cells to restore β-cell function. We conclude with tissue engineering approaches that might improve the engraftment, function, and survival of β-cell replacement therapies.
Collapse
Affiliation(s)
- Nicole A J Krentz
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Lonnie D Shea
- Departments of Biomedical Engineering, Chemical Engineering, and Surgery, College of Engineering and School of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mark O Huising
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, Davis, CA, USA; Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA, USA
| | - James A M Shaw
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
152
|
Feng Y, Quinnell SP, Lanzi AM, Vegas AJ. Alginate-Based Amphiphilic Block Copolymers as a Drug Codelivery Platform. NANO LETTERS 2021; 21:7495-7504. [PMID: 34495662 PMCID: PMC8768502 DOI: 10.1021/acs.nanolett.1c01525] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Structured nanoassemblies are biomimetic structures that are enabling applications from nanomedicine to catalysis. One approach to achieve these spatially organized architectures is utilizing amphiphilic diblock copolymers with one or two macromolecular backbones that self-assemble in solution. To date, the impact of alternating backbone architectures on self-assembly and drug delivery is still an area of active research limited by the strategies used to synthesize these multiblock polymers. Here, we report self-assembling ABC-type alginate-based triblock copolymers with the backbones of three distinct biomaterials utilizing a facile conjugation approach. This "polymer mosaic" was synthesized by the covalent attachment of alginate with a PLA/PEG diblock copolymer. The combination of alginate, PEG, and PLA domains resulted in an amphiphilic copolymer that self-assembles into nanoparticles with a unique morphology of alginate domain compartmentalization. These particles serve as a versatile platform for co-encapsulation of hydrophilic and hydrophobic small molecules, their spatiotemporal release, and show potential as a drug delivery system for combination therapy.
Collapse
Affiliation(s)
- Yunpeng Feng
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States
| | - Sean P. Quinnell
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Alison M. Lanzi
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States
| | - Arturo J. Vegas
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
- Corresponding Author: Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States;
| |
Collapse
|
153
|
Whitaker R, Hernaez-Estrada B, Hernandez RM, Santos-Vizcaino E, Spiller KL. Immunomodulatory Biomaterials for Tissue Repair. Chem Rev 2021; 121:11305-11335. [PMID: 34415742 DOI: 10.1021/acs.chemrev.0c00895] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
All implanted biomaterials are targets of the host's immune system. While the host inflammatory response was once considered a detrimental force to be blunted or avoided, in recent years, it has become a powerful force to be leveraged to augment biomaterial-tissue integration and tissue repair. In this review, we will discuss the major immune cells that mediate the inflammatory response to biomaterials, with a focus on how biomaterials can be designed to modulate immune cell behavior to promote biomaterial-tissue integration. In particular, the intentional activation of monocytes and macrophages with controlled timing, and modulation of their interactions with other cell types involved in wound healing, have emerged as key strategies to improve biomaterial efficacy. To this end, careful design of biomaterial structure and controlled release of immunomodulators can be employed to manipulate macrophage phenotype for the maximization of the wound healing response with enhanced tissue integration and repair, as opposed to a typical foreign body response characterized by fibrous encapsulation and implant isolation. We discuss current challenges in the clinical translation of immunomodulatory biomaterials, such as limitations in the use of in vitro studies and animal models to model the human immune response. Finally, we describe future directions and opportunities for understanding and controlling the biomaterial-immune system interface, including the application of new imaging tools, new animal models, the discovery of new cellular targets, and novel techniques for in situ immune cell reprogramming.
Collapse
Affiliation(s)
- Ricardo Whitaker
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Beatriz Hernaez-Estrada
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States.,NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
| | - Rosa Maria Hernandez
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Kara L Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
154
|
Correa S, Grosskopf AK, Lopez Hernandez H, Chan D, Yu AC, Stapleton LM, Appel EA. Translational Applications of Hydrogels. Chem Rev 2021; 121:11385-11457. [PMID: 33938724 PMCID: PMC8461619 DOI: 10.1021/acs.chemrev.0c01177] [Citation(s) in RCA: 463] [Impact Index Per Article: 115.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Indexed: 12/17/2022]
Abstract
Advances in hydrogel technology have unlocked unique and valuable capabilities that are being applied to a diverse set of translational applications. Hydrogels perform functions relevant to a range of biomedical purposes-they can deliver drugs or cells, regenerate hard and soft tissues, adhere to wet tissues, prevent bleeding, provide contrast during imaging, protect tissues or organs during radiotherapy, and improve the biocompatibility of medical implants. These capabilities make hydrogels useful for many distinct and pressing diseases and medical conditions and even for less conventional areas such as environmental engineering. In this review, we cover the major capabilities of hydrogels, with a focus on the novel benefits of injectable hydrogels, and how they relate to translational applications in medicine and the environment. We pay close attention to how the development of contemporary hydrogels requires extensive interdisciplinary collaboration to accomplish highly specific and complex biological tasks that range from cancer immunotherapy to tissue engineering to vaccination. We complement our discussion of preclinical and clinical development of hydrogels with mechanical design considerations needed for scaling injectable hydrogel technologies for clinical application. We anticipate that readers will gain a more complete picture of the expansive possibilities for hydrogels to make practical and impactful differences across numerous fields and biomedical applications.
Collapse
Affiliation(s)
- Santiago Correa
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Abigail K. Grosskopf
- Chemical
Engineering, Stanford University, Stanford, California 94305, United States
| | - Hector Lopez Hernandez
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Doreen Chan
- Chemistry, Stanford University, Stanford, California 94305, United States
| | - Anthony C. Yu
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
| | | | - Eric A. Appel
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
- Bioengineering, Stanford University, Stanford, California 94305, United States
- Pediatric
Endocrinology, Stanford University School
of Medicine, Stanford, California 94305, United States
- ChEM-H Institute, Stanford
University, Stanford, California 94305, United States
- Woods
Institute for the Environment, Stanford
University, Stanford, California 94305, United States
| |
Collapse
|
155
|
Puiggalí-Jou A, Babeli I, Roa JJ, Zoppe JO, Garcia-Amorós J, Ginebra MP, Alemán C, García-Torres J. Remote Spatiotemporal Control of a Magnetic and Electroconductive Hydrogel Network via Magnetic Fields for Soft Electronic Applications. ACS APPLIED MATERIALS & INTERFACES 2021; 13:42486-42501. [PMID: 34469100 PMCID: PMC8594865 DOI: 10.1021/acsami.1c12458] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Multifunctional hydrogels are a class of materials offering new opportunities for interfacing living organisms with machines due to their mechanical compliance, biocompatibility, and capacity to be triggered by external stimuli. Here, we report a dual magnetic- and electric-stimuli-responsive hydrogel with the capacity to be disassembled and reassembled up to three times through reversible cross-links. This allows its use as an electronic device (e.g., temperature sensor) in the cross-linked state and spatiotemporal control through narrow channels in the disassembled state via the application of magnetic fields, followed by reassembly. The hydrogel consists of an interpenetrated polymer network of alginate (Alg) and poly(3,4-ethylenedioxythiophene) (PEDOT), which imparts mechanical and electrical properties, respectively. In addition, the incorporation of magnetite nanoparticles (Fe3O4 NPs) endows the hydrogel with magnetic properties. After structural, (electro)chemical, and physical characterization, we successfully performed dynamic and continuous transport of the hydrogel through disassembly, transporting the polymer-Fe3O4 NP aggregates toward a target using magnetic fields and its final reassembly to recover the multifunctional hydrogel in the cross-linked state. We also successfully tested the PEDOT/Alg/Fe3O4 NP hydrogel for temperature sensing and magnetic hyperthermia after various disassembly/re-cross-linking cycles. The present methodology can pave the way to a new generation of soft electronic devices with the capacity to be remotely transported.
Collapse
Affiliation(s)
- Anna Puiggalí-Jou
- Departament
d’Enginyeria Química, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany, 10-14, 08019 Barcelona, Spain
| | - Ismael Babeli
- Departament
d’Enginyeria Química, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany, 10-14, 08019 Barcelona, Spain
| | - Joan Josep Roa
- CIEFMA
(Center for Research in Structural Integrity, Reliability and Micromechanics
of Materials)-Department of Materials Science and Engineering, EEBE, Universitat Politècnica de Catalunya-BarcelonaTech, 08019 Barcelona, Spain
- Barcelona
Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, 08930 Barcelona, Spain
| | - Justin O. Zoppe
- Department
of Materials Science and Engineering, Universitat
Politècnica de Catalunya (UPC), 08019 Barcelona, Spain
| | - Jaume Garcia-Amorós
- Grup
de Materials Orgànics, Departament de Química Inorgànica
i Orgànica (Secció de Química Orgànica), Universitat de Barcelona, Martí i Franquès, 1, 08028 Barcelona, Spain
- Institut
de Nanociència i Nanotecnologia (IN2UB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Maria-Pau Ginebra
- Barcelona
Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, 08930 Barcelona, Spain
- Biomaterials,
Biomechanics and Tissue Engineering Group, Department of Materials
Science and Engineering, Universitat Politècnica
de Catalunya (UPC), 08019 Barcelona, Spain
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Carlos Alemán
- Departament
d’Enginyeria Química, EEBE, Universitat Politècnica de Catalunya, C/ Eduard Maristany, 10-14, 08019 Barcelona, Spain
- Barcelona
Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, 08930 Barcelona, Spain
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Jose García-Torres
- Barcelona
Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, 08930 Barcelona, Spain
- Biomaterials,
Biomechanics and Tissue Engineering Group, Department of Materials
Science and Engineering, Universitat Politècnica
de Catalunya (UPC), 08019 Barcelona, Spain
| |
Collapse
|
156
|
Zhang D, Chen Q, Bi Y, Zhang H, Chen M, Wan J, Shi C, Zhang W, Zhang J, Qiao Z, Li J, Chen S, Liu R. Bio-inspired poly-DL-serine materials resist the foreign-body response. Nat Commun 2021; 12:5327. [PMID: 34493717 PMCID: PMC8423817 DOI: 10.1038/s41467-021-25581-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 08/02/2021] [Indexed: 11/09/2022] Open
Abstract
Implantation-caused foreign-body response (FBR) is a commonly encountered issue and can result in failure of implants. The high L-serine content in low immunogenic silk sericin, and the high D-serine content as a neurotransmitter together inspire us to prepare poly-DL-serine (PSer) materials in mitigating the FBR. Here we report highly water soluble, biocompatible and easily accessible PSer hydrogels that cause negligible inflammatory response after subcutaneous implantation in mice for 1 week and 2 weeks. No obvious collagen capsulation is found surrounding the PSer hydrogels after 4 weeks, 3 months and 7 months post implantation. Histological analysis on inflammatory cytokines and RNA-seq assay both indicate that PSer hydrogels show low FBR, comparable to the Mock group. The anti-FBR performance of PSer hydrogels at all time points surpass the poly(ethyleneglycol) hydrogels that is widely utilized as bio-inert materials, implying the potent and wide application of PSer materials in implantable biomaterials and biomedical devices.
Collapse
Affiliation(s)
- Donghui Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Qi Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Yufang Bi
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Haodong Zhang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Minzhang Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Jianglin Wan
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Chao Shi
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Wenjing Zhang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Junyu Zhang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Zhongqian Qiao
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Jin Li
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengfu Chen
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, China
| | - Runhui Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China. .,Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
157
|
Xue K, Wang F, Suwardi A, Han MY, Teo P, Wang P, Wang S, Ye E, Li Z, Loh XJ. Biomaterials by design: Harnessing data for future development. Mater Today Bio 2021; 12:100165. [PMID: 34877520 PMCID: PMC8628044 DOI: 10.1016/j.mtbio.2021.100165] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 01/18/2023] Open
Abstract
Biomaterials is an interdisciplinary field of research to achieve desired biological responses from new materials, regardless of material type. There have been many exciting innovations in this discipline, but commercialization suffers from a lengthy discovery to product pipeline, with many failures along the way. Success can be greatly accelerated by harnessing machine learning techniques to comb through large amounts of data. There are many potential benefits of moving from an unstructured empirical approach to a development strategy that is entrenched in data. Here, we discuss the recent work on the use of machine learning in the discovery and design of biomaterials, including new polymeric, metallic, ceramics, and nanomaterials, and how machine learning can interface with emerging use cases of 3D printing. We discuss the steps for closer integration of machine learning to make this exciting possibility a reality.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Enyi Ye
- Institute of Materials Research and Engineering, A∗STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Zibiao Li
- Institute of Materials Research and Engineering, A∗STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering, A∗STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| |
Collapse
|
158
|
Cai SS, Li T, Akinade T, Zhu Y, Leong KW. Drug delivery carriers with therapeutic functions. Adv Drug Deliv Rev 2021; 176:113884. [PMID: 34302897 PMCID: PMC8440421 DOI: 10.1016/j.addr.2021.113884] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 01/07/2023]
Abstract
Design of micro- or nanocarriers for drug delivery has primarily been focused on properties such as hydrophobicity, biodegradability, size, shape, surface charge, and toxicity, so that they can achieve optimal delivery with respect to drug loading, release kinetics, biodistribution, cellular uptake, and biocompatibility. Incorporation of stimulus-sensitive moieties into the carriers would lead to "smart" delivery systems. A further evolution would be to endow the carrier with a therapeutic function such that it no longer serves as a mere passive entity to release the drug at the target tissue but can be viewed as a therapeutic agent in itself. In this review, we will discuss recent and ongoing efforts over the past decade to design therapeutic drug carriers that confer a biological benefit, including ROS scavenging or generating, pro- or anti-inflammatory, and immuno-evasive properties, to enhance the overall therapeutic efficacy of the delivery systems.
Collapse
Affiliation(s)
- Shuting S. Cai
- Department of Biomedical Engineering, Columbia University, New York 10027, New York, United States
| | - Tianyu Li
- Department of Biomedical Engineering, Columbia University, New York 10027, New York, United States
| | - Tolulope Akinade
- Graduate Program in Cellular, Molecular and Biomedical Studies, Vagelos College of Physicians and Surgeons, Columbia University, New York 10027, New York, United States
| | - Yuefei Zhu
- Department of Biomedical Engineering, Columbia University, New York 10027, New York, United States
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York 10027, New York, United States,Department of Systems Biology, Columbia University, New York 10027, New York, United States,Corresponding author , Mailing address: 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY 10027
| |
Collapse
|
159
|
The evolution of commercial drug delivery technologies. Nat Biomed Eng 2021; 5:951-967. [PMID: 33795852 DOI: 10.1038/s41551-021-00698-w] [Citation(s) in RCA: 658] [Impact Index Per Article: 164.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
Abstract
Drug delivery technologies have enabled the development of many pharmaceutical products that improve patient health by enhancing the delivery of a therapeutic to its target site, minimizing off-target accumulation and facilitating patient compliance. As therapeutic modalities expanded beyond small molecules to include nucleic acids, peptides, proteins and antibodies, drug delivery technologies were adapted to address the challenges that emerged. In this Review Article, we discuss seminal approaches that led to the development of successful therapeutic products involving small molecules and macromolecules, identify three drug delivery paradigms that form the basis of contemporary drug delivery and discuss how they have aided the initial clinical successes of each class of therapeutic. We also outline how the paradigms will contribute to the delivery of live-cell therapies.
Collapse
|
160
|
Elalouf A. Immune response against the biomaterials used in 3D bioprinting of organs. Transpl Immunol 2021; 69:101446. [PMID: 34389430 DOI: 10.1016/j.trim.2021.101446] [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: 06/20/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 12/26/2022]
Abstract
Regenerative medicine has developed promising approaches for healing and replacing defective and damaged organs or tissues with functional ones. Three-dimensional (3D) bioprinting innovation has integrated a potential to design organs or tissues specific to the patient with the capability of rapid construction to fulfill the storage of organs and the need for transplantation. 3D bioprinting of organs has the main goal to develop a structural and functional organ or tissue mimic to the original one. The highly complex fabrication of tissue engineering scaffolds containing biomaterials, tissue models, and biomedical devices has made it possible to print small blood vessels to mimic organs to reduce organ or tissue rejection. 3D bioprinting has the concept of bioinks containing biomaterials that may trigger the immune responses in the body. Nevertheless, foreign body response (FBR) is mediated by various cell types such as B-cells, dendritic cells, macrophages, natural killer cells, neutrophils, and T-cells, and molecular signals such as antibodies (Abs), cytokines, and reactive radical species. Typically, the biomaterial is shielded by the fibrous encapsulation that is regulated by molecular signals. This review explored the progress in 3D bioprinting of vital organs and basic immune response against the biomaterials used in this approach. Thus, evaluating immune response against biomaterials used in 3D printed organs is necessary to mitigate tissue rejection after the transplantation.
Collapse
Affiliation(s)
- Amir Elalouf
- Bar-Ilan University, Department of Management, Ramat Gan 5290002, Israel.
| |
Collapse
|
161
|
Kusamori K. Development of Advanced Cell-Based Therapy by Regulating Cell-Cell Interactions. Biol Pharm Bull 2021; 44:1029-1036. [PMID: 34334488 DOI: 10.1248/bpb.b21-00276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cell-based therapy for disease treatment involves the transplantation of cells obtained either from self or others into relevant patients. While cells constituting the body tissues maintain homeostasis by performing remarkable functions through complicated cell-cell interactions, transplanted cells, which are generally cultured as a monolayer, are unable to recapitulate similar interactions in vivo. The regulation of cell-cell interactions can immensely increase the function and therapeutic effect of transplanted cells. This review aims to summarize the methods of regulating cell-cell interactions that could significantly increase the therapeutic effects of transplanted cells. The first method involves the generation of multicellular spheroids by three-dimensional cell culture. Spheroid formation greatly improved the survival and therapeutic effects of insulin-secreting cells in diabetic mice after transplantation. Moreover, mixed multicellular spheroids, composed of insulin-secreting cells and aorta endothelial cells or fibroblasts, were found to significantly improve insulin secretion. Secondly, adhesamine derivatives, which are low-molecular-weight compounds that accelerate cell adhesion and avoid anoikis and anchorage-dependent apoptosis, have been used to improve the survival of bone marrow-derived cells and significantly enhanced the therapeutic effects in a diabetic mouse model of delayed wound healing. Finally, the avidin-biotin complex method, a cell surface modification method, has been applied to endow tumor-homing mesenchymal stem cells with anti-tumor ability by modifying them with doxorubicin-encapsulated liposomes. The modified cells showed excellent effectiveness in cell-based cancer-targeting therapy. The discussed methods can be useful tools for advanced cell-based therapy, promising future clinical applications.
Collapse
Affiliation(s)
- Kosuke Kusamori
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| |
Collapse
|
162
|
Goswami D, Domingo‐Lopez DA, Ward NA, Millman JR, Duffy GP, Dolan EB, Roche ET. Design Considerations for Macroencapsulation Devices for Stem Cell Derived Islets for the Treatment of Type 1 Diabetes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100820. [PMID: 34155834 PMCID: PMC8373111 DOI: 10.1002/advs.202100820] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/24/2021] [Indexed: 05/08/2023]
Abstract
Stem cell derived insulin producing cells or islets have shown promise in reversing Type 1 Diabetes (T1D), yet successful transplantation currently necessitates long-term modulation with immunosuppressant drugs. An alternative approach to avoiding this immune response is to utilize an islet macroencapsulation device, where islets are incorporated into a selectively permeable membrane that can protect the transplanted cells from acute host response, whilst enabling delivery of insulin. These macroencapsulation systems have to meet a number of stringent and challenging design criteria in order to achieve the ultimate goal of reversing T1D. In this progress report, the design considerations and functional requirements of macroencapsulation systems are reviewed, specifically for stem-cell derived islets (SC-islets), highlighting distinct design parameters. Additionally, a perspective on the future for macroencapsulation systems is given, and how incorporating continuous sensing and closed-loop feedback can be transformative in advancing toward an autonomous biohybrid artificial pancreas.
Collapse
Affiliation(s)
- Debkalpa Goswami
- Institute for Medical Engineering and ScienceMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Daniel A. Domingo‐Lopez
- Department of AnatomyCollege of Medicine, Nursing, and Health SciencesNational University of Ireland GalwayGalwayH91 TK33Ireland
| | - Niamh A. Ward
- Department of Biomedical EngineeringSchool of EngineeringCollege of Science and EngineeringNational University of Ireland GalwayGalwayH91 TK33Ireland
| | - Jeffrey R. Millman
- Division of Endocrinology, Metabolism & Lipid ResearchWashington University School of MedicineSt. LouisMO63110USA
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMO63110USA
| | - Garry P. Duffy
- Department of AnatomyCollege of Medicine, Nursing, and Health SciencesNational University of Ireland GalwayGalwayH91 TK33Ireland
- Advanced Materials and BioEngineering Research Centre (AMBER)Trinity College DublinDublinD02 PN40Ireland
- CÚRAM, Centre for Research in Medical DevicesNational University of Ireland GalwayGalwayH91 TK33Ireland
| | - Eimear B. Dolan
- Department of Biomedical EngineeringSchool of EngineeringCollege of Science and EngineeringNational University of Ireland GalwayGalwayH91 TK33Ireland
| | - Ellen T. Roche
- Institute for Medical Engineering and ScienceMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
163
|
Lumelsky N. Creating a Pro-Regenerative Tissue Microenvironment: Local Control is the Key. Front Bioeng Biotechnol 2021; 9:712685. [PMID: 34368106 PMCID: PMC8334550 DOI: 10.3389/fbioe.2021.712685] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/09/2021] [Indexed: 01/01/2023] Open
Affiliation(s)
- Nadya Lumelsky
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
164
|
Gessner I. Optimizing nanoparticle design and surface modification toward clinical translation. MRS BULLETIN 2021; 46:643-649. [PMID: 34305307 PMCID: PMC8279028 DOI: 10.1557/s43577-021-00132-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 05/14/2023]
Abstract
The field of nanomedicine is a rapidly evolving field driven by the need for safer and more efficient therapies as well as ultrasensitive and fast diagnostics. Although the advantages of nanoparticles for diagnostic and therapeutic applications are unambiguous, in vivo requirements, including low toxicity, long blood circulation time, proper clearance, sufficient stability, and reproducible synthesis have, in most cases, bedeviled their clinical translation. Nevertheless, researchers have the opportunity to have a decisive influence on the future of nanomedicine by developing new multifunctional molecules and adapting the material design to the requirements. Ultimately, the goal is to find the right level of functionality without adding unnecessary complexity to the system. This article aims to emphasize the potential and current challenges of nanoparticle-based medical agents and highlights how smart and functional material design considerations can help to overcome many of the current limitations and increase the clinical value of nanoparticles.
Collapse
Affiliation(s)
- Isabel Gessner
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
165
|
Kharbikar BN, Chendke GS, Desai TA. Modulating the foreign body response of implants for diabetes treatment. Adv Drug Deliv Rev 2021; 174:87-113. [PMID: 33484736 PMCID: PMC8217111 DOI: 10.1016/j.addr.2021.01.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/30/2020] [Accepted: 01/10/2021] [Indexed: 02/06/2023]
Abstract
Diabetes Mellitus is a group of diseases characterized by high blood glucose levels due to patients' inability to produce sufficient insulin. Current interventions often require implants that can detect and correct high blood glucose levels with minimal patient intervention. However, these implantable technologies have not reached their full potential in vivo due to the foreign body response and subsequent development of fibrosis. Therefore, for long-term function of implants, modulating the initial immune response is crucial in preventing the activation and progression of the immune cascade. This review discusses the different molecular mechanisms and cellular interactions involved in the activation and progression of foreign body response (FBR) and fibrosis, specifically for implants used in diabetes. We also highlight the various strategies and techniques that have been used for immunomodulation and prevention of fibrosis. We investigate how these general strategies have been applied to implants used for the treatment of diabetes, offering insights on how these devices can be further modified to circumvent FBR and fibrosis.
Collapse
Affiliation(s)
- Bhushan N Kharbikar
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gauree S Chendke
- University of California Berkeley - University of California San Francisco Graduate Program in Bioengineering, San Francisco, CA 94143, USA
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94143, USA; University of California Berkeley - University of California San Francisco Graduate Program in Bioengineering, San Francisco, CA 94143, USA; Department of Bioengineering, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
166
|
Satin LS, Soleimanpour SA, Walker EM. New Aspects of Diabetes Research and Therapeutic Development. Pharmacol Rev 2021; 73:1001-1015. [PMID: 34193595 PMCID: PMC8274312 DOI: 10.1124/pharmrev.120.000160] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Both type 1 and type 2 diabetes mellitus are advancing at exponential rates, placing significant burdens on health care networks worldwide. Although traditional pharmacologic therapies such as insulin and oral antidiabetic stalwarts like metformin and the sulfonylureas continue to be used, newer drugs are now on the market targeting novel blood glucose-lowering pathways. Furthermore, exciting new developments in the understanding of beta cell and islet biology are driving the potential for treatments targeting incretin action, islet transplantation with new methods for immunologic protection, and the generation of functional beta cells from stem cells. Here we discuss the mechanistic details underlying past, present, and future diabetes therapies and evaluate their potential to treat and possibly reverse type 1 and 2 diabetes in humans. SIGNIFICANCE STATEMENT: Diabetes mellitus has reached epidemic proportions in the developed and developing world alike. As the last several years have seen many new developments in the field, a new and up to date review of these advances and their careful evaluation will help both clinical and research diabetologists to better understand where the field is currently heading.
Collapse
Affiliation(s)
- Leslie S Satin
- Department of Pharmacology (L.S.S.), Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine (L.S.S., S.A.S., E.M.W.), and Brehm Diabetes Center (L.S.S., S.A.S., E.M.W.), University of Michigan Medical School, Ann Arbor, Michigan; and VA Ann Arbor Healthcare System, Ann Arbor, Michigan (S.A.S.) ; ;
| | - Scott A Soleimanpour
- Department of Pharmacology (L.S.S.), Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine (L.S.S., S.A.S., E.M.W.), and Brehm Diabetes Center (L.S.S., S.A.S., E.M.W.), University of Michigan Medical School, Ann Arbor, Michigan; and VA Ann Arbor Healthcare System, Ann Arbor, Michigan (S.A.S.)
| | - Emily M Walker
- Department of Pharmacology (L.S.S.), Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine (L.S.S., S.A.S., E.M.W.), and Brehm Diabetes Center (L.S.S., S.A.S., E.M.W.), University of Michigan Medical School, Ann Arbor, Michigan; and VA Ann Arbor Healthcare System, Ann Arbor, Michigan (S.A.S.) ; ;
| |
Collapse
|
167
|
Lopez-Mendez TB, Santos-Vizcaino E, Pedraz JL, Orive G, Hernandez RM. Cell microencapsulation technologies for sustained drug delivery: Latest advances in efficacy and biosafety. J Control Release 2021; 335:619-636. [PMID: 34116135 DOI: 10.1016/j.jconrel.2021.06.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 06/04/2021] [Accepted: 06/06/2021] [Indexed: 10/21/2022]
Abstract
The development of cell microencapsulation systems began several decades ago. However, today few systems have been tested in clinical trials. For this reason, in the last years, researchers have directed efforts towards trying to solve some of the key aspects that still limit efficacy and biosafety, the two major criteria that must be satisfied to reach the clinical practice. Regarding the efficacy, which is closely related to biocompatibility, substantial improvements have been made, such as the purification or chemical modification of the alginates that normally form the microspheres. Each of the components that make up the microcapsules has been carefully selected to avoid toxicities that can damage the encapsulated cells or generate an immune response leading to pericapsular fibrosis. As for the biosafety, researchers have developed biological circuits capable of actively responding to the needs of the patients to precisely and accurately release the demanded drug dose. Furthermore, the structure of the devices has been subject of study to adequately protect the encapsulated cells and prevent their spread in the body. The objective of this review is to describe the latest advances made by scientist to improve the efficacy and biosafety of cell microencapsulation systems for sustained drug delivery, also highlighting those points that still need to be optimized.
Collapse
Affiliation(s)
- Tania B Lopez-Mendez
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad, 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad, 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Jose Luis Pedraz
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad, 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad, 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), BTI Biotechnology Institute, Vitoria-Gasteiz, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore.
| | - Rosa Maria Hernandez
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad, 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, C/Monforte de Lemos 3-5, 28029 Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
| |
Collapse
|
168
|
Prox J, Seicol B, Qi H, Argall A, Araya N, Behnke N, Guo L. Toward living neuroprosthetics: developing a biological brain pacemaker as a living neuromodulatory implant for improving parkinsonian symptoms. J Neural Eng 2021; 18. [PMID: 34010821 DOI: 10.1088/1741-2552/ac02dd] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 05/19/2021] [Indexed: 12/21/2022]
Abstract
Objective.Therapeutic intervention for Parkinson's disease (PD) via deep brain stimulation (DBS) represents the current paradigm for managing the advanced stages of the disease in patients when treatment with pharmaceuticals becomes inadequate. Although DBS is the prevailing therapy in these cases, the overall effectiveness and reliability of DBS can be diminished over time due to hardware complications and biocompatibility issues with the electronic implants. To achieve a lifetime solution, we envision that the next generation of neural implants will be entirely 'biological' and 'autologous', both physically and functionally. Thus, in this study, we set forth toward developing a biological brain pacemaker for treating PD. Our focus is to investigate engineering strategies for creating a multicellular biological circuit that integrates innate biological design and function while incorporating principles of neuromodulation to create a biological mechanism for delivering high-frequency stimulation with cellular specificity.Approach.We engineer a 3D multicellular circuit design built entirely from biological and biocompatible components using established tissue engineering protocols to demonstrate the feasibility of creating a living neural implant. Furthermore, using 2D co-culture systems, we investigate the physiologically relevant parameters that would be necessary to further develop a therapeutic benefit of high-frequency stimulation with cellular specificity within our construct design.Main results.Our results demonstrate the feasibility of fabricating a 3D multicellular circuit device in an implantable form. Furthermore, we show we can organize cellular materials to create potential functional connections in normal physiological conditions, thus laying down the foundation of designing a high-frequency pacing system for selective and controlled therapeutic neurostimulation.Significance.The findings from this study may lead to the future development of autologous living neural implants that both circumvent the issues inherent in electronic neural implants and form more biocompatible devices with lifelong robustness to repair and restore motor functions, with the ultimate benefit for patients with PD.
Collapse
Affiliation(s)
- Jordan Prox
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, United States of America
| | - Benjamin Seicol
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States of America
| | - Hao Qi
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States of America
| | - Aaron Argall
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, United States of America
| | - Neway Araya
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States of America
| | - Nicholas Behnke
- Department of Food, Agricultural and Biological Engineering, The Ohio State University, Columbus, OH, United States of America
| | - Liang Guo
- Department of Electrical and Computer Engineering, The Ohio State University, Columbus, OH, United States of America
| |
Collapse
|
169
|
Leon Plata P, Zaroudi M, Lee CY, Foster C, Nitsche LC, Rios PD, Wang Y, Oberholzer J, Liu Y. Heterogeneous toroidal spiral particles for islet encapsulation. Biomater Sci 2021; 9:3954-3967. [PMID: 33620354 DOI: 10.1039/d0bm02082f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Transplantable cell encapsulation systems present a promising approach to deliver a therapeutic solution from hormone-producing cells for the treatment of endocrine diseases like type 1 diabetes. However, the development of a broadly effective and safe transplantation system has been challenging. While some current micro-sized capsules have been optimized for adequate nutrient and metabolic transport, they lack the robustness and retrievability for the clinical safety translation that macro-devices may offer. An existing challenge to be addressed in the current macro-devices is their configuration which may lead to unsatisfactory mass transfer. Here, we design and characterize a millimeter-size particle system of poly-ethylene glycol (PEG) featuring internal toroidal spiral channels, called toroidal spiral particles (TSPs). The characteristic internal structure of the TSPs allows for large encapsulation capacity and large surface area available to all the encapsulated cell mass for effective molecular diffusion. The polymeric matrix renders the particle flexible yet robust for safe transplantation and retrieval. We demonstrate the feasibility of fabricating these particles with various polymer compositions, while optimizing their mechanical properties as well as glucose and insulin permeability. Encapsulation of islets of Langerhans is achieved with high loading capacity (∼160 IEQ per TSP) and excellent cell viability. TSP-encapsulated islets showed similar glucose-stimulated insulin secretion to the naked islets. Preliminary biocompatibility of the TSPs on naïve C57BL/6 mice shows minimal inflammatory response after 4-week transplantation into the intraperitoneal (IP) space. Long-term therapeutic efficacy of encapsulated islets needs to be confirmed in diabetic rodent models in the future, while determining minimal mass required to reverse diabetes. However, we believe from the in vitro favorable results and the TSPs' unique design that TSPs may provide a safe, effective method to transplant and retrieve therapeutic cells for type 1 diabetes treatment and may also be applicable for other cell therapies.
Collapse
Affiliation(s)
- Paola Leon Plata
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Mohammadi MR, Rodriguez SM, Luong JC, Li S, Cao R, Alshetaiwi H, Lau H, Davtyan H, Jones MB, Jafari M, Kessenbrock K, Villalta SA, de Vos P, Zhao W, Lakey JRT. Exosome loaded immunomodulatory biomaterials alleviate local immune response in immunocompetent diabetic mice post islet xenotransplantation. Commun Biol 2021; 4:685. [PMID: 34083739 PMCID: PMC8175379 DOI: 10.1038/s42003-021-02229-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 05/07/2021] [Indexed: 12/16/2022] Open
Abstract
Foreign body response (FBR) to biomaterials compromises the function of implants and leads to medical complications. Here, we report a hybrid alginate microcapsule (AlgXO) that attenuated the immune response after implantation, through releasing exosomes derived from human Umbilical Cord Mesenchymal Stem Cells (XOs). Upon release, XOs suppress the local immune microenvironment, where xenotransplantation of rat islets encapsulated in AlgXO led to >170 days euglycemia in immunocompetent mouse model of Type 1 Diabetes. In vitro analyses revealed that XOs suppressed the proliferation of CD3/CD28 activated splenocytes and CD3+ T cells. Comparing suppressive potency of XOs in purified CD3+ T cells versus splenocytes, we found XOs more profoundly suppressed T cells in the splenocytes co-culture, where a heterogenous cell population is present. XOs also suppressed CD3/CD28 activated human peripheral blood mononuclear cells (PBMCs) and reduced their cytokine secretion including IL-2, IL-6, IL-12p70, IL-22, and TNFα. We further demonstrate that XOs mechanism of action is likely mediated via myeloid cells and XOs suppress both murine and human macrophages partly by interfering with NFκB pathway. We propose that through controlled release of XOs, AlgXO provide a promising new platform that could alleviate the local immune response to implantable biomaterials.
Collapse
Affiliation(s)
- M Rezaa Mohammadi
- Department of Materials Science and Engineering, University of California Irvine, Irvine, CA, USA
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Department of Surgery, University of California Irvine, Irvine, CA, USA
| | | | - Jennifer Cam Luong
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Shiri Li
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Rui Cao
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Hamad Alshetaiwi
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Hien Lau
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA
| | - Hayk Davtyan
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, USA
| | - Mathew Blurton Jones
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Mahtab Jafari
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - S Armando Villalta
- Institute for Immunology, University of California Irvine, Irvine, CA, USA
| | - Paul de Vos
- Department of Pathology and Medical Biology, Section Immunoendocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Weian Zhao
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center; Edwards Life Sciences Center for Advanced Cardiovascular Technology; Department of Biomedical Engineering, Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Jonathan R T Lakey
- Sue and Bill Stem Cell Center, University of California Irvine, Irvine, CA, USA.
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA.
- Department of Surgery, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
171
|
Wang X, Maxwell KG, Wang K, Bowers DT, Flanders JA, Liu W, Wang LH, Liu Q, Liu C, Naji A, Wang Y, Wang B, Chen J, Ernst AU, Melero-Martin JM, Millman JR, Ma M. A nanofibrous encapsulation device for safe delivery of insulin-producing cells to treat type 1 diabetes. Sci Transl Med 2021; 13:eabb4601. [PMID: 34078744 PMCID: PMC8563008 DOI: 10.1126/scitranslmed.abb4601] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 02/09/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022]
Abstract
Transplantation of stem cell-derived β (SC-β) cells represents a promising therapy for type 1 diabetes (T1D). However, the delivery, maintenance, and retrieval of these cells remain a challenge. Here, we report the design of a safe and functional device composed of a highly porous, durable nanofibrous skin and an immunoprotective hydrogel core. The device consists of electrospun medical-grade thermoplastic silicone-polycarbonate-urethane and is soft but tough (~15 megapascal at a rupture strain of >2). Tuning the nanofiber size to less than ~500 nanometers prevented cell penetration while maintaining maximum mass transfer and decreased cellular overgrowth on blank (cell-free) devices to as low as a single-cell layer (~3 micrometers thick) when implanted in the peritoneal cavity of mice. We confirmed device safety, indicated as continuous containment of proliferative cells within the device for 5 months. Encapsulating syngeneic, allogeneic, or xenogeneic rodent islets within the device corrected chemically induced diabetes in mice and cells remained functional for up to 200 days. The function of human SC-β cells was supported by the device, and it reversed diabetes within 1 week of implantation in immunodeficient and immunocompetent mice, for up to 120 and 60 days, respectively. We demonstrated the scalability and retrievability of the device in dogs and observed viable human SC-β cells despite xenogeneic immune responses. The nanofibrous device design may therefore provide a translatable solution to the balance between safety and functionality in developing stem cell-based therapies for T1D.
Collapse
Affiliation(s)
- Xi Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Kristina G Maxwell
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Kai Wang
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel T Bowers
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - James A Flanders
- Department of Clinical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Wanjun Liu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Long-Hai Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Qingsheng Liu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Chengyang Liu
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ali Naji
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yong Wang
- Division of Transplant Surgery, University of Virginia, Charlottesville, VA 22904, USA
| | - Bo Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Jing Chen
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Alexander U Ernst
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA.
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
172
|
Xu Y, Patino Gaillez M, Rothe R, Hauser S, Voigt D, Pietzsch J, Zhang Y. Conductive Hydrogels with Dynamic Reversible Networks for Biomedical Applications. Adv Healthc Mater 2021; 10:e2100012. [PMID: 33930246 PMCID: PMC11468162 DOI: 10.1002/adhm.202100012] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/21/2021] [Indexed: 12/30/2022]
Abstract
Conductive hydrogels (CHs) are emerging as a promising and well-utilized platform for 3D cell culture and tissue engineering to incorporate electron signals as biorelevant physical cues. In conventional covalently crosslinked conductive hydrogels, the network dynamics (e.g., stress relaxation, shear shining, and self-healing) required for complex cellular functions and many biomedical utilities (e.g., injection) cannot be easily realized. In contrast, dynamic conductive hydrogels (DCHs) are fabricated by dynamic and reversible crosslinks. By allowing for the breaking and reforming of the reversible linkages, DCHs can provide dynamic environments for cellular functions while maintaining matrix integrity. These dynamic materials can mimic some properties of native tissues, making them well-suited for several biotechnological and medical applications. An overview of the design, synthesis, and engineering of DCHs is presented in this review, focusing on the different dynamic crosslinking mechanisms of DCHs and their biomedical applications.
Collapse
Affiliation(s)
- Yong Xu
- Technische Universität DresdenB CUBE Center for Molecular BioengineeringDresden01307Germany
| | | | - Rebecca Rothe
- Helmholtz‐Zentrum Dresden‐Rossendorf (HZDR)Institute of Radiopharmaceutical Cancer ResearchDepartment of Radiopharmaceutical and Chemical BiologyDresden01328Germany
- Technische Universität DresdenSchool of ScienceFaculty of Chemistry and Food ChemistryDresden01062Germany
| | - Sandra Hauser
- Helmholtz‐Zentrum Dresden‐Rossendorf (HZDR)Institute of Radiopharmaceutical Cancer ResearchDepartment of Radiopharmaceutical and Chemical BiologyDresden01328Germany
| | - Dagmar Voigt
- Technische Universität Dresden, School of ScienceFaculty of BiologyInstitute of BotanyDresden01062Germany
| | - Jens Pietzsch
- Helmholtz‐Zentrum Dresden‐Rossendorf (HZDR)Institute of Radiopharmaceutical Cancer ResearchDepartment of Radiopharmaceutical and Chemical BiologyDresden01328Germany
- Technische Universität DresdenSchool of ScienceFaculty of Chemistry and Food ChemistryDresden01062Germany
| | - Yixin Zhang
- Technische Universität DresdenB CUBE Center for Molecular BioengineeringDresden01307Germany
- Cluster of Excellence Physics of LifeTechnische Universität DresdenDresden01062Germany
| |
Collapse
|
173
|
Wang Y, Li Z, Hu Q. Emerging self-regulated micro/nano drug delivery devices: A step forward towards intelligent diagnosis and therapy. NANO TODAY 2021; 38:101127. [DOI: 10.1016/j.nantod.2021.101127] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
174
|
Crawford L, Wyatt M, Bryers J, Ratner B. Biocompatibility Evolves: Phenomenology to Toxicology to Regeneration. Adv Healthc Mater 2021; 10:e2002153. [PMID: 33829678 PMCID: PMC8221530 DOI: 10.1002/adhm.202002153] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/26/2021] [Indexed: 12/20/2022]
Abstract
The word "biocompatibility," is inconsistent with the observations of healing for so-called biocompatible biomaterials. The vast majority of the millions of medical implants in humans today, presumably "biocompatible," are walled off by a dense, avascular, crosslinked collagen capsule, hardly suggestive of life or compatibility. In contrast, one is now seeing examples of implant biomaterials that lead to a vascularized reconstruction of localized tissue, a biological reaction different from traditional biocompatible materials that generate a foreign body capsule. Both the encapsulated biomaterials and the reconstructive biomaterials qualify as "biocompatible" by present day measurements of biocompatibility. Yet, this new generation of materials would seem to heal "compatibly" with the living organism, where older biomaterials are isolated from the living organism by the dense capsule. This review/perspective article will explore this biocompatibility etymological conundrum by reviewing the history of the concepts around biocompatibility, today's standard methods for assessing biocompatibility, a contemporary view of the foreign body reaction and finally, a compendium of new biomaterials that heal without the foreign body capsule. A new definition of biocompatibility is offered here to address advances in biomaterials design leading to biomaterials that heal into the body in a facile manner.
Collapse
Affiliation(s)
- Lars Crawford
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Meghan Wyatt
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - James Bryers
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Buddy Ratner
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
175
|
Ghanta RK, Aghlara-Fotovat S, Pugazenthi A, Ryan CT, Singh VP, Mathison M, Jarvis MI, Mukherjee S, Hernandez A, Veiseh O. Immune-modulatory alginate protects mesenchymal stem cells for sustained delivery of reparative factors to ischemic myocardium. Biomater Sci 2021; 8:5061-5070. [PMID: 32797143 DOI: 10.1039/d0bm00855a] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Paracrine factors secreted by mesenchymal stem cells (MSCs) have been previously shown to improve cardiac function following acute myocardial infarction (MI). However, cell therapy activates the innate immune response, leading to the rapid elimination of transplanted cells and only short-term therapeutic delivery. Herein, we describe a new strategy to deliver sustained paracrine-mediated MSC therapy to ischemic myocardium. Using an immune evasive, small molecule modified alginate, we encapsulated rat MSC cells in a core-shell hydrogel capsule and implanted them in the pericardial sac of post-MI rats. Encapsulated cells allowed diffusion of reparative paracrine factors at levels similar to non-encapsulated cells in vitro. Encapsulation enabled sustained cell survival with localization over the heart for 2 weeks. The effect of the experimental group on ventricular function and fibrosis was compared with blank (cell free) capsules and unencapsulated MSCs injected into infarcted myocardium. MSC capsules improved post-MI ventricular function ∼2.5× greater than MSC injection. After 4 weeks, post-MI fibrosis was reduced ∼2/3 with MSC capsules, but unchanged with MSC injection. MSC encapsulation with alginate core-shell capsules sustains cell survival and potentiates efficacy of therapy.
Collapse
Affiliation(s)
- Ravi K Ghanta
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | | | - Aarthi Pugazenthi
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Christopher T Ryan
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Vivek P Singh
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Megumi Mathison
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Maria I Jarvis
- Department of Bioengineering, Rice University, Houston, TX, USA.
| | - Sudip Mukherjee
- Department of Bioengineering, Rice University, Houston, TX, USA.
| | - Andrea Hernandez
- Department of Bioengineering, Rice University, Houston, TX, USA.
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|
176
|
Akbarian M. Insulin therapy; a valuable legacy and its future perspective. Int J Biol Macromol 2021; 181:1224-1230. [PMID: 33989689 DOI: 10.1016/j.ijbiomac.2021.05.052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/21/2021] [Accepted: 05/06/2021] [Indexed: 11/30/2022]
Abstract
Proteins and peptides are widely used in various areas including pharmaceutical, health, food, textile and biofuel industries. At present, pharmaceutical proteins and peptides have attracted the attention of many researchers. These types of drugs are superior to chemical drugs in many ways so that every year the number of drugs with a protein or peptide moiety is increasing. Due to high performance and low side effects, the demand for these drugs has increased year by year. The beginning of the protein and peptide drug industry dates back to 1982 with the introduction of the protein hormone insulin into the field of treatment. From this year onwards, a new number of protein and peptide drugs have entered the field of treatment every year. In this article, we focus on human therapeutic insulin. First, the history of the hormone will be introduced, then-current methods for insulin therapy will be discussed and finally, the treatments by this hormone in the future will be pointed. Reading this article would be very helpful for nano researchers, biochemists, organic chemists, material scientists and other people who are interested in soft and hard matters interfaces.
Collapse
Affiliation(s)
- Mohsen Akbarian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Chemistry, National Cheng Kung University, Tainan 701, Taiwan..
| |
Collapse
|
177
|
Shen P, Chen Y, Luo S, Fan Z, Wang J, Chang J, Deng J. Applications of biomaterials for immunosuppression in tissue repair and regeneration. Acta Biomater 2021; 126:31-44. [PMID: 33722787 DOI: 10.1016/j.actbio.2021.03.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/24/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022]
Abstract
The immune system plays an essential role in tissue repair and regeneration. Regardless of innate or adaptive immune responses, immunosuppressive strategies such as macrophage polarization and regulatory T (Treg) cell induction can be used to modulate the immune system to promote tissue repair and regeneration. Biomaterials can improve the production of anti-inflammatory macrophages and Treg cells by providing physiochemical cues or delivering therapeutics such as cytokines, small molecules, microRNA, growth factors, or stem cells in the damaged tissues. Herein, we present an overview of immunosuppressive modulation by biomaterials in tissue regeneration and highlight the mechanisms of macrophage polarization and Treg cell induction. Overall, we foresee that future biomaterials for regenerative strategies will entail more interactions between biomaterials and the immune cells, and more mechanisms of immunosuppression related to T cell subsets remain to be discovered and applied to develop novel biomaterials for tissue repair and regeneration. STATEMENT OF SIGNIFICANCE: Immunosuppression plays a key role in tissue repair and regeneration, and biomaterials can interact with the immune system through their biological properties and by providing physiochemical cues. Here, we summarize the studies on biomaterials that have been used for immunosuppression to facilitate tissue regeneration. In the first part of this review, we demonstrate the crucial role of macrophage polarization and induction of T regulatory (Treg) cells in immunosuppression. In the second part, distinct approaches used by biomaterials to induce immunosuppression are introduced, which show excellent performance in terms of promoting tissue regeneration.
Collapse
Affiliation(s)
- Peng Shen
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Yanxin Chen
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Shuai Luo
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Zhiyuan Fan
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA 19104, USA
| | - Jilong Wang
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Jiang Chang
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China; State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China.
| | - Junjie Deng
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China.
| |
Collapse
|
178
|
Yang L, Pijuan-Galito S, Rho HS, Vasilevich AS, Eren AD, Ge L, Habibović P, Alexander MR, de Boer J, Carlier A, van Rijn P, Zhou Q. High-Throughput Methods in the Discovery and Study of Biomaterials and Materiobiology. Chem Rev 2021; 121:4561-4677. [PMID: 33705116 PMCID: PMC8154331 DOI: 10.1021/acs.chemrev.0c00752] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 02/07/2023]
Abstract
The complex interaction of cells with biomaterials (i.e., materiobiology) plays an increasingly pivotal role in the development of novel implants, biomedical devices, and tissue engineering scaffolds to treat diseases, aid in the restoration of bodily functions, construct healthy tissues, or regenerate diseased ones. However, the conventional approaches are incapable of screening the huge amount of potential material parameter combinations to identify the optimal cell responses and involve a combination of serendipity and many series of trial-and-error experiments. For advanced tissue engineering and regenerative medicine, highly efficient and complex bioanalysis platforms are expected to explore the complex interaction of cells with biomaterials using combinatorial approaches that offer desired complex microenvironments during healing, development, and homeostasis. In this review, we first introduce materiobiology and its high-throughput screening (HTS). Then we present an in-depth of the recent progress of 2D/3D HTS platforms (i.e., gradient and microarray) in the principle, preparation, screening for materiobiology, and combination with other advanced technologies. The Compendium for Biomaterial Transcriptomics and high content imaging, computational simulations, and their translation toward commercial and clinical uses are highlighted. In the final section, current challenges and future perspectives are discussed. High-throughput experimentation within the field of materiobiology enables the elucidation of the relationships between biomaterial properties and biological behavior and thereby serves as a potential tool for accelerating the development of high-performance biomaterials.
Collapse
Affiliation(s)
- Liangliang Yang
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Sara Pijuan-Galito
- School
of Pharmacy, Biodiscovery Institute, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Hoon Suk Rho
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Aliaksei S. Vasilevich
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aysegul Dede Eren
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Lu Ge
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Pamela Habibović
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Morgan R. Alexander
- School
of Pharmacy, Boots Science Building, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Jan de Boer
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aurélie Carlier
- Department
of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Patrick van Rijn
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Qihui Zhou
- Institute
for Translational Medicine, Department of Stomatology, The Affiliated
Hospital of Qingdao University, Qingdao
University, Qingdao 266003, China
| |
Collapse
|
179
|
Promoting musculoskeletal system soft tissue regeneration by biomaterial-mediated modulation of macrophage polarization. Bioact Mater 2021; 6:4096-4109. [PMID: 33997496 PMCID: PMC8091177 DOI: 10.1016/j.bioactmat.2021.04.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/27/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023] Open
Abstract
Musculoskeletal disorders are common in clinical practice. Repairing critical-sized defects in musculoskeletal systems remains a challenge for researchers and surgeons, requiring the application of tissue engineering biomaterials. Successful application depends on the response of the host tissue to the biomaterial and specific healing process of each anatomical structure. The commonly-held view is that biomaterials should be biocompatible to minimize local host immune response. However, a growing number of studies have shown that active modulation of the immune cells, particularly macrophages, via biomaterials is an effective way to control immune response and promote tissue regeneration as well as biomaterial integration. Therefore, we critically review the role of macrophages in the repair of injured musculoskeletal system soft tissues, which have relatively poor regenerative capacities, as well as discuss further enhancement of target tissue regeneration via modulation of macrophage polarization by biomaterial-mediated immunomodulation (biomaterial properties and delivery systems). This active regulation approach rather than passive-evade strategy maximizes the potential of biomaterials to promote musculoskeletal system soft tissue regeneration and provides alternative therapeutic options for repairing critical-sized defects. Different phenotypes of macrophages play a crucial role in musculoskeletal system soft tissue regeneration. Biomaterials and biomaterial-based delivery systems can be utilized to modulate macrophage polarization. This review summarizes immunomodulatory biomaterials to spur musculoskeletal system soft tissue regeneration.
Collapse
|
180
|
Dargaville TR, Harkin DG, Park JR, Cavalcanti A, Bolle ECL, Savi FM, Farrugia BL, Monnery BD, Bernhard Y, Van Guyse JFR, Podevyn A, Hoogenboom R. Poly(2-allylamidopropyl-2-oxazoline)-Based Hydrogels: From Accelerated Gelation Kinetics to In Vivo Compatibility in a Murine Subdermal Implant Model. Biomacromolecules 2021; 22:1590-1599. [PMID: 33764748 DOI: 10.1021/acs.biomac.1c00046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A rapid photo-curing system based on poly(2-ethyl-2-oxazoline-co-2-allylamidopropyl-2-oxazoline) and its in vivo compatibility are presented. The base polymer was synthesized from the copolymerization of 2-ethyl-2-oxazoline (EtOx) and the methyl ester containing 2-methoxycarboxypropyl-2-oxazoline (C3MestOx) followed by amidation with allylamine to yield a highly water-soluble macromer. We showed that spherical hydrogels can be obtained by a simple water-in-oil gelation method using thiol-ene coupling and investigated the in vivo biocompatibility of these hydrogel spheres in a 28-day murine subdermal model. For comparison, hydrogel spheres prepared from poly(ethylene glycol) were also implanted. Both materials displayed mild, yet typical foreign body responses with little signs of fibrosis. This is the first report on the foreign body response of a poly(2-oxazoline) hydrogel, which paves the way for future investigations into how this highly tailorable class of materials can be used for implantable hydrogel devices.
Collapse
Affiliation(s)
- Tim R Dargaville
- Centre for Materials Science, Queensland University of Technology, 2 George Street, Brisbane, Queensland 4000, Australia.,School of Chemistry and Physics, Queensland University of Technology, 2 George Street, Brisbane, Queensland 4000, Australia.,Institute of Health and Biomedical Innovation, Science and Engineering Faculty, Queensland University of Technology, Brisbane, Queensland 4001, Australia
| | - Damien G Harkin
- Institute of Health and Biomedical Innovation, Science and Engineering Faculty, Queensland University of Technology, Brisbane, Queensland 4001, Australia.,School of Biomedical Sciences, Queensland University of Technology, 2 George Street, Brisbane, Queensland, 4000, Australia
| | - Jong-Ryul Park
- Centre for Materials Science, Queensland University of Technology, 2 George Street, Brisbane, Queensland 4000, Australia.,School of Chemistry and Physics, Queensland University of Technology, 2 George Street, Brisbane, Queensland 4000, Australia.,Institute of Health and Biomedical Innovation, Science and Engineering Faculty, Queensland University of Technology, Brisbane, Queensland 4001, Australia
| | - Amanda Cavalcanti
- Centre for Materials Science, Queensland University of Technology, 2 George Street, Brisbane, Queensland 4000, Australia.,School of Chemistry and Physics, Queensland University of Technology, 2 George Street, Brisbane, Queensland 4000, Australia.,Institute of Health and Biomedical Innovation, Science and Engineering Faculty, Queensland University of Technology, Brisbane, Queensland 4001, Australia
| | - Eleonore C L Bolle
- Centre for Materials Science, Queensland University of Technology, 2 George Street, Brisbane, Queensland 4000, Australia.,School of Chemistry and Physics, Queensland University of Technology, 2 George Street, Brisbane, Queensland 4000, Australia.,Institute of Health and Biomedical Innovation, Science and Engineering Faculty, Queensland University of Technology, Brisbane, Queensland 4001, Australia
| | - Flavia Medeiros Savi
- Institute of Health and Biomedical Innovation, Science and Engineering Faculty, Queensland University of Technology, Brisbane, Queensland 4001, Australia
| | - Brooke L Farrugia
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Bryn D Monnery
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, B-9000 Ghent, Belgium
| | - Yann Bernhard
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, B-9000 Ghent, Belgium
| | - Joachim F R Van Guyse
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, B-9000 Ghent, Belgium
| | - Annelore Podevyn
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, B-9000 Ghent, Belgium
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, B-9000 Ghent, Belgium
| |
Collapse
|
181
|
Cell membrane-biomimetic coating via click-mediated liposome fusion for mitigating the foreign-body reaction. Biomaterials 2021; 271:120768. [PMID: 33812321 DOI: 10.1016/j.biomaterials.2021.120768] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/02/2021] [Accepted: 03/15/2021] [Indexed: 12/19/2022]
Abstract
The foreign-body reaction (FBR) caused by the implantation of synthetic polymer scaffolds seriously affects tissue-biomaterial integration and tissue repair. To address this issue, we developed a cell membrane-biomimetic coating formed by "click"-mediated liposome immobilization and fusion on the surface of electrospun fibers to mitigate the FBR. Utilization of electrospun polystyrene microfibrous scaffold as a model matrix, we deposited azide-incorporated silk fibroin on the surface of the fibers by the layer-by-layer assembly, finally, covalently modified with clickable liposomes via copper-free SPAAC click reaction. Compared with physical adsorption, liposomes click covalently binding can quickly fuse to form lipid film and maintain fluidity, which also improved liposome stability in vitro and in vivo. Molecular dynamics simulation proved that "click" improves the binding rate and strength of liposome to silk substrate. Importantly, histological observation and in vivo fluorescent probes imaging showed that liposome-functionalized electrospun fibers had negligible characteristics of the FBR and were accompanied by many infiltrated host cells and new blood vessels. We believe that the promotion of macrophage polarization toward a pro-regenerative phenotype plays an important role in vascularization. This bioinspired strategy paves the way for utilizing cell membrane biomimetic coating to resist the FBR and promote tissue-scaffold integration.
Collapse
|
182
|
Kuwabara R, Hu S, Smink AM, Orive G, Lakey JRT, de Vos P. Applying Immunomodulation to Promote Longevity of Immunoisolated Pancreatic Islet Grafts. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:129-140. [PMID: 33397201 DOI: 10.1089/ten.teb.2020.0326] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Islet transplantation is a promising therapy for insulin-dependent diabetes, but large-scale application is hampered by the lack of a consistent source of insulin-producing cells and need for lifelong administration of immunosuppressive drugs, which are associated with severe side effects. To avoid chronic immunosuppression, islet grafts can be enveloped in immunoisolating polymeric membranes. These immunoisolating polymeric membranes protect islet grafts from cell-mediated rejection while allowing diffusion of oxygen, nutrients, and insulin. Although clinical trials have shown the safety and feasibility of encapsulated islets to control glucose homeostasis, the strategy does up till now not support long-term graft survival. This partly can be explained by a significant loss of insulin-producing cells in the immediate period after implantation. The loss can be prevented by combining immunoisolation with immunomodulation, such as combined administration of immunomodulating cytokines or coencapsulation of immunomodulating cell types such as regulatory T cells, mesenchymal stem cells, or Sertoli cells. Also, administration of specific antibodies or apoptotic donor leucocytes is considered to create a tolerant microenvironment around immunoisolated grafts. In this review, we describe the outcomes and limitations of these approaches, as well as the recent progress in immunoisolating devices. Impact statement Immunoisolation by enveloping islets in semipermeable membranes allows for successful transplantation of islet grafts in the absence of chronic immunosuppression, but the duration of graft survival is still not permanent. The reasons for long-term final graft failure is not fully understood, but combining immunoisolation with immunomodulation of tissues or host immune system has been proposed to enhance the longevity of grafts. This article reviews the recent progress and challenges of immunoisolation, as well as the benefits and feasibility of combining encapsulation approaches with immunomodulation to promote longevity of encapsulated grafts.
Collapse
Affiliation(s)
- Rei Kuwabara
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Biomaterials, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shuxian Hu
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alexandra M Smink
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Jonathan R T Lakey
- Department of Surgery and Biomedical Engineering, University of California Irvine, Irvine, California, USA
| | - Paul de Vos
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
183
|
Gelain F, Luo Z, Rioult M, Zhang S. Self-assembling peptide scaffolds in the clinic. NPJ Regen Med 2021; 6:9. [PMID: 33597509 PMCID: PMC7889856 DOI: 10.1038/s41536-020-00116-w] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022] Open
Abstract
Well-defined scaffold hydrogels made of self-assembling peptides have found their way into clinical products. By examining the properties and applications of two self-assembling peptides-EAK16 and RADA16-we highlight the potential for translating designer biological scaffolds into commercial products.
Collapse
Affiliation(s)
- Fabrizio Gelain
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBREMIT), IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, Italy.
- Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, 20162, Milan, Italy.
| | - Zhongli Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Centre, Chongqing Medical University, Chongqing, 400016, China.
| | | | - Shuguang Zhang
- Laboratory of Molecular Architecture, Media Lab, Massachusetts Institute of Technology, Cambridge, MA, 02139-4307, USA.
| |
Collapse
|
184
|
Li T, Chen X, Qian Y, Shao J, Li X, Liu S, Zhu L, Zhao Y, Ye H, Yang Y. A synthetic BRET-based optogenetic device for pulsatile transgene expression enabling glucose homeostasis in mice. Nat Commun 2021; 12:615. [PMID: 33504786 PMCID: PMC7840992 DOI: 10.1038/s41467-021-20913-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 12/21/2020] [Indexed: 12/26/2022] Open
Abstract
Pulsing cellular dynamics in genetic circuits have been shown to provide critical capabilities to cells in stress response, signaling and development. Despite the fascinating discoveries made in the past few years, the mechanisms and functional capabilities of most pulsing systems remain unclear, and one of the critical challenges is the lack of a technology that allows pulsatile regulation of transgene expression both in vitro and in vivo. Here, we describe the development of a synthetic BRET-based transgene expression (LuminON) system based on a luminescent transcription factor, termed luminGAVPO, by fusing NanoLuc luciferase to the light-switchable transcription factor GAVPO. luminGAVPO allows pulsatile and quantitative activation of transgene expression via both chemogenetic and optogenetic approaches in mammalian cells and mice. Both the pulse amplitude and duration of transgene expression are highly tunable via adjustment of the amount of furimazine. We further demonstrated LuminON-mediated blood-glucose homeostasis in type 1 diabetic mice. We believe that the BRET-based LuminON system with the pulsatile dynamics of transgene expression provides a highly sensitive tool for precise manipulation in biological systems that has strong potential for application in diverse basic biological studies and gene- and cell-based precision therapies in the future.
Collapse
Affiliation(s)
- Ting Li
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
- School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
| | - Xianjun Chen
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
- School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yajie Qian
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
- School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
| | - Jiawei Shao
- Synthetic Biology and Biomedical Engineering Laboratory, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Dongchuan Road 500, Shanghai, 200241, China
| | - Xie Li
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
- School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
| | - Shuning Liu
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
- School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
| | - Linyong Zhu
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
| | - Yuzheng Zhao
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
- School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China
| | - Haifeng Ye
- Synthetic Biology and Biomedical Engineering Laboratory, Biomedical Synthetic Biology Research Center, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Dongchuan Road 500, Shanghai, 200241, China.
| | - Yi Yang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China.
- School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai, 200237, China.
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
185
|
Karpov AA, Anikin NA, Mihailova AM, Smirnov SS, Vaulina DD, Shilenko LA, Ivkin DY, Bagrov AY, Moiseeva OM, Galagudza MM. Model of Chronic Thromboembolic Pulmonary Hypertension in Rats Caused by Repeated Intravenous Administration of Partially Biodegradable Sodium Alginate Microspheres. Int J Mol Sci 2021; 22:ijms22031149. [PMID: 33498971 PMCID: PMC7865986 DOI: 10.3390/ijms22031149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/23/2022] Open
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a rare and life-threatening complication of pulmonary embolism. As existing animal models of CTEPH do not fully recapitulate complex disease pathophysiology, we report a new rat model for CTEPH evoked by repetitive embolization of the distal pulmonary artery branches with partially biodegradable alginate microspheres (MSs). MSs (180 ± 28 μm) were intravenously administered eight times at 4-day intervals; control animals received saline. The validity of the model was confirmed using transthoracic echocardiography, exercise testing, catheterization of the right ventricle, and histological examination of the lung and heart. The animals in the CTEPH group demonstrated a stable increase in right ventricular systolic pressure (RVSP) and decreased exercise tolerance. Histopathological examination revealed advanced medial hypertrophy in the small pulmonary arteries associated with fibrosis. The diameter of the main pulmonary artery was significantly larger in the CTEPH group than in the control group. Marinobufagenin and endothelin-1 serum levels were significantly elevated in rats with CTEPH. In conclusion, repetitive administration of alginate MSs in rats resulted in CTEPH development characterized by specific lung vasculature remodeling, reduced exercise tolerance, and a persistent rise in RVSP. The developed model can be used for pre-clinical testing of promising drug candidates.
Collapse
Affiliation(s)
- Andrei A. Karpov
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (N.A.A.); (A.M.M.); (S.S.S.); (D.D.V.); (L.A.S.); (O.M.M.)
- Correspondence: (A.A.K.); (M.M.G.); Tel.: +7-951-678-7006 (A.A.K.)
| | - Nikita A. Anikin
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (N.A.A.); (A.M.M.); (S.S.S.); (D.D.V.); (L.A.S.); (O.M.M.)
| | - Aleksandra M. Mihailova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (N.A.A.); (A.M.M.); (S.S.S.); (D.D.V.); (L.A.S.); (O.M.M.)
| | - Sergey S. Smirnov
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (N.A.A.); (A.M.M.); (S.S.S.); (D.D.V.); (L.A.S.); (O.M.M.)
- First Pavlov State Medical University of Saint Petersburg, 197022 St. Petersburg, Russia
| | - Dariya D. Vaulina
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (N.A.A.); (A.M.M.); (S.S.S.); (D.D.V.); (L.A.S.); (O.M.M.)
- N.P. Bechtereva Institute of Human Brain, Russian Academy of Science, 197376 St. Petersburg, Russia
| | - Leonid A. Shilenko
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (N.A.A.); (A.M.M.); (S.S.S.); (D.D.V.); (L.A.S.); (O.M.M.)
- First Pavlov State Medical University of Saint Petersburg, 197022 St. Petersburg, Russia
| | - Dmitry Yu. Ivkin
- Saint Petersburg State Chemical Pharmaceutical University, 197376 St. Petersburg, Russia;
| | - Alexei Y. Bagrov
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 194223 St. Petersburg, Russia;
| | - Olga M. Moiseeva
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (N.A.A.); (A.M.M.); (S.S.S.); (D.D.V.); (L.A.S.); (O.M.M.)
| | - Michael M. Galagudza
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (N.A.A.); (A.M.M.); (S.S.S.); (D.D.V.); (L.A.S.); (O.M.M.)
- Correspondence: (A.A.K.); (M.M.G.); Tel.: +7-951-678-7006 (A.A.K.)
| |
Collapse
|
186
|
Molakandov K, Berti DA, Beck A, Elhanani O, Walker MD, Soen Y, Yavriyants K, Zimerman M, Volman E, Toledo I, Erukhimovich A, Levy AM, Hasson A, Itskovitz-Eldor J, Chebath J, Revel M. Selection for CD26 - and CD49A + Cells From Pluripotent Stem Cells-Derived Islet-Like Clusters Improves Therapeutic Activity in Diabetic Mice. Front Endocrinol (Lausanne) 2021; 12:635405. [PMID: 34025576 PMCID: PMC8131825 DOI: 10.3389/fendo.2021.635405] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/07/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cell therapy of diabetes aims at restoring the physiological control of blood glucose by transplantation of functional pancreatic islet cells. A potentially unlimited source of cells for such transplantations would be islet cells derived from an in vitro differentiation of human pluripotent stem cells (hESC/hiPSC). The islet-like clusters (ILC) produced by the known differentiation protocols contain various cell populations. Among these, the β-cells that express both insulin and the transcription factor Nkx6.1 seem to be the most efficient to restore normoglycemia in diabetes animal models. Our aim was to find markers allowing selection of these efficient cells. METHODS Functional Cell-Capture Screening (FCCS) was used to identify markers that preferentially capture the cells expressing both insulin and Nkx6.1, from hESC-derived ILC cells. In order to test whether selection for such markers could improve cell therapy in diabetic mouse models, we used ILC produced from a clinical-grade line of hESC by a refined differentiation protocol adapted to up-scalable bioreactors. Re-aggregated MACS sorted cells were encapsulated in microspheres made of alginate modified to reduce foreign body reaction. Implantation was done intraperitoneally in STZ-treated C57BL/6 immuno-competent mice. RESULTS CD49A (integrin alpha1) was identified by FCCS as a marker for cells that express insulin (or C-peptide) as well as Nkx6.1 in ILC derived by hESC differentiation. The ILC fraction enriched in CD49A + cells rapidly reduced glycemia when implanted in diabetic mice, whereas mice receiving the CD49A depleted population remained highly diabetic. CD49A-enriched ILC cells also produced higher levels of human C-peptide in the blood of transplanted mice. However, the difference between CD49A-enriched and total ILC cells remained small. Another marker, CD26 (DPP4), was identified by FCCS as binding insulin-expressing cells which are Nkx6.1 negative. Depletion of CD26 + cells followed by enrichment for CD49A + cells increased insulin+/Nkx6.1+ cells fraction to ~70%. The CD26 - /CD49A + enriched ILC exhibited improved function over non-sorted ILC or CD49A + cells in diabetic mice and maintain prolonged blood C-peptide levels. CONCLUSIONS Refining the composition of ILC differentiated from hPSC by negative selection to remove cells expressing CD26 and positive selection for CD49A expressing cells could enable more effective cell therapy of diabetes.
Collapse
Affiliation(s)
- Kfir Molakandov
- Kadimastem Ltd., Weizmann Science Park, Ness Ziona, Israel
- *Correspondence: Kfir Molakandov,
| | | | - Avital Beck
- Kadimastem Ltd., Weizmann Science Park, Ness Ziona, Israel
| | - Ofer Elhanani
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Michael D. Walker
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Yoav Soen
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | | | - Ella Volman
- Kadimastem Ltd., Weizmann Science Park, Ness Ziona, Israel
| | - Itzik Toledo
- Kadimastem Ltd., Weizmann Science Park, Ness Ziona, Israel
| | | | - Alon M. Levy
- Kadimastem Ltd., Weizmann Science Park, Ness Ziona, Israel
| | - Arik Hasson
- Kadimastem Ltd., Weizmann Science Park, Ness Ziona, Israel
| | | | - Judith Chebath
- Kadimastem Ltd., Weizmann Science Park, Ness Ziona, Israel
| | - Michel Revel
- Kadimastem Ltd., Weizmann Science Park, Ness Ziona, Israel
- Department of Molecular Genetics (emeritus), Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
187
|
Dong D, Tsao C, Hung HC, Yao F, Tang C, Niu L, Ma J, MacArthur J, Sinclair A, Wu K, Jain P, Hansen MR, Ly D, Tang SGH, Luu TM, Jain P, Jiang S. High-strength and fibrous capsule-resistant zwitterionic elastomers. SCIENCE ADVANCES 2021; 7:7/1/eabc5442. [PMID: 33523839 PMCID: PMC7775767 DOI: 10.1126/sciadv.abc5442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/06/2020] [Indexed: 05/20/2023]
Abstract
The high mechanical strength and long-term resistance to the fibrous capsule formation are two major challenges for implantable materials. Unfortunately, these two distinct properties do not come together and instead compromise each other. Here, we report a unique class of materials by integrating two weak zwitterionic hydrogels into an elastomer-like high-strength pure zwitterionic hydrogel via a "swelling" and "locking" mechanism. These zwitterionic-elastomeric-networked (ZEN) hydrogels are further shown to efficaciously resist the fibrous capsule formation upon implantation in mice for up to 1 year. Such materials with both high mechanical properties and long-term fibrous capsule resistance have never been achieved before. This work not only demonstrates a class of durable and fibrous capsule-resistant materials but also provides design principles for zwitterionic elastomeric hydrogels.
Collapse
Affiliation(s)
- Dianyu Dong
- Department of Chemical Engineering, University of Washington, Seattle, WA 98185, USA
- School of Chemical Engineering and Technology, and Key Laboratory of Systems Bioengineering of Ministry of Education, Tianjin University, Tianjin 300350, China
| | - Caroline Tsao
- Department of Chemical Engineering, University of Washington, Seattle, WA 98185, USA
| | - Hsiang-Chieh Hung
- Department of Chemical Engineering, University of Washington, Seattle, WA 98185, USA
| | - Fanglian Yao
- School of Chemical Engineering and Technology, and Key Laboratory of Systems Bioengineering of Ministry of Education, Tianjin University, Tianjin 300350, China
| | - Chenjue Tang
- Department of Chemical Engineering, University of Washington, Seattle, WA 98185, USA
| | - Liqian Niu
- Department of Chemical Engineering, University of Washington, Seattle, WA 98185, USA
| | - Jinrong Ma
- Department of Chemical Engineering, University of Washington, Seattle, WA 98185, USA
| | - Joel MacArthur
- Department of Chemical Engineering, University of Washington, Seattle, WA 98185, USA
| | - Andrew Sinclair
- Department of Chemical Engineering, University of Washington, Seattle, WA 98185, USA
| | - Kan Wu
- Department of Chemical Engineering, University of Washington, Seattle, WA 98185, USA
| | - Priyesh Jain
- Department of Chemical Engineering, University of Washington, Seattle, WA 98185, USA
| | - Mitchell Ryan Hansen
- Department of Chemical Engineering, University of Washington, Seattle, WA 98185, USA
| | - Dorathy Ly
- Department of Chemical Engineering, University of Washington, Seattle, WA 98185, USA
| | | | - Tammy My Luu
- Department of Chemical Engineering, University of Washington, Seattle, WA 98185, USA
| | - Parul Jain
- Department of Chemical Engineering, University of Washington, Seattle, WA 98185, USA
| | - Shaoyi Jiang
- Department of Chemical Engineering, University of Washington, Seattle, WA 98185, USA.
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
188
|
Fuchs S, Ernst AU, Wang LH, Shariati K, Wang X, Liu Q, Ma M. Hydrogels in Emerging Technologies for Type 1 Diabetes. Chem Rev 2020; 121:11458-11526. [DOI: 10.1021/acs.chemrev.0c01062] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Stephanie Fuchs
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Alexander U. Ernst
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Long-Hai Wang
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Kaavian Shariati
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Xi Wang
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Qingsheng Liu
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Minglin Ma
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
189
|
Welch NG, Winkler DA, Thissen H. Antifibrotic strategies for medical devices. Adv Drug Deliv Rev 2020; 167:109-120. [PMID: 32553685 DOI: 10.1016/j.addr.2020.06.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/02/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022]
Abstract
A broad range of medical devices initiate an immune reaction known as the foreign body response (FBR) upon implantation. Here, collagen deposition at the surface of the implant occurs as a result of the FBR, ultimately leading to fibrous encapsulation and, in many cases, reduced function or failure of the device. Despite significant efforts, the prevention of fibrotic encapsulation has not been realized at this point in time. However, many next-generation medical technologies including cellular therapies, sensors and devices depend on the ability to modulate and control the FBR. For these technologies to become viable, significant advances must be made in understanding the underlying mechanism of this response as well as in the methods modulating this response. In this review, we highlight recent advances in the development of materials and coatings providing a reduced FBR and emphasize key characteristics of high-performing approaches. We also provide a detailed overview of the state-of-the-art in strategies relying on controlled drug release, the surface display of bioactive signals, materials-based approaches, and combinations of these approaches. Finally, we offer perspectives on future directions in this field.
Collapse
|
190
|
Ghoneim MA, Refaie AF, Elbassiouny BL, Gabr MM, Zakaria MM. From Mesenchymal Stromal/Stem Cells to Insulin-Producing Cells: Progress and Challenges. Stem Cell Rev Rep 2020; 16:1156-1172. [PMID: 32880857 PMCID: PMC7667138 DOI: 10.1007/s12015-020-10036-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stromal cells (MSCs) are an attractive option for cell therapy for type 1 diabetes mellitus (DM). These cells can be obtained from many sources, but bone marrow and adipose tissue are the most studied. MSCs have distinct advantages since they are nonteratogenic, nonimmunogenic and have immunomodulatory functions. Insulin-producing cells (IPCs) can be generated from MSCs by gene transfection, gene editing or directed differentiation. For directed differentiation, MSCs are usually cultured in a glucose-rich medium with various growth and activation factors. The resulting IPCs can control chemically-induced diabetes in immune-deficient mice. These findings are comparable to those obtained from pluripotent cells. PD-L1 and PD-L2 expression by MSCs is upregulated under inflammatory conditions. Immunomodulation occurs due to the interaction between these ligands and PD-1 receptors on T lymphocytes. If this function is maintained after differentiation, life-long immunosuppression or encapsulation could be avoided. In the clinical setting, two sites can be used for transplantation of IPCs: the subcutaneous tissue and the omentum. A 2-stage procedure is required for the former and a laparoscopic procedure for the latter. For either site, cells should be transplanted within a scaffold, preferably one from fibrin. Several questions remain unanswered. Will the transplanted cells be affected by the antibodies involved in the pathogenesis of type 1 DM? What is the functional longevity of these cells following their transplantation? These issues have to be addressed before clinical translation is attempted. Graphical Abstract Bone marrow MSCs are isolated from the long bone of SD rats. Then they are expanded and through directed differentiation insulin-producing cells are formed. The differentiated cells are loaded onto a collagen scaffold. If one-stage transplantation is planned, a drug delivery system must be incorporated to ensure immediate oxygenation, promote vascularization and provide some growth factors. Some mechanisms involved in the immunomodulatory function of MSCs. These are implemented either by cell to cell contact or by the release of soluble factors. Collectively, these pathways results in an increase in T-regulatory cells.
Collapse
|
191
|
Gunawardana M, Remedios-Chan M, Sanchez D, Webster S, Galvan P, Fanter R, Castonguay AE, Webster P, Moss JA, Kuo J, Gallay PA, Vincent KL, Motamedi M, Weinberger D, Marzinke MA, Hendrix CW, Baum MM. Multispecies Evaluation of a Long-Acting Tenofovir Alafenamide Subdermal Implant for HIV Prophylaxis. Front Pharmacol 2020; 11:569373. [PMID: 33536904 PMCID: PMC7849190 DOI: 10.3389/fphar.2020.569373] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/19/2020] [Indexed: 12/17/2022] Open
Abstract
New HIV-1 infection rates far outpace the targets set by global health organizations, despite important progress in curbing the progression of the epidemic. Long-acting (LA) formulations delivering antiretroviral (ARV) agents for HIV-1 pre-exposure prophylaxis (PrEP) hold significant promise, potentially facilitating adherence due to reduced dosing frequency compared to oral regimens. We have developed a subdermal implant delivering the potent ARV drug tenofovir alafenamide that could provide protection from HIV-1 infection for 6 months, or longer. Implants from the same lot were investigated in mice and sheep for local safety and pharmacokinetics (PKs). Ours is the first report using these animal models to evaluate subdermal implants for HIV-1 PrEP. The devices appeared safe, and the plasma PKs as well as the drug and metabolite concentrations in dermal tissue adjacent to the implants were studied and contrasted in two models spanning the extremes of the body weight spectrum. Drug and drug metabolite concentrations in dermal tissue are key in assessing local exposure and any toxicity related to the active agent. Based on our analysis, both animal models were shown to hold significant promise in LA product development.
Collapse
Affiliation(s)
- Manjula Gunawardana
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, CA, United States
| | - Mariana Remedios-Chan
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, CA, United States
| | - Debbie Sanchez
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, CA, United States
| | - Simon Webster
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, CA, United States
| | - Patricia Galvan
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, CA, United States
| | - Rob Fanter
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, CA, United States
| | - Amalia E. Castonguay
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, CA, United States
| | - Paul Webster
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, CA, United States
| | - John A. Moss
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, CA, United States
| | - Joseph Kuo
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| | - Philippe A. Gallay
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| | - Kathleen L. Vincent
- Center for Biomedical Engineering, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Massoud Motamedi
- Center for Biomedical Engineering, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | | | - Mark A. Marzinke
- Department of Medicine, Johns Hopkins University, Baltimore, MD, United States
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Craig W. Hendrix
- Department of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Marc M. Baum
- Department of Chemistry, Oak Crest Institute of Science, Monrovia, CA, United States
| |
Collapse
|
192
|
Xie M, Viviani M, Fussenegger M. Engineering precision therapies: lessons and motivations from the clinic. Synth Biol (Oxf) 2020; 6:ysaa024. [PMID: 33817342 PMCID: PMC7998714 DOI: 10.1093/synbio/ysaa024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/18/2022] Open
Abstract
In the past decade, gene- and cell-based therapies have been at the forefront of the biomedical revolution. Synthetic biology, the engineering discipline of building sophisticated 'genetic software' to enable precise regulation of gene activities in living cells, has been a decisive success factor of these new therapies. Here, we discuss the core technologies and treatment strategies that have already gained approval for therapeutic applications in humans. We also review promising preclinical work that could either enhance the efficacy of existing treatment strategies or pave the way for new precision medicines to treat currently intractable human conditions.
Collapse
Affiliation(s)
- Mingqi Xie
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zheijang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zheijang, China
| | - Mirta Viviani
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zheijang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zheijang, China
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Faculty of Science, University of Basel, Basel, Switzerland
| |
Collapse
|
193
|
Lopez-Mendez TB, Santos-Vizcaino E, Pedraz JL, Hernandez RM, Orive G. Cell microencapsulation technologies for sustained drug delivery: Clinical trials and companies. Drug Discov Today 2020; 26:852-861. [PMID: 33242694 DOI: 10.1016/j.drudis.2020.11.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/03/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022]
Abstract
In recent years, cell microencapsulation technology has advanced, mainly driven by recent developments in the use of stem cells or the optimization of biomaterials. Old challenges have been addressed from new perspectives, and systems developed and improved for decades are now being transferred to the market by novel start-ups and consolidated companies. These products are mainly intended for the treatment of diabetes mellitus (DM), but also cancer, central nervous system (CNS) disorders or lysosomal diseases, among others. In this review, we analyze the results obtained in clinical trials to date and define the global key players that will lead the cell microencapsulation market to bring this technology to the clinic in the future.
Collapse
Affiliation(s)
- Tania B Lopez-Mendez
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Jose Luis Pedraz
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Rosa Maria Hernandez
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua); BTI Biotechnology Institute, Vitoria-Gasteiz, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore.
| |
Collapse
|
194
|
Caserto JS, Bowers DT, Shariati K, Ma M. Biomaterial Applications in Islet Encapsulation and Transplantation. ACS APPLIED BIO MATERIALS 2020; 3:8127-8135. [DOI: 10.1021/acsabm.0c01235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Julia S. Caserto
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Daniel T. Bowers
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Kaavian Shariati
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
195
|
O’Dwyer J, O’Cearbhaill RE, Wylie R, O’Mahony S, O’Dwyer M, Duffy GP, Dolan EB. Enhancing delivery of small molecule and cell-based therapies for ovarian cancer using advanced delivery strategies. ADVANCED THERAPEUTICS 2020; 3:2000144. [PMID: 33709016 PMCID: PMC7942751 DOI: 10.1002/adtp.202000144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Indexed: 12/17/2022]
Abstract
Ovarian cancer is the most lethal gynecological malignancy with a global five-year survival rate of 30-50%. First-line treatment involves cytoreductive surgery and administration of platinum-based small molecules and paclitaxel. These therapies were traditionally administered via intravenous infusion, although intraperitoneal delivery has also been investigated. Initial clinical trials of intraperitoneal administration for ovarian cancer indicated significant improvements in overall survival compared to intravenous delivery, but this result is not consistent across all studies performed. Recently cell-based immunotherapy has been of interest for ovarian cancer. Direct intraperitoneal delivery of cell-based immunotherapies might prompt local immunoregulatory mechanisms to act synergistically with the delivered immunotherapy. Based on this theory, pre-clinical in vivo studies have delivered these cell-based immunotherapies via the intraperitoneal route, with promising results. However, successful intraperitoneal delivery of cell-based immunotherapy and clinical adoption of this technique will depend on overcoming challenges of intraperitoneal delivery and finding the optimal combinations of dose, therapeutic and delivery route. We review the potential advantages and disadvantages of intraperitoneal delivery of cell-based immunotherapy for ovarian cancer and the pre-clinical and clinical work performed so far. Potential advanced delivery strategies, which might improve the efficacy and adoption of intraperitoneal delivery of therapy for ovarian cancer, are also outlined.
Collapse
Affiliation(s)
- Joanne O’Dwyer
- Department of Biomedical Engineering, School of Engineering, College of Science and Engineering, National University of Ireland Galway, Ireland; Anatomy & Regenerative Medicine Institute, School of Medicine, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Ireland
| | - Roisin E. O’Cearbhaill
- Anatomy & Regenerative Medicine Institute, School of Medicine, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Ireland; Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Robert Wylie
- Anatomy & Regenerative Medicine Institute, School of Medicine, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Ireland
| | - Saoirse O’Mahony
- Department of Biomedical Engineering, School of Engineering, College of Science and Engineering, National University of Ireland Galway, Ireland
| | - Michael O’Dwyer
- Apoptosis Research Centre, National University of Ireland Galway, Ireland
| | - Garry P. Duffy
- Anatomy & Regenerative Medicine Institute, School of Medicine, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Ireland
| | - Eimear B. Dolan
- Department of Biomedical Engineering, School of Engineering, College of Science and Engineering, National University of Ireland Galway, Ireland
| |
Collapse
|
196
|
Clough DW, King JL, Li F, Shea LD. Integration of Islet/Beta-Cell Transplants with Host Tissue Using Biomaterial Platforms. Endocrinology 2020; 161:bqaa156. [PMID: 32894299 PMCID: PMC8253249 DOI: 10.1210/endocr/bqaa156] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/27/2020] [Indexed: 12/30/2022]
Abstract
Cell-based therapies are emerging for type I diabetes mellitus (T1D), an autoimmune disease characterized by the destruction of insulin-producing pancreatic β-cells, as a means to provide long-term restoration of glycemic control. Biomaterial scaffolds provide an opportunity to enhance the manufacturing and transplantation of islets or stem cell-derived β-cells. In contrast to encapsulation strategies that prevent host contact with the graft, recent approaches aim to integrate the transplant with the host to facilitate glucose sensing and insulin distribution, while also needing to modulate the immune response. Scaffolds can provide a supportive niche for cells either during the manufacturing process or following transplantation at extrahepatic sites. Scaffolds are being functionalized to deliver oxygen, angiogenic, anti-inflammatory, or trophic factors, and may facilitate cotransplantation of cells that can enhance engraftment or modulate immune responses. This local engineering of the transplant environment can complement systemic approaches for maximizing β-cell function or modulating immune responses leading to rejection. This review discusses the various scaffold platforms and design parameters that have been identified for the manufacture of human pluripotent stem cell-derived β-cells, and the transplantation of islets/β-cells to maintain normal blood glucose levels.
Collapse
Affiliation(s)
- Daniel W Clough
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Jessica L King
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Feiran Li
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
197
|
Mohammadi MR, Dehkordi-Vakil F, Ricks-Oddie J, Mansfield R, Kashimiri H, Daniels M, Zhao W, Lakey JR. Preferences of Type 1 Diabetic Patients on Devices for Islet Transplantation. Cell Transplant 2020; 29:963689720952343. [PMID: 33023311 PMCID: PMC7784499 DOI: 10.1177/0963689720952343] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Transplantation of pancreatic islets within a biomaterial device is currently
under investigation in clinical trials for the treatment of patients with type 1
diabetes (T1D). Patients’ preferences on such implants could guide the designs
of next-generation implantable devices; however, such information is not
currently available. We surveyed the preferences of 482 patients with T1D on the
size, shape, visibility, and transplantation site of islet containing implants.
More than 83% of participants were willing to receive autologous stem cells, and
there was no significant association between implant fabricated by one’s own
stem cell with gender (χ2 (1, n = 468) = 0.28; P = 0.6) or
with age (χ2 (4, n = 468) = 2.92; P = 0.6).
Preferred location for islet transplantation within devices was under the skin
(52.7%). 48.3% preferred microscopic disks, and 32.3% preferred a thin device
(like a credit card). Moreover, 58.4% preferred the implant to be as small as
possible, 25.4% did not care about visibility, and 16.2% preferred their
implants not to be visible. Among female participants, 81% cared about the
implant visibility, whereas this number was 64% for male respondents
(χ2 test (1, n = 468) = 16.34; P <
0.0001). 22% of those younger than 50 years of age and 30% of those older than
50 did not care about the visibility of implant (χ2 test (4, n = 468) = 23.69; P <
0.0001). These results suggest that subcutaneous sites and micron-sized devices
are preferred choices among patients with T1D who participated in our
survey.
Collapse
Affiliation(s)
- M Rezaa Mohammadi
- Department of Materials Science and Engineering, 8788University of California, Irvine, CA, USA.,Sue and Bill Gross Stem Cell Research Center, 8788University of California, Irvine, CA, USA
| | - Farideh Dehkordi-Vakil
- Center for Statistical Consulting, Department of Statistics, 8788University of California, Irvine, CA, USA
| | - Joni Ricks-Oddie
- Center for Statistical Consulting, Department of Statistics, 8788University of California, Irvine, CA, USA
| | - Robert Mansfield
- 369679Juvenile Diabetes Research Foundation Orange County Chapter, Irvine, CA, USA
| | | | - Mark Daniels
- CHOC Children's Endocrine & Diabetes Center, Orange, CA, USA
| | - Weian Zhao
- Sue and Bill Gross Stem Cell Research Center, 8788University of California, Irvine, CA, USA.,Department of Pharmaceutical Sciences, Chao Family Comprehensive Cancer Center, Edwards Life Sciences Center for Advanced Cardiovascular Technology, 8788University of California, Irvine, Irvine, CA, USA.,Department of Biomedical Engineering, 8788University of California, Irvine, Irvine, CA, USA.,Department of Biological Chemistry, 8788University of California, Irvine, Irvine, CA, USA
| | - Jonathan Rt Lakey
- Sue and Bill Gross Stem Cell Research Center, 8788University of California, Irvine, CA, USA.,Department of Surgery and Biomedical Engineering, 8788University of California Irvine, Orange, CA, USA
| |
Collapse
|
198
|
Amin Yavari S, Castenmiller SM, van Strijp JAG, Croes M. Combating Implant Infections: Shifting Focus from Bacteria to Host. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002962. [PMID: 32914481 DOI: 10.1002/adma.202002962] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/28/2020] [Indexed: 05/06/2023]
Abstract
The widespread use of biomaterials to support or replace body parts is increasingly threatened by the risk of implant-associated infections. In the quest for finding novel anti-infective biomaterials, there generally has been a one-sided focus on biomaterials with direct antibacterial properties, which leads to excessive use of antibacterial agents, compromised host responses, and unpredictable effectiveness in vivo. This review sheds light on how host immunomodulation, rather than only targeting bacteria, can endow biomaterials with improved anti-infective properties. How antibacterial surface treatments are at risk to be undermined by biomaterial features that dysregulate the protection normally provided by critical immune cell subsets, namely, neutrophils and macrophages, is discussed. Accordingly, how the precise modification of biomaterial surface biophysical cues, or the incorporation of immunomodulatory drug delivery systems, can render biomaterials with the necessary immune-compatible and immune-protective properties to potentiate the host defense mechanisms is reviewed. Within this context, the protective role of host defense peptides, metallic particles, quorum sensing inhibitors, and therapeutic adjuvants is discussed. The highlighted immunomodulatory strategies may lay a foundation to develop anti-infective biomaterials, while mitigating the increasing threat of antibacterial drug resistance.
Collapse
Affiliation(s)
- Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | - Suzanne M Castenmiller
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | - Michiel Croes
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| |
Collapse
|
199
|
Watanabe T, Okitsu T, Ozawa F, Nagata S, Matsunari H, Nagashima H, Nagaya M, Teramae H, Takeuchi S. Millimeter-thick xenoislet-laden fibers as retrievable transplants mitigate foreign body reactions for long-term glycemic control in diabetic mice. Biomaterials 2020; 255:120162. [PMID: 32562943 DOI: 10.1016/j.biomaterials.2020.120162] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 05/19/2020] [Accepted: 05/29/2020] [Indexed: 12/20/2022]
Abstract
Transplantation technologies of pancreatic islets as well as stem cell-derived pancreatic beta cells encapsulated in hydrogel for the induction of immunoprotection could advance to treat type 1 diabetes mellitus, if the hydrogel transplants acquire retrievability through mitigating foreign body reactions after transplantation. Here, we demonstrate that the diameter of the fiber-shaped hydrogel transplants determines both in vivo cellular deposition onto themselves and their retrievability. Specifically, we found that the in vivo cellular deposition is significantly mitigated when the diameter is 1.0 mm and larger, and that 1.0 mm-thick xenoislet-laden fiber-shaped hydrogel transplants can be retrieved after being placed in the intraperitoneal cavities of immunocompetent diabetic mice for more than 100 days, during which period the hydrogel transplants can normalize the blood glucose concentrations of the mice. These findings could provide an innovative concept of a transplant that would promote the clinical application of stem cell-derived functional cells through improving their in vivo efficacy and safety.
Collapse
Affiliation(s)
- Takaichi Watanabe
- Institute of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Teru Okitsu
- Institute of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Fumisato Ozawa
- Institute of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Shogo Nagata
- Institute of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Hitomi Matsunari
- Meiji University International Institute for Bio-Resource Research, Kawasaki, 214-8571, Japan
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, Kawasaki, 214-8571, Japan; Laboratory of Developmental Engineering, Meiji University, Kawasaki, 214-8571, Japan
| | - Masaki Nagaya
- Meiji University International Institute for Bio-Resource Research, Kawasaki, 214-8571, Japan
| | - Hiroki Teramae
- Institute of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Shoji Takeuchi
- Institute of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan; Department of Mechano-Informatics, Graduate School of Information Science and Technology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| |
Collapse
|
200
|
Li Y, Frei AW, Yang EY, Labrada-Miravet I, Sun C, Rong Y, Samojlik MM, Bayer AL, Stabler CL. In vitro platform establishes antigen-specific CD8 + T cell cytotoxicity to encapsulated cells via indirect antigen recognition. Biomaterials 2020; 256:120182. [PMID: 32599358 PMCID: PMC7480933 DOI: 10.1016/j.biomaterials.2020.120182] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/04/2020] [Accepted: 06/06/2020] [Indexed: 02/07/2023]
Abstract
The curative potential of non-autologous cellular therapy is hindered by the requirement of anti-rejection therapy. Cellular encapsulation within nondegradable biomaterials has the potential to inhibit immune rejection, but the efficacy of this approach in robust preclinical and clinical models remains poor. While the responses of innate immune cells to the encapsulating material have been characterized, little attention has been paid to the contributions of adaptive immunity in encapsulated graft destabilization. Avoiding the limitations of animal models, we established an efficient, antigen-specific in vitro platform capable of delineating direct and indirect host T cell recognition to microencapsulated cellular grafts and evaluated their consequential impacts. Using ovalbumin (OVA) as a model antigen, we determined that alginate microencapsulation abrogates direct CD8+ T cell activation by interrupting donor-host interaction; however, indirect T cell activation, mediated by host antigen presenting cells (APCs) primed with shed donor antigens, still occurs. These activated T cells imparted cytotoxicity on the encapsulated cells, likely via diffusion of cytotoxic solutes. Overall, this platform delivers unique mechanistic insight into the impacts of hydrogel encapsulation on host adaptive immune responses, comprehensively addressing a long-standing hypothesis of the field. Furthermore, it provides an efficient benchtop screening tool for the investigation of new encapsulation methods and/or synergistic immunomodulatory agents.
Collapse
Affiliation(s)
- Ying Li
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Graduate Program in Biomedical Sciences, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Anthony W Frei
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Ethan Y Yang
- Diabetes Research Institute, College of Medicine, University of Miami, Miami, FL, USA
| | - Irayme Labrada-Miravet
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Chuqiao Sun
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Yanan Rong
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Magdalena M Samojlik
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Allison L Bayer
- Diabetes Research Institute, College of Medicine, University of Miami, Miami, FL, USA; Department of Microbiology and Immunology, University of Miami, Miami, FL, USA
| | - Cherie L Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Graduate Program in Biomedical Sciences, College of Medicine, University of Florida, Gainesville, FL, USA; University of Florida Diabetes Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|