1
|
Pham JPA, Coronel MM. Unlocking Transplant Tolerance with Biomaterials. Adv Healthc Mater 2024:e2400965. [PMID: 38843866 DOI: 10.1002/adhm.202400965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/31/2024] [Indexed: 07/04/2024]
Abstract
For patients suffering from organ failure due to injury or autoimmune disease, allogeneic organ transplantation with chronic immunosuppression is considered the god standard in terms of clinical treatment. However, the true "holy grail" of transplant immunology is operational tolerance, in which the recipient exhibits a sustained lack of alloreactivity toward unencountered antigen presented by the donor graft. This outcome is resultant from critical changes to the phenotype and genotype of the immune repertoire predicated by the activation of specific signaling pathways responsive to soluble and mechanosensitive cues. Biomaterials have emerged as a medium for interfacing with and reprogramming these endogenous pathways toward tolerance in precise, minimally invasive, and spatiotemporally defined manners. By viewing seminal and contemporary breakthroughs in transplant tolerance induction through the lens of biomaterials-mediated immunomodulation strategies-which include intrinsic material immunogenicity, the depot effect, graft coatings, induction and delivery of tolerogenic immune cells, biomimicry of tolerogenic immune cells, and in situ reprogramming-this review emphasizes the stunning diversity of approaches in the field and spotlights exciting future directions for research to come.
Collapse
Affiliation(s)
- John-Paul A Pham
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Elizabeth Caswell Diabetes Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - María M Coronel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Elizabeth Caswell Diabetes Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
2
|
Cooper DKC, Mou L, Bottino R. A brief review of the current status of pig islet xenotransplantation. Front Immunol 2024; 15:1366530. [PMID: 38464515 PMCID: PMC10920266 DOI: 10.3389/fimmu.2024.1366530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 02/07/2024] [Indexed: 03/12/2024] Open
Abstract
An estimated 1.5 million Americans suffer from Type I diabetes mellitus, and its incidence is increasing worldwide. Islet allotransplantation offers a treatment, but the availability of deceased human donor pancreases is limited. The transplantation of islets from gene-edited pigs, if successful, would resolve this problem. Pigs are now available in which the expression of the three known xenoantigens against which humans have natural (preformed) antibodies has been deleted, and in which several human 'protective' genes have been introduced. The transplantation of neonatal pig islets has some advantages over that of adult pig islets. Transplantation into the portal vein of the recipient results in loss of many islets from the instant blood-mediated inflammatory reaction (IBMIR) and so the search for an alternative site continues. The adaptive immune response can be largely suppressed by an immunosuppressive regimen based on blockade of the CD40/CD154 T cell co-stimulation pathway, whereas conventional therapy (e.g., based on tacrolimus) is less successful. We suggest that, despite the need for effective immunosuppressive therapy, the transplantation of 'free' islets will prove more successful than that of encapsulated islets. There are data to suggest that, in the absence of rejection, the function of pig islets, though less efficient than human islets, will be sufficient to maintain normoglycemia in diabetic recipients. Pig islets transplanted into immunosuppressed nonhuman primates have maintained normoglycemia for periods extending more than two years, illustrating the potential of this novel form of therapy.
Collapse
Affiliation(s)
- David K. C. Cooper
- Center for Transplantation Sciences, Massachusetts General Hospital/Harvard Medical School, Boston, MA, United States
| | - Lisha Mou
- Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
- MetaLife Center, Shenzhen Institute of Translational Medicine, Shenzhen, Guangdong, China
| | - Rita Bottino
- Imagine Islet Center, Imagine Pharma, Pittsburgh, PA, United States
| |
Collapse
|
3
|
Leishman DJ, Oppler SH, Stone LLH, O’Brien TD, Ramachandran S, Willenberg BJ, Adams AB, Hering BJ, Graham ML. Targeted mapping and utilization of the perihepatic surface for therapeutic beta cell replacement and retrieval in diabetic non-human primates. FRONTIERS IN TRANSPLANTATION 2024; 3:1352777. [PMID: 38993753 PMCID: PMC11235263 DOI: 10.3389/frtra.2024.1352777] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/09/2024] [Indexed: 07/13/2024]
Abstract
Introduction Successful diabetes reversal using pancreatic islet transplantation by various groups illustrates the significant achievements made in cell-based diabetes therapy. While clinically, intraportal islet delivery is almost exclusively used, it is not without obstacles, including instant blood-mediated inflammatory reaction (IBMIR), relative hypoxia, and loss of function over time, therefore hindering long-term success. Here we demonstrate the perihepatic surface of non-human primates (NHPs) as a potential islet delivery site maximizing favorable characteristics, including proximity to a dense vascular network for adequate oxygenation while avoiding IBMIR exposure, maintenance of portal insulin delivery, and relative ease of accessibility through minimally invasive surgery or percutaneous means. In addition, we demonstrate a targeted mapping technique of the perihepatic surface, allowing for the testing of multiple experimental conditions, including a semi-synthetic hydrogel as a possible three-dimensional framework to improve islet viability. Methods Perihepatic allo-islet cell transplants were performed in immunosuppressed cynomolgus macaques using a targeted mapping technique to test multiple conditions for biocompatibility. Transplant conditions included islets or carriers (including hydrogel, autologous plasma, and media) alone or in various combinations. Necropsy was performed at day 30, and histopathology was performed to assess biocompatibility, immune response, and islet viability. Subsequently, single-injection perihepatic allo-islet transplant was performed in immunosuppressed diabetic cynomolgus macaques. Metabolic assessments were measured frequently (i.e., blood glucose, insulin, C-peptide) until final graft retrieval for histopathology. Results Targeted mapping biocompatibility studies demonstrated mild inflammatory changes with islet-plasma constructs; however, significant inflammatory cell infiltration and fibrosis were seen surrounding sites with the hydrogel carrier affecting islet viability. In diabetic NHPs, perihepatic islet transplant using an autologous plasma carrier demonstrated prolonged function up to 6 months with improvements in blood glucose, exogenous insulin requirements, and HbA1c. Histopathology of these islets was associated with mild peri-islet mononuclear cell infiltration without evidence of rejection. Discussion The perihepatic surface serves as a viable site for islet cell transplantation demonstrating sustained islet function through 6 months. The targeted mapping approach allows for the testing of multiple conditions simultaneously to evaluate immune response to biomaterials at this site. Compared to traditional intraportal injection, the perihepatic site is a minimally invasive approach that allows the possibility for graft recovery and avoids IBMIR.
Collapse
Affiliation(s)
- David J. Leishman
- Preclinical Research Center, Department of Surgery, University of Minnesota, Minneapolis, MN, United States
| | - Scott H. Oppler
- Preclinical Research Center, Department of Surgery, University of Minnesota, Minneapolis, MN, United States
| | - Laura L. Hocum Stone
- Preclinical Research Center, Department of Surgery, University of Minnesota, Minneapolis, MN, United States
| | - Timothy D. O’Brien
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, United States
| | - Sabarinathan Ramachandran
- Preclinical Research Center, Department of Surgery, University of Minnesota, Minneapolis, MN, United States
| | - Bradley J. Willenberg
- Department of Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Andrew B. Adams
- Division of Transplantation, Department of Surgery, University of Minnesota, Minneapolis, MN, United States
| | - Bernhard J. Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, United States
| | - Melanie L. Graham
- Preclinical Research Center, Department of Surgery, University of Minnesota, Minneapolis, MN, United States
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, United States
| |
Collapse
|
4
|
Sremac M, Luo H, Deng H, Parr MFE, Hutcheson J, Verde PS, Alagpulinsa DA, Kitzmann JM, Papas KK, Brauns T, Markmann JF, Lei J, Poznansky MC. Short-term function and immune-protection of microencapsulated adult porcine islets with alginate incorporating CXCL12 in healthy and diabetic non-human primates without systemic immune suppression: A pilot study. Xenotransplantation 2023; 30:e12826. [PMID: 37712342 DOI: 10.1111/xen.12826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/10/2023] [Accepted: 08/29/2023] [Indexed: 09/16/2023]
Abstract
Replacement of insulin-producing pancreatic beta-cells by islet transplantation offers a functional cure for type-1 diabetes (T1D). We recently demonstrated that a clinical grade alginate micro-encapsulant incorporating the immune-repellent chemokine and pro-survival factor CXCL12 could protect and sustain the integrity and function of autologous islets in healthy non-human primates (NHPs) without systemic immune suppression. In this pilot study, we examined the impact of the CXCL12 micro encapsulant on the function and inflammatory and immune responses of xenogeneic islets transplanted into the omental tissue bilayer sac (OB; n = 4) and diabetic (n = 1) NHPs. Changes in the expression of cytokines after implantation were limited to 2-6-fold changes in blood, most of which did not persist over the first 4 weeks after implantation. Flow cytometry of PBMCs following transplantation showed minimal changes in IFNγ or TNFα expression on xenoantigen-specific CD4+ or CD8+ T cells compared to unstimulated cells, and these occurred mainly in the first 4 weeks. Microbeads were readily retrievable for assessment at day 90 and day 180 and at retrieval were without microscopic signs of degradation or foreign body responses (FBR). In vitro and immunohistochemistry studies of explanted microbeads indicated the presence of functional xenogeneic islets at day 30 post transplantation in all biopsied NHPs. These results from a small pilot study revealed that CXCL12-microencapsulated xenogeneic islets abrogate inflammatory and adaptive immune responses to the xenograft. This work paves the way toward future larger scale studies of the transplantation of alginate microbeads with CXCL12 and porcine or human stem cell-derived beta cells or allogeneic islets into diabetic NHPs without systemic immunosuppression.
Collapse
Affiliation(s)
- Marinko Sremac
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hao Luo
- Division of Transplant Surgery and Center of Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of General Surgery, General Hospital of Western Theater Command, Chengdu, China
| | - Hongping Deng
- Division of Transplant Surgery and Center of Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Madeline F E Parr
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Pushkar S Verde
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David A Alagpulinsa
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jenna Miner Kitzmann
- Department of Surgery, Institute for Cellular Transplantation, University of Arizona, Tucson, Arizona, USA
| | - Klearchos K Papas
- Department of Surgery, Institute for Cellular Transplantation, University of Arizona, Tucson, Arizona, USA
| | - Timothy Brauns
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - James F Markmann
- Division of Transplant Surgery and Center of Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ji Lei
- Division of Transplant Surgery and Center of Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Abbaszadeh S, Nosrati-Siahmazgi V, Musaie K, Rezaei S, Qahremani M, Xiao B, Santos HA, Shahbazi MA. Emerging strategies to bypass transplant rejection via biomaterial-assisted immunoengineering: Insights from islets and beyond. Adv Drug Deliv Rev 2023; 200:115050. [PMID: 37549847 DOI: 10.1016/j.addr.2023.115050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/14/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Novel transplantation techniques are currently under development to preserve the function of impaired tissues or organs. While current technologies can enhance the survival of recipients, they have remained elusive to date due to graft rejection by undesired in vivo immune responses despite systemic prescription of immunosuppressants. The need for life-long immunomodulation and serious adverse effects of current medicines, the development of novel biomaterial-based immunoengineering strategies has attracted much attention lately. Immunomodulatory 3D platforms can alter immune responses locally and/or prevent transplant rejection through the protection of the graft from the attack of immune system. These new approaches aim to overcome the complexity of the long-term administration of systemic immunosuppressants, including the risks of infection, cancer incidence, and systemic toxicity. In addition, they can decrease the effective dose of the delivered drugs via direct delivery at the transplantation site. In this review, we comprehensively address the immune rejection mechanisms, followed by recent developments in biomaterial-based immunoengineering strategies to prolong transplant survival. We also compare the efficacy and safety of these new platforms with conventional agents. Finally, challenges and barriers for the clinical translation of the biomaterial-based immunoengineering transplants and prospects are discussed.
Collapse
Affiliation(s)
- Samin Abbaszadeh
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Vahideh Nosrati-Siahmazgi
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Science, 45139-56184 Zanjan, Iran
| | - Kiyan Musaie
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Saman Rezaei
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Science, 45139-56184 Zanjan, Iran
| | - Mostafa Qahremani
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Science, 45139-56184 Zanjan, Iran
| | - Bo Xiao
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715 China.
| | - Hélder A Santos
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands; Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands; W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
6
|
Deng J, Yang L, Wang Z, Ouyang H, Yu H, Yuan H, Pang D. Advance of genetically modified pigs in xeno-transplantation. Front Cell Dev Biol 2022; 10:1033197. [PMID: 36299485 PMCID: PMC9590650 DOI: 10.3389/fcell.2022.1033197] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
As the standard of living improves, chronic diseases and end-stage organ failure have been a regular occurrence in human beings. Organ transplantation has become one of the hopes in the fight against chronic diseases and end-stage organ failure. However, organs available for transplantation are far from sufficient to meet the demand, leading to a major organ shortage crisis. To solve this problem, researchers have turned to pigs as their target since pigs have many advantages as xenograft donors. Pigs are considered the ideal organ donor for human xenotransplantation, but direct transplantation of porcine organs to humans faces many obstacles, such as hyperacute rejection, acute humoral xenograft rejection, coagulation dysregulation, inflammatory response, coagulation dysregulation, and endogenous porcine retroviral infection. Many transgenic strategies have been developed to overcome these obstacles. This review provides an overview of current advances in genetically modified pigs for xenotransplantation. Future genetic engineering-based delivery of safe and effective organs and tissues for xenotransplantation remains our goal.
Collapse
Affiliation(s)
- Jiacheng Deng
- College of Animal Sciences, Jilin University, Changchun, China
| | - Lin Yang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Ziru Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Hongsheng Ouyang
- College of Animal Sciences, Jilin University, Changchun, China
- Chongqing Research Institute, Jilin University, Chongqing, China
- Chongqing Jitang Biotechnology Research Institute, Chongqing, China
| | - Hao Yu
- College of Animal Sciences, Jilin University, Changchun, China
| | - Hongming Yuan
- College of Animal Sciences, Jilin University, Changchun, China
- Chongqing Research Institute, Jilin University, Chongqing, China
- Chongqing Jitang Biotechnology Research Institute, Chongqing, China
- *Correspondence: Hongming Yuan, ; Daxin Pang,
| | - Daxin Pang
- College of Animal Sciences, Jilin University, Changchun, China
- Chongqing Research Institute, Jilin University, Chongqing, China
- Chongqing Jitang Biotechnology Research Institute, Chongqing, China
- *Correspondence: Hongming Yuan, ; Daxin Pang,
| |
Collapse
|
7
|
Viability and Functionality of Neonatal Porcine Islet-like Cell Clusters Bioprinted in Alginate-Based Bioinks. Biomedicines 2022; 10:biomedicines10061420. [PMID: 35740440 PMCID: PMC9220255 DOI: 10.3390/biomedicines10061420] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
The transplantation of pancreatic islets can prevent severe long-term complications in diabetes mellitus type 1 patients. With respect to a shortage of donor organs, the transplantation of xenogeneic islets is highly attractive. To avoid rejection, islets can be encapsulated in immuno-protective hydrogel-macrocapsules, whereby 3D bioprinted structures with macropores allow for a high surface-to-volume ratio and reduced diffusion distances. In the present study, we applied 3D bioprinting to encapsulate the potentially clinically applicable neonatal porcine islet-like cell clusters (NICC) in alginate-methylcellulose. The material was additionally supplemented with bovine serum albumin or the human blood plasma derivatives platelet lysate and fresh frozen plasma. NICC were analysed for viability, proliferation, the presence of hormones, and the release of insulin in reaction to glucose stimulation. Bioprinted NICC are homogeneously distributed, remain morphologically intact, and show a comparable viability and proliferation to control NICC. The number of insulin-positive cells is comparable between the groups and over time. The amount of insulin release increases over time and is released in response to glucose stimulation over 4 weeks. In summary, we show the successful bioprinting of NICC and could demonstrate functionality over the long-term in vitro. Supplementation resulted in a trend for higher viability, but no additional benefit on functionality was observed.
Collapse
|
8
|
Chenodeoxycholic Acid Pharmacology in Biotechnology and Transplantable Pharmaceutical Applications for Tissue Delivery: An Acute Preclinical Study. Cells 2021; 10:cells10092437. [PMID: 34572086 PMCID: PMC8472107 DOI: 10.3390/cells10092437] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/30/2021] [Accepted: 09/06/2021] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Primary bile acids (PBAs) are produced and released into human gut as a result of cholesterol catabolism in the liver. A predominant PBA is chenodeoxycholic acid (CDCA), which in a recent study in our laboratory, showed significant excipient-stabilizing effects on microcapsules carrying insulinoma β-cells, in vitro, resulting in improved cell functions and insulin release, in the hyperglycemic state. Hence, this study aimed to investigate the applications of CDCA in bio-encapsulation and transplantation of primary healthy viable islets, preclinically, in type 1 diabetes. METHODS Healthy islets were harvested from balb/c mice, encapsulated in CDCA microcapsules, and transplanted into the epididymal tissues of 6 syngeneic diabetic mice, post diabetes confirmation. Pre-transplantation, the microcapsules' morphology, size, CDCA-deep layer distribution, and physical features such as swelling ratio and mechanical strength were analyzed. Post-transplantation, animals' weight, bile acids', and proinflammatory biomarkers' concentrations were analyzed. The control group was diabetic mice that were transplanted encapsulated islets (without PBA). RESULTS AND CONCLUSION Islet encapsulation by PBA microcapsules did not compromise the microcapsules' morphology or features. Furthermore, the PBA-graft performed better in terms of glycemic control and resulted in modulation of the bile acid profile in the brain. This is suggestive that the improved glycemic control was mediated via brain-related effects. However, the improvement in graft insulin delivery and glycemic control was short-term.
Collapse
|
9
|
Osteoclast-Mediated Cell Therapy as an Attempt to Treat Elastin Specific Vascular Calcification. Molecules 2021; 26:molecules26123643. [PMID: 34203711 PMCID: PMC8232296 DOI: 10.3390/molecules26123643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 01/03/2023] Open
Abstract
Inflammation and stiffness in the arteries is referred to as vascular calcification. This process is a prevalent yet poorly understood consequence of cardiovascular disease and diabetes mellitus, comorbidities with few treatments clinically available. Because this is an active process similar to bone formation, it is hypothesized that osteoclasts (OCs), bone-resorbing cells in the body, could potentially work to reverse existing calcification by resorbing bone material. The receptor activator of nuclear kappa B-ligand (RANKL) is a molecule responsible for triggering a response in monocytes and macrophages that allows them to differentiate into functional OCs. In this study, OC and RANKL delivery were employed to determine whether calcification could be attenuated. OCs were either delivered via direct injection, collagen/alginate microbeads, or collagen gel application, while RANKL was delivered via injection, through either a porcine subdermal model or aortic injury model. While in vitro results yielded a decrease in calcification using OC therapy, in vivo delivery mechanisms did not provide control or regulation to keep cells localized long enough to induce calcification reduction. However, these results do provide context and direction for the future of OC therapy, revealing necessary steps for this treatment to effectively reduce calcification in vivo. The discrepancy between in vivo and in vitro success for OC therapy points to the need for a more stable and time-controlled delivery mechanism that will allow OCs not only to remain at the site of calcification, but also to be regulated so that they are healthy and functioning normally when introduced to diseased tissue.
Collapse
|
10
|
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: 27] [Impact Index Per Article: 9.0] [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
|
11
|
Encapsulation Strategies for Pancreatic Islet Transplantation without Immune Suppression. CURRENT STEM CELL REPORTS 2021. [DOI: 10.1007/s40778-021-00190-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
12
|
Mahkam M, Bazmi Zeynabad F, Alizadeh E, Rahimi M, Rahimi F, Salehi R. Novel Methotrexate-Ciprofloxacin Loaded Alginate-Clay Based Nanocomposite as Anticancer and Antibacterial Co-Drug Delivery System. Adv Pharm Bull 2021; 11:477-489. [PMID: 34513622 PMCID: PMC8421626 DOI: 10.34172/apb.2021.055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 05/22/2020] [Accepted: 06/30/2020] [Indexed: 01/03/2023] Open
Abstract
Purpose: In last decades, by increasing multi-drug resistant microbial pathogens an urgent demand was felt in the development of novel antimicrobial agents. Methods: Promising nanocomposites composed of clay/alginate/imidazolium-based ionic liquid, have been developed via intercalation of calcium alginate and ionic liquid by ion exchange method. These tailored nanocomposites were used as nanocarriers to simultaneously deliver methotrexate (MTX), and ciprofloxacin (CIP), as anticancer and antibacterial agents, respectively to MCF-7 breast cancer cells. Nanocomposites were fully characterized by scanning electron microscopy studies (SEM), X-ray diffraction (XRD), Fourier transforms infrared (FTIR) spectroscopy, and thermogravimetric analysis (TGA) methods. The in vitro antimicrobial potential of the mentioned nanocomposites in free and dual-drug loaded form was investigated on Pseudomonas aeruginosa and Escherichia coli bacteria. The antitumor activity of nano-formulations was evaluated by both MTT assay and cell cycle arrest. Results: The dual drug-loaded nanocomposites with exceptionally high loading efficiency (MTX: 99 ±0.4% and CIP: 98 ±1.2%) and mean particle size of 70 nm were obtained with obvious pH-responsive MTX and CIP release (both drugs release rate was increased at pH 5.8 compared to 7.4). The antibacterial activity of CIP-loaded nanocomposites was significantly higher in comparison with free CIP (P <0.001). The antitumor activity results revealed that MTX cytotoxicity on MCF-7 cells was significantly higher in nano-formulations compared to free MTX (P <0.001). Both MTX-loaded nanocomposites caused S-phase arrest in MCF-7 cells compared to non-treated cells (P ˂ 0.001). Conclusion: Newly developed smart nanocomposites are potentially effective pH-sustainable delivery systems for enhanced tumor therapy.
Collapse
Affiliation(s)
- Mehrdad Mahkam
- Chemistry Department, Azarbaijan Shahid Madani University, Tabriz, Iran
| | | | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Rahimi
- Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Fariborz Rahimi
- Department of Electrical Engineering, University of Bonab, Bonab, Iran
| | - Roya Salehi
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
13
|
Izeia L, Eufrasio-da-Silva T, Dolatshahi-Pirouz A, Ostrovidov S, Paolone G, Peppas NA, De Vos P, Emerich D, Orive G. Cell-laden alginate hydrogels for the treatment of diabetes. Expert Opin Drug Deliv 2021; 17:1113-1118. [PMID: 32515621 DOI: 10.1080/17425247.2020.1778667] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Diabetes mellitus is an ever-increasing medical condition that currently suffers 1 of 11 adults who may have lifelong commitment with insulin injections. Cell-laden hydrogels releasing insulin may provide the ultimate means of correcting diabetes. Here, we provide insights of this cell-based approach including latest preclinical and clinical progress both from academia and industry. AREA COVERED The present article focuses on reviewing latest advances in cell-laden hydrogels both from the technological and biological perspective. The most relevant clinical results including clinical trials are also discussed. EXPERT OPINION Current progress in technological issues (stem cells, devices, biomaterials) have contributed cell encapsulation science to have a very relevant progress in the field of diabetes treatment.
Collapse
Affiliation(s)
- Lukin Izeia
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU , Vitoria-Gasteiz, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) , Vitoria-Gasteiz, Spain
| | - Tatiane Eufrasio-da-Silva
- Department of Dentistry - Regenerative Biomaterials, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen, The Netherlands
| | - Alireza Dolatshahi-Pirouz
- Department of Dentistry - Regenerative Biomaterials, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen, The Netherlands.,Department of Health Technology, Institute of Biotherapeutic Engineering and Drug Targeting, Center for Intestinal Absorption and Transport of Biopharmaceuticals Technical University of Denmark , Lyngby, Denmark
| | - Serge Ostrovidov
- Center for Minimally Invasive Therapeutics (C-MIT) Department of Radiological Sciences, University of California , Los Angeles, CA, USA
| | - Giovanna Paolone
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona , Verona, Italy
| | - Nicholas A Peppas
- Departments of Pharmaceutics, Chemical and Biomedical Engineering, The University of Texas at Austin , Austin, TX, USA
| | - Paul De Vos
- Pathology and Medical Biology Section, Immunoendocrinology, University of Groningen , Groningen, The Netherlands
| | - Dwaine Emerich
- Gloriana Therapeutics, Inc. (Formerly NsGene Inc.) , Providence, RI, USA
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU , Vitoria-Gasteiz, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) , Vitoria-Gasteiz, Spain.,University Institute for Regenerative Medicine and Oral Implantology - UIRMI (Upv/ehu-fundación Eduardo Anitua) , Vitoria, Spain.,The Academia, Singapore Eye Research Institute , Discovery Tower, Singapore
| |
Collapse
|
14
|
Huang CP, Yang CY, Shyr CR. Utilizing Xenogeneic Cells As a Therapeutic Agent for Treating Diseases. Cell Transplant 2021; 30:9636897211011995. [PMID: 33975464 PMCID: PMC8120531 DOI: 10.1177/09636897211011995] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/19/2021] [Accepted: 04/05/2021] [Indexed: 12/27/2022] Open
Abstract
The utilization of biologically produced cells to treat diseases is a revolutionary invention in modern medicine after chemically synthesized small molecule drugs and biochemically made protein drugs. Cells are basic units of life with diverse functions in mature and developing organs, which biological properties could be utilized as a promising therapeutic approach for currently intractable and incurable diseases. Xenogeneic cell therapy utilizing animal cells other than human for medicinal purpose has been studied as a new way of treating diseases. Xenogeneic cell therapy is considered as a potential regenerative approach to fulfill current unmet medical needs because xenogeneic cells could be isolated from different animal organs and expanded ex vivo as well as maintain the characteristics of original organs, providing a versatile and plenty cell source for cell-based therapeutics beside autologous and allogeneic sources. The swine species is considered the most suitable source because of the similarity with humans in size and physiology of many organs in addition to the economic and ethical reasons plus the possibility of genetic modification. This review discusses the old proposed uses of xenogeneic cells such as xenogeneic pancreatic islet cells, hepatocytes and neuronal cells as a living drug for the treatment of degenerative and organ failure diseases. Novel applications of xenogeneic mesenchymal stroma cells and urothelial cells are also discussed. There are formidable immunological barriers toward successful cellular xenotransplantation in clinic despite major progress in the development of novel immunosuppression regimens and genetically multimodified donor pigs. However, immunological barriers could be turn into immune boosters by using xenogeneic cells of specific tissue types as a novel immunotherapeutic agent to elicit bystander antitumor immunity due to rejection immune responses. Xenogeneic cells have the potential to become a safe and efficacious option for intractable diseases and hard-to-treat cancers, adding a new class of cellular medicine in our drug armamentarium.
Collapse
Affiliation(s)
- Chi-Ping Huang
- Department of Urology, School of Medicine, China Medical University and Hospital, Taichung, Taiwan
| | - Chi-Yu Yang
- Animal Technology Research Center/Division of Animal Technology, Agriculture Technology Research Institute, Miaoli, Taiwan
| | - Chih-Rong Shyr
- Sex Hormone Research Center, Department of Medical Laboratory Science and Biotechnology, China Medical University and Hospital, Taichung, Taiwan
| |
Collapse
|
15
|
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: 25] [Impact Index Per Article: 6.3] [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
|
16
|
Elnashar M, Vaccarezza M, Al-Salami H. Cutting-edge biotechnological advancement in islet delivery using pancreatic and cellular approaches. Future Sci OA 2020; 7:FSO660. [PMID: 33552541 PMCID: PMC7849926 DOI: 10.2144/fsoa-2020-0105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
There are approximately 1 billion prediabetic people worldwide, and the global cost for diabetes mellitus (DM) is estimated to be $825 billion. In regard to Type 1 DM, transplanting a whole pancreas or its islets has gained the attention of researchers in the last few decades. Recent studies showed that islet transplantation (ILT) containing insulin-producing β cells is the most notable advancement cure for Type 1 DM. However, this procedure has been hindered by shortage and lack of sufficient islet donors and the need for long-term immunosuppression of any potential graft rejection. The strategy of encapsulation may avoid the rejection of stem-cell-derived allogeneic islets or xenogeneic islets. This review article describes various biotechnology features in encapsulation-of-islet-cell therapy for humans, including the use of bile acids.
Collapse
Affiliation(s)
- Magdy Elnashar
- Biotechnology & Drug Development Research Laboratory, School of Pharmacy & Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.,Centre of Excellence, Department of Polymers, National Research Centre, Cairo, Egypt
| | - Mauro Vaccarezza
- School of Pharmacy & Biomedical Science, Faculty of Health Sciences, Curtin University, Bentley, Perth, WA 6102, Australia
| | - Hani Al-Salami
- Biotechnology & Drug Development Research Laboratory, School of Pharmacy & Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| |
Collapse
|
17
|
Kupikowska-Stobba B, Lewińska D. Polymer microcapsules and microbeads as cell carriers for in vivo biomedical applications. Biomater Sci 2020; 8:1536-1574. [PMID: 32110789 DOI: 10.1039/c9bm01337g] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Polymer microcarriers are being extensively explored as cell delivery vehicles in cell-based therapies and hybrid tissue and organ engineering. Spherical microcarriers are of particular interest due to easy fabrication and injectability. They include microbeads, composed of a porous matrix, and microcapsules, where matrix core is additionally covered with a semipermeable membrane. Microcarriers provide cell containment at implantation site and protect the cells from host immunoresponse, degradation and shear stress. Immobilized cells may be genetically altered to release a specific therapeutic product directly at the target site, eliminating side effects of systemic therapies. Cell microcarriers need to fulfil a number of extremely high standards regarding their biocompatibility, cytocompatibility, immunoisolating capacity, transport, mechanical and chemical properties. To obtain cell microcarriers of specified parameters, a wide variety of polymers, both natural and synthetic, and immobilization methods can be applied. Yet so far, only a few approaches based on cell-laden microcarriers have reached clinical trials. The main issue that still impedes progress of these systems towards clinical application is limited cell survival in vivo. Herein, we review polymer biomaterials and methods used for fabrication of cell microcarriers for in vivo biomedical applications. We describe their key limitations and modifications aiming at improvement of microcarrier in vivo performance. We also present the main applications of polymer cell microcarriers in regenerative medicine, pancreatic islet and hepatocyte transplantation and in the treatment of cancer. Lastly, we outline the main challenges in cell microimmobilization for biomedical purposes, the strategies to overcome these issues and potential future improvements in this area.
Collapse
Affiliation(s)
- Barbara Kupikowska-Stobba
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| | - Dorota Lewińska
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| |
Collapse
|
18
|
Beh CW, Fu Y, Weiss CR, Hu C, Arepally A, Mao HQ, Wang TH, Kraitchman DL. Microfluidic-prepared, monodisperse, X-ray-visible, embolic microspheres for non-oncological embolization applications. LAB ON A CHIP 2020; 20:3591-3600. [PMID: 32869821 PMCID: PMC7531348 DOI: 10.1039/d0lc00098a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Embolotherapy using particle embolics is normally performed with exogenous contrast to assist in visualization. However, the exact location of the embolics cannot be identified after contrast washout. We developed a novel, pseudo-check valve-integrated microfluidic device, that partitions barium- impregnated alginate from crosslinking solution, thereby preventing nozzle failure. This enables rapid and continuous generation of inherently X-ray-visible embolic microspheres (XEMs) with uniform size. The XEMs are visible under clinical X-ray and cone beam CT both in vitro and in vivo. In particular, we demonstrated the embolization properties of these XEMs in large animals, performing direct intra- and post-procedural assessment of embolic delivery. The persistent radiopacity of these XEMs enables real-time evaluation of embolization precision and offers great promise for non-invasive follow-up examination without exogenous contrast. We also demonstrated that bariatric arterial embolization with XEMs significantly suppresses weight gain in swine, as an example of a non-oncological application of embolotherapy.
Collapse
Affiliation(s)
- Cyrus W Beh
- Department of Biomedical Engineering, Johns Hopkins University, 3400 N, Charles St, Baltimore, MD 21218, USA.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Laporte C, Tubbs E, Pierron M, Gallego A, Moisan A, Lamarche F, Lozano T, Hernandez A, Cottet-Rousselle C, Gauchez AS, Persoons V, Bottausci F, Fontelaye C, Boizot F, Lablanche S, Rivera F. Improved human islets’ viability and functionality with mesenchymal stem cells and arg-gly-asp tripeptides supplementation of alginate micro-encapsulated islets in vitro. Biochem Biophys Res Commun 2020; 528:650-657. [DOI: 10.1016/j.bbrc.2020.05.107] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 05/15/2020] [Indexed: 12/19/2022]
|
20
|
Marfil‐Garza BA, Polishevska K, Pepper AR, Korbutt GS. Current State and Evidence of Cellular Encapsulation Strategies in Type 1 Diabetes. Compr Physiol 2020; 10:839-878. [DOI: 10.1002/cphy.c190033] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
21
|
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: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
22
|
Delcassian D, Luzhansky I, Spanoudaki V, Bochenek M, McGladrigan C, Nguyen A, Norcross S, Zhu Y, Shan CS, Hausser R, Shakesheff KM, Langer R, Anderson DG. Magnetic Retrieval of Encapsulated Beta Cell Transplants from Diabetic Mice Using Dual-Function MRI Visible and Retrievable Microcapsules. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1904502. [PMID: 32134138 DOI: 10.1002/adma.201904502] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 12/03/2019] [Indexed: 05/18/2023]
Abstract
Encapsulated beta cell transplantation offers a potential cure for a subset of diabetic patients. Once transplanted, beta cell grafts can help to restore glycemic control; however, locating and retrieving cells in the event of graft failure may pose a surgical challenge. Here, a dual-function nanoparticle-loaded hydrogel microcapsule is developed that enables graft retrieval under an applied magnetic field. Additionally, this system facilitates graft localization via magnetic resonance imaging (MRI), and graft isolation from the immune system. Iron oxide nanoparticles encapsulated within alginate hydrogel capsules containing viable islets are transplanted and the in vitro and in vivo retrieval of capsules containing nanoparticles functionalized with various ligands are compared. Capsules containing islets co-encapsulated with COOH-coated nanoparticles restore normal glycemia in immunocompetent diabetic mice for at least 6 weeks, can be visualized using MRI, and are retrievable in a magnetic field. Application of a magnetic field for 90 s via a magnetically assisted retrieval device facilitates rapid retrieval of up to 94% (±3.1%) of the transplant volume 24 h after surgical implantation. This strategy aids monitoring of cell-capsule locations in vivo, facilitates graft removal at the end of the transplant lifetime, and may be applicable to many encapsulated cell transplant systems.
Collapse
Affiliation(s)
- Derfogail Delcassian
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Division of Regenerative Medicine and Cellular Therapies, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Igor Luzhansky
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Virginia Spanoudaki
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Matthew Bochenek
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Collin McGladrigan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Amy Nguyen
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Samuel Norcross
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Yuhan Zhu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Crystal Shuo Shan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Reed Hausser
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Kevin M Shakesheff
- Division of Regenerative Medicine and Cellular Therapies, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| |
Collapse
|
23
|
Safley SA, Graham ML, Weegman BP, Einstein SA, Barber GF, Janecek JJ, Mutch LA, Singh A, Ramachandran S, Garwood M, Sambanis A, Papas KK, Hering BJ, Weber CJ. Noninvasive Fluorine-19 Magnetic Resonance Relaxometry Measurement of the Partial Pressure of Oxygen in Acellular Perfluorochemical-loaded Alginate Microcapsules Implanted in the Peritoneal Cavity of Nonhuman Primates. Transplantation 2020; 104:259-269. [PMID: 31385927 PMCID: PMC6994361 DOI: 10.1097/tp.0000000000002896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND We have utilized a noninvasive technique for measuring the partial pressure of oxygen (pO2) in alginate microcapsules implanted intraperitoneally in healthy nonhuman primates (NHPs). Average pO2 is important for determining if a transplant site and capsules with certain passive diffusion characteristics can support the islet viability, metabolic activity, and dose necessary to reverse diabetes. METHODS Perfluoro-15-crown-5-ether alginate capsules were infused intraperitoneally into 3 healthy NHPs. Peritoneal pO2 levels were measured on days 0 and 7 using fluorine-19 magnetic resonance relaxometry and a fiber-optic probe. Fluorine-19 MRI was used to determine the locations of capsules within the peritoneal space on days 0 and 7. Gross and histologic evaluations of the capsules were used to assess their biocompatibility postmortem. RESULTS At day 0 immediately after infusion of capsules equilibrated to room air, capsules were concentrated near the infusion site, and the pO2 measurement using magnetic resonance relaxometry was 147 ± 9 mm Hg. On day 7 after capsules were dispersed throughout the peritoneal cavity, the pO2 level was 61 ± 11 mm Hg. Measurements using the fiber-optic oxygen sensor were 132 ± 7.5 mm Hg (day 0) and 89 ± 6.1 mm Hg (day 7). Perfluoro-15-crown-5-ether capsules retrieved on day 7 were intact and free-floating without host cell attachment, although the numbers of peritoneal CD20 B cells, CD4 and CD8 T cells, and CD14 macrophages increased consistent with a mild foreign body reaction. CONCLUSIONS The peritoneal pO2 of normal NHPs is relatively low and we predict would decrease further when encapsulated islets are transplanted intraperitoneally.
Collapse
Affiliation(s)
| | - Melanie L. Graham
- Preclinical Research Center, Department of Surgery, University of Minnesota, St. Paul, MN
| | - Bradley P. Weegman
- Sylvatica Biotech, Inc., Charleston, SC
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
| | - Samuel A. Einstein
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Jody J. Janecek
- Preclinical Research Center, Department of Surgery, University of Minnesota, St. Paul, MN
| | - Lucas A. Mutch
- Preclinical Research Center, Department of Surgery, University of Minnesota, St. Paul, MN
| | - Amar Singh
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | | | - Michael Garwood
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
| | | | | | - Bernhard J. Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | | |
Collapse
|
24
|
Liu Q, Chiu A, Wang LH, An D, Zhong M, Smink AM, de Haan BJ, de Vos P, Keane K, Vegge A, Chen EY, Song W, Liu WF, Flanders J, Rescan C, Grunnet LG, Wang X, Ma M. Zwitterionically modified alginates mitigate cellular overgrowth for cell encapsulation. Nat Commun 2019; 10:5262. [PMID: 31748525 PMCID: PMC6868136 DOI: 10.1038/s41467-019-13238-7] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 10/30/2019] [Indexed: 01/16/2023] Open
Abstract
Foreign body reaction (FBR) to implanted biomaterials and medical devices is common and can compromise the function of implants or cause complications. For example, in cell encapsulation, cellular overgrowth (CO) and fibrosis around the cellular constructs can reduce the mass transfer of oxygen, nutrients and metabolic wastes, undermining cell function and leading to transplant failure. Therefore, materials that mitigate FBR or CO will have broad applications in biomedicine. Here we report a group of zwitterionic, sulfobetaine (SB) and carboxybetaine (CB) modifications of alginates that reproducibly mitigate the CO of implanted alginate microcapsules in mice, dogs and pigs. Using the modified alginates (SB-alginates), we also demonstrate improved outcome of islet encapsulation in a chemically-induced diabetic mouse model. These zwitterion-modified alginates may contribute to the development of cell encapsulation therapies for type 1 diabetes and other hormone-deficient diseases.
Collapse
Affiliation(s)
- Qingsheng Liu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Alan Chiu
- 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
| | - Duo An
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Monica Zhong
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Alexandra M Smink
- Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Bart J de Haan
- Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Paul de Vos
- Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Kevin Keane
- Stem Cell Biology, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Andreas Vegge
- Diabetes Research, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Esther Y Chen
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, 92697, USA
| | - Wei Song
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Wendy F Liu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, 92697, USA
| | - James Flanders
- Department of Clinical Sciences, Cornell University, Ithaca, NY, 14853, USA
| | - Claude Rescan
- Stem Cell Pharmacology, Novo Nordisk A/S, 2760, Måløv, Denmark
| | | | - Xi Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
25
|
Toda S, Fattah A, Asawa K, Nakamura N, N. Ekdahl K, Nilsson B, Teramura Y. Optimization of Islet Microencapsulation with Thin Polymer Membranes for Long-Term Stability. MICROMACHINES 2019; 10:E755. [PMID: 31698737 PMCID: PMC6915491 DOI: 10.3390/mi10110755] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 10/30/2019] [Accepted: 11/04/2019] [Indexed: 12/23/2022]
Abstract
Microencapsulation of islets can protect against immune reactions from the host immune system after transplantation. However, sufficient numbers of islets cannot be transplanted due to the increase of the size and total volume. Therefore, thin and stable polymer membranes are required for the microencapsulation. Here, we undertook the cell microencapsulation using poly(ethylene glycol)-conjugated phospholipid (PEG-lipid) and layer-by-layer membrane of multiple-arm PEG. In order to examine the membrane stability, we used different molecular weights of 4-arm PEG (10k, 20k and 40k)-Mal to examine the influence on the polymer membrane stability. We found that the polymer membrane made of 4-arm PEG(40k)-Mal showed the highest stability on the cell surface. Also, the polymer membrane did not disturb the insulin secretion from beta cells.
Collapse
Affiliation(s)
- Shota Toda
- Department of Bioscience and Engineering, College of Systems Engineering and Science, Shibaura Institute of Technology, Saitama 337-8570, Japan; (S.T.); (N.N.)
| | - Artin Fattah
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden; (A.F.); (K.N.E.); (B.N.)
| | - Kenta Asawa
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan;
| | - Naoko Nakamura
- Department of Bioscience and Engineering, College of Systems Engineering and Science, Shibaura Institute of Technology, Saitama 337-8570, Japan; (S.T.); (N.N.)
| | - Kristina N. Ekdahl
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden; (A.F.); (K.N.E.); (B.N.)
- Linnaeus Center of Biomaterials Chemistry, Linnaeus University, SE-391 82 Kalmar, Sweden
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden; (A.F.); (K.N.E.); (B.N.)
| | - Yuji Teramura
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden; (A.F.); (K.N.E.); (B.N.)
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan;
| |
Collapse
|
26
|
Bowers DT, Song W, Wang LH, Ma M. Engineering the vasculature for islet transplantation. Acta Biomater 2019; 95:131-151. [PMID: 31128322 PMCID: PMC6824722 DOI: 10.1016/j.actbio.2019.05.051] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 04/13/2019] [Accepted: 05/20/2019] [Indexed: 12/17/2022]
Abstract
The microvasculature in the pancreatic islet is highly specialized for glucose sensing and insulin secretion. Although pancreatic islet transplantation is a potentially life-changing treatment for patients with insulin-dependent diabetes, a lack of blood perfusion reduces viability and function of newly transplanted tissues. Functional vasculature around an implant is not only necessary for the supply of oxygen and nutrients but also required for rapid insulin release kinetics and removal of metabolic waste. Inadequate vascularization is particularly a challenge in islet encapsulation. Selectively permeable membranes increase the barrier to diffusion and often elicit a foreign body reaction including a fibrotic capsule that is not well vascularized. Therefore, approaches that aid in the rapid formation of a mature and robust vasculature in close proximity to the transplanted cells are crucial for successful islet transplantation or other cellular therapies. In this paper, we review various strategies to engineer vasculature for islet transplantation. We consider properties of materials (both synthetic and naturally derived), prevascularization, local release of proangiogenic factors, and co-transplantation of vascular cells that have all been harnessed to increase vasculature. We then discuss the various other challenges in engineering mature, long-term functional and clinically viable vasculature as well as some emerging technologies developed to address them. The benefits of physiological glucose control for patients and the healthcare system demand vigorous pursuit of solutions to cell transplant challenges. STATEMENT OF SIGNIFICANCE: Insulin-dependent diabetes affects more than 1.25 million people in the United States alone. Pancreatic islets secrete insulin and other endocrine hormones that control glucose to normal levels. During preparation for transplantation, the specialized islet blood vessel supply is lost. Furthermore, in the case of cell encapsulation, cells are protected within a device, further limiting delivery of nutrients and absorption of hormones. To overcome these issues, this review considers methods to rapidly vascularize sites and implants through material properties, pre-vascularization, delivery of growth factors, or co-transplantation of vessel supporting cells. Other challenges and emerging technologies are also discussed. Proper vascular growth is a significant component of successful islet transplantation, a treatment that can provide life-changing benefits to patients.
Collapse
Affiliation(s)
- Daniel T Bowers
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Wei Song
- 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
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
27
|
Pathak S, Pham TT, Jeong JH, Byun Y. Immunoisolation of pancreatic islets via thin-layer surface modification. J Control Release 2019; 305:176-193. [DOI: 10.1016/j.jconrel.2019.04.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/15/2019] [Accepted: 04/22/2019] [Indexed: 12/13/2022]
|
28
|
Hu S, de Vos P. Polymeric Approaches to Reduce Tissue Responses Against Devices Applied for Islet-Cell Encapsulation. Front Bioeng Biotechnol 2019; 7:134. [PMID: 31214587 PMCID: PMC6558039 DOI: 10.3389/fbioe.2019.00134] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/20/2019] [Indexed: 12/15/2022] Open
Abstract
Immunoisolation of pancreatic islets is a technology in which islets are encapsulated in semipermeable but immunoprotective polymeric membranes. The technology allows for successful transplantation of insulin-producing cells in the absence of immunosuppression. Different approaches of immunoisolation are currently under development. These approaches involve intravascular devices that are connected to the bloodstream and extravascular devices that can be distinguished in micro- and macrocapsules and are usually implanted in the peritoneal cavity or under the skin. The technology has been subject of intense fundamental research in the past decade. It has co-evolved with novel replenishable cell sources for cure of diseases such as Type 1 Diabetes Mellitus that need to be protected for the host immune system. Although the devices have shown significant success in animal models and even in human safety studies most technologies still suffer from undesired tissue responses in the host. Here we review the past and current approaches to modulate and reduce tissue responses against extravascular cell-containing micro- and macrocapsules with a focus on rational choices for polymer (combinations). Choices for polymers but also choices for crosslinking agents that induce more stable and biocompatible capsules are discussed. Combining beneficial properties of molecules in diblock polymers or application of these molecules or other anti-biofouling molecules have been reviewed. Emerging are also the principles of polymer brushes that prevent protein and cell-adhesion. Recently also immunomodulating biomaterials that bind to specific immune receptors have entered the field. Several natural and synthetic polymers and even combinations of these polymers have demonstrated significant improvement in outcomes of encapsulated grafts. Adequate polymeric surface properties have been shown to be essential but how the surface should be composed to avoid host responses remains to be identified. Current insight is that optimal biocompatible devices can be created which raises optimism that immunoisolating devices can be created that allows for long term survival of encapsulated replenishable insulin-producing cell sources for treatment of Type 1 Diabetes Mellitus.
Collapse
Affiliation(s)
- Shuixan Hu
- Division of Medical Biology, Department of Pathology and Medical Biology, Immunoendocrinology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | | |
Collapse
|
29
|
Cao R, Avgoustiniatos E, Papas K, de Vos P, Lakey JRT. Mathematical predictions of oxygen availability in micro- and macro-encapsulated human and porcine pancreatic islets. J Biomed Mater Res B Appl Biomater 2019; 108:343-352. [PMID: 31013399 DOI: 10.1002/jbm.b.34393] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 02/12/2019] [Accepted: 04/04/2019] [Indexed: 12/22/2022]
Abstract
Optimal function of immunoisolated islets requires adequate supply of oxygen to metabolically active insulin producing beta-cells. Using mathematical modeling, we investigated the influence of the pO2 on islet insulin secretory capacity and evaluated conditions that could lead to the development of tissue anoxia, modeled for a 300 μm islet in a 500 μm microcapsule or a 500 μm planar, slab-shaped macrocapsule. The pO2 was used to assess the part of islets that contributed to insulin secretion. Assuming a 500 μm macrocapsule with a 300 μm islet, with oxygen consumption rate (OCR) of 100-300 nmol min-1 mg-1 DNA, islets did not develop any necrotic core. The nonfunctional zone (with no insulin secretion if pO2 < 0.1 mmHg) was 0.3% for human islets (OCR ~100 nmol/min/mg DNA) and 35% for porcine islets (OCR ~300 nmol/min/mg DNA). The OCR of the islet preparation is profoundly affected by islet size, with optimal size of <250 μm in diameter (human) or <150 μm (porcine). Our data suggest that microcapsules afford superior oxygen delivery to encapsulated islets than macrocapsules, and optimal islet function can be achieved by encapsulating multiple, small (<150 μm) islets with OCR of ~100 nmol min-1 mg-1 DNA (human islets) or ~200 nmol min-1 mg-1 DNA (porcine islets).
Collapse
Affiliation(s)
- Rui Cao
- Department of Surgery, University of California, Irvine, Orange, California
| | | | - Klearchos Papas
- Department of Surgery, University of Arizona, Tucson, Arizona
| | - Paul de Vos
- Departments of Pathology and Laboratory Medicine, Division of Immuno-Endocrinology, University of Groningen, Groningen, The Netherlands
| | - Jonathan R T Lakey
- Department of Surgery, University of California, Irvine, Orange, California
- Department of Biomedical Engineering, University of California, Irvine, California
| |
Collapse
|
30
|
Preliminary Studies of the Impact of CXCL12 on the Foreign Body Reaction to Pancreatic Islets Microencapsulated in Alginate in Nonhuman Primates. Transplant Direct 2019; 5:e447. [PMID: 31165082 PMCID: PMC6511446 DOI: 10.1097/txd.0000000000000890] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 03/04/2019] [Indexed: 12/18/2022] Open
Abstract
Supplemental Digital Content is available in the text. Background. We previously demonstrated that the incorporation of the chemokine CXCL12 into alginate microbeads supported long-term survival of microencapsulated auto-, allo-, and xenogeneic islets in murine models of diabetes without systemic immune suppression. The purpose of this study was to test whether CXCL12 could abrogate foreign body responses (FBRs) against alginate microbeads which were empty or contained autologous islets in healthy nonhuman primates (NHPs; n = 4). Methods. Two NHPs received intraperitoneal implants of 400 000 alginate microbeads with or without CXCL12, and postimplantation immunological and histopathological changes were evaluated up to 6 months postimplantation. A similar evaluation of autologous islets in CXCL12-containing alginate microbeads was performed in NHPs (n = 2). Results. CXCL12-containing alginate microbeads were associated with a markedly reduced FBR to microbeads. Host responses to microbead implants were minimal, as assessed by clinical observations, blood counts, and chemistry. Evaluation of encapsulated islets was limited by the development of necrotizing pancreatitis after hemipancreatectomy in 1 NHP. A limited number of functioning islets were detectable at 6 months posttransplantation in the second NHP. In general, empty microbeads or islet-containing beads were found to be evenly distributed through the intraperitoneal cavity and did not accumulate in the Pouch of Douglas. Conclusions. Inclusion of CXCL12 in alginate microbeads minimized localized FBR. The NHP autologous islet implant model had limited utility for excluding inflammatory/immune responses to implanted islets because of the complexity of pancreatic surgery (hemipancreatectomy) before transplantation and the need to microencapsulate and transplant encapsulated autologous islets immediately after pancreatectomy and islet isolation.
Collapse
|
31
|
Cao Y, Hassan M, Cheng Y, Chen Z, Wang M, Zhang X, Haider Z, Zhao G. Multifunctional Photo- and Magnetoresponsive Graphene Oxide-Fe 3O 4 Nanocomposite-Alginate Hydrogel Platform for Ice Recrystallization Inhibition. ACS APPLIED MATERIALS & INTERFACES 2019; 11:12379-12388. [PMID: 30865418 DOI: 10.1021/acsami.9b02887] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Tuning ice recrystallization (IR) has attracted tremendous interest in fundamental research and a variety of practical applications, including food and pharmaceutical engineering, fabrication of anti-icing coating and porous materials, and cryopreservation of biological cells and tissues. Although great efforts have been devoted to modulation of IR for better microstructure control of various materials, it still remains a challenge, especially in cryopreservation, where insufficient suppression of IR during warming is fatal to the cells. Herein, we report an all-in-one platform, combining the external physical fields and the functional materials for both active and passive suppression of IR, where the photo- and magnetothermal dual-modal heating of GO-Fe3O4 nanocomposites (NCs) can be used to suppress IR with both enhanced global warming and microscale thermal disturbance. Moreover, the materials alginate hydrogels and GO-Fe3O4 NCs can act as IR inhibitors for further suppression of the IR effect. As a typical application, we show that this GO-Fe3O4 nanocomposite-alginate hydrogel platform can successfully enable low-cryoprotectant, high-quality vitrification of stem cell-laden hydrogels. We believe that the versatile ice recrystallization inhibition platform will have a profound influence on cryopreservation and tremendously facilitate stem cell-based medicine to meet its ever-increasing demand in clinical settings.
Collapse
Affiliation(s)
- Yuan Cao
- Department of Electronic Science and Technology , University of Science and Technology of China , Hefei 230027 , Anhui , China
| | - Muhammad Hassan
- Division of Nanomaterials & Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, Department of Chemistry , University of Science and Technology of China , Hefei 230026 , China
| | - Yue Cheng
- Department of Electronic Science and Technology , University of Science and Technology of China , Hefei 230027 , Anhui , China
| | - Zhongrong Chen
- Department of Electronic Science and Technology , University of Science and Technology of China , Hefei 230027 , Anhui , China
| | - Meng Wang
- Department of Electronic Science and Technology , University of Science and Technology of China , Hefei 230027 , Anhui , China
| | - Xiaozhang Zhang
- Department of Electronic Science and Technology , University of Science and Technology of China , Hefei 230027 , Anhui , China
| | - Zeeshan Haider
- Department of Electronic Science and Technology , University of Science and Technology of China , Hefei 230027 , Anhui , China
| | - Gang Zhao
- Department of Electronic Science and Technology , University of Science and Technology of China , Hefei 230027 , Anhui , China
| |
Collapse
|
32
|
Noninvasive Monitoring of Allogeneic Stem Cell Delivery with Dual-Modality Imaging-Visible Microcapsules in a Rabbit Model of Peripheral Arterial Disease. Stem Cells Int 2019; 2019:9732319. [PMID: 31001343 PMCID: PMC6437732 DOI: 10.1155/2019/9732319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/06/2019] [Accepted: 01/28/2019] [Indexed: 01/08/2023] Open
Abstract
Stem cell therapies, although promising for treating peripheral arterial disease (PAD), often suffer from low engraftment rates and the inability to confirm the delivery success and track cell distribution and engraftment. Stem cell microencapsulation combined with imaging contrast agents may provide a means to simultaneously enhance cell survival and enable cell tracking with noninvasive imaging. Here, we have evaluated a novel MRI- and X-ray-visible microcapsule formulation for allogeneic mesenchymal stem cell (MSC) delivery and tracking in a large animal model. Bone marrow-derived MSCs from male New Zealand White rabbits were encapsulated using a modified cell encapsulation method to incorporate a dual-modality imaging contrast agent, perfluorooctyl bromide (PFOB). PFOB microcapsules (PFOBCaps) were then transplanted into the medial thigh of normal or PAD female rabbits. In vitro MSC viability remained high (79 ± 5% at 4 weeks of postencapsulation), and as few as two and ten PFOBCaps could be detected in phantoms using clinical C-arm CT and 19F MRI, respectively. Successful injections of PFOBCaps in the medial thigh of normal (n = 15) and PAD (n = 16) rabbits were demonstrated on C-arm CT at 1-14 days of postinjection. Using 19F MRI, transplanted PFOBCaps were clearly identified as “hot spots” and showed one-to-one correspondence to the radiopacities on C-arm CT. Concordance of 19F MRI and C-arm CT locations of PFOBCaps with postmortem locations was high (95%). Immunohistological analysis revealed high MSC survival in PFOBCaps (>56%) two weeks after transplantation while naked MSCs were no longer viable beyond three days after delivery. These findings demonstrate that PFOBCaps could maintain cell viability even in the ischemic tissue and provide a means to monitor cell delivery and track engraftment using clinical noninvasive imaging systems.
Collapse
|
33
|
Li X, Meng Q, Zhang L. Overcoming Immunobiological Barriers Against Porcine Islet Xenografts: What Should Be Done? Pancreas 2019; 48:299-308. [PMID: 30855426 DOI: 10.1097/mpa.0000000000001259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Porcine islets might represent an ideal solution to the severe shortage of living donor islets available for transplantation and thus have great potential for the treatment of diabetes. Although tremendous progress has been achieved through recent experiments, the immune response remains a major obstacle. This review first describes the 3 major pathways of rejection: hyperacute rejection mediated by preformed natural antibodies and complement, instant blood-mediated inflammatory reactions, and acute cell-mediated rejection. Furthermore, this review examines immune-related strategies, including major advances, which have been shown to extend the life and/or function of porcine islets in vitro and in vivo: (1) genetic modification to make porcine islets more compatible with the recipient, (2) optimization of the newly defined biological agents that have been shown to promote long-term survival of xenografts in nonhuman primates, and (3) development of novel immunoisolation technologies that maintain the long-term survival of islet xenografts without the use of systemic immunosuppressive drugs. Finally, the clinical application of porcine islet transplantation is presented. Even though less clinical information is available, experimental data indicate that porcine islet xenografts are likely to become a standard treatment for patients with type 1 diabetes in the future.
Collapse
Affiliation(s)
- Xinyu Li
- From the Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | | |
Collapse
|
34
|
Navarro-Tableros V, Gomez Y, Brizzi MF, Camussi G. Generation of Human Stem Cell-Derived Pancreatic Organoids (POs) for Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1212:179-220. [PMID: 31025308 DOI: 10.1007/5584_2019_340] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Insulin-dependent diabetes mellitus or type 1 diabetes mellitus (T1DM) is an auto-immune condition characterized by the loss of pancreatic β-cells. The curative approach for highly selected patients is the pancreas or the pancreatic islet transplantation. Nevertheless, these options are limited by a growing shortage of donor organs and by the requirement of immunosuppression.Xenotransplantation of porcine islets has been extensively investigated. Nevertheless, the strong xenoimmunity and the risk of transmission of porcine endogenous retroviruses, have limited their application in clinic. Generation of β-like cells from stem cells is one of the most promising strategies in regenerative medicine. Embryonic, and more recently, adult stem cells are currently the most promising cell sources exploited to generate functional β-cells in vitro. A number of studies demonstrated that stem cells could generate functional pancreatic organoids (POs), able to restore normoglycemia when implanted in different preclinical diabetic models. Nevertheless, a gradual loss of function and cell dead are commonly detected when POs are transplanted in immunocompetent animals. So far, the main issue to be solved is the post-transplanted islet loss, due to the host immune attack. To avoid this hurdle, nanotechnology has provided a number of polymers currently under investigation for islet micro and macro-encapsulation. These new approaches, besides conferring PO immune protection, are able to supply oxygen and nutrients and to preserve PO morphology and long-term viability.Herein, we summarize the current knowledge on bioengineered POs and the stem cell differentiation platforms. We also discuss the in vitro strategies used to generate functional POs, and the protocols currently used to confer immune-protection against the host immune attack (micro- and macro-encapsulation). In addition, the most relevant ongoing clinical trials, and the most relevant hurdles met to move towards clinical application are revised.
Collapse
Affiliation(s)
- Victor Navarro-Tableros
- 2i3T Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico Scarl, University of Turin, Turin, Italy
| | - Yonathan Gomez
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin, Italy.
- Fondazione per la Ricerca Biomedica-ONLUS, Turin, Italy.
| |
Collapse
|
35
|
De Mesmaeker I, Robert T, Suenens KG, Stangé GM, Van Hulle F, Ling Z, Tomme P, Jacobs-Tulleneers-Thevissen D, Keymeulen B, Pipeleers DG. Increase Functional β-Cell Mass in Subcutaneous Alginate Capsules With Porcine Prenatal Islet Cells but Loss With Human Adult Islet Cells. Diabetes 2018; 67:2640-2649. [PMID: 30305364 DOI: 10.2337/db18-0709] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/25/2018] [Indexed: 11/13/2022]
Abstract
Alginate (Alg)-encapsulated porcine islet cell grafts are developed to overcome limitations of human islet transplantation. They can generate functional implants in animals when prepared from fetal, perinatal, and adult pancreases. Implants have not yet been examined for efficacy to establish sustained, metabolically adequate functional β-cell mass (FBM) in comparison with human islet cells. This study in immune-compromised mice demonstrates that subcutaneous implants of Alg-encapsulated porcine prenatal islet cells with 4 × 105 β-cells form, over 10 weeks, a FBM that results in glucose-induced plasma C-peptide >2 ng/mL and metabolic control over the following 10 weeks, with higher efficiency than nonencapsulated, while failing in peritoneum. This intracapsular FBM formation involves β-cell replication, increasing number fourfold, and maturation toward human adult β-cells. Subcutaneous Alg-encapsulated human islet cells with similar β-cell number establish implants with plasma C-peptide >2 ng/mL for the first 10 weeks, with nonencapsulated cells failing; their β-cells do not replicate but progressively die (>70%), explaining C-peptide decline and insufficient metabolic control. An Alg matrix thus helps establish β-cell functions in subcutis. It allows formation of sustained metabolically adequate FBM by immature porcine β-cells with proliferative activity but not by human adult islet cells. These findings define conditions for evaluating its immune-protecting properties.
Collapse
Affiliation(s)
- Ines De Mesmaeker
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Thomas Robert
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Krista G Suenens
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Geert M Stangé
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Freya Van Hulle
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- University Hospital Brussels-Vrije Universiteit Brussel, Brussels, Belgium
| | - Zhidong Ling
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- University Hospital Brussels-Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Daniel Jacobs-Tulleneers-Thevissen
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- University Hospital Brussels-Vrije Universiteit Brussel, Brussels, Belgium
| | - Bart Keymeulen
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- University Hospital Brussels-Vrije Universiteit Brussel, Brussels, Belgium
| | - Daniel G Pipeleers
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- University Hospital Brussels-Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Engineering endocrine pancreatic tissue is an emerging topic in type 1 diabetes with the intent to overcome the current limitation of β cell transplantation. During islet isolation, the vascularized structure and surrounding extracellular matrix (ECM) are completely disrupted. Once implanted, islets slowly engraft and mostly are lost for the initial avascular phase. This review discusses the main building blocks required to engineer the endocrine pancreas: (i) islet niche ECM, (ii) islet niche vascular network, and (iii) new available sources of endocrine cells. RECENT FINDINGS Current approaches include the following: tissue engineering of endocrine grafts by seeding of native or synthetic ECM scaffolds with human islets, vascularization of native or synthetic ECM prior to implantation, vascular functionalization of ECM structures to enhance angiogenesis after implantation, generation of engineered animals as human organ donors, and embryonic and pluripotent stem cell-derived endocrine cells that may be encapsulated or genetically engineered to be immunotolerated. Substantial technological improvements have been made to regenerate or engineer endocrine pancreatic tissue; however, significant hurdles remain, and more research is needed to develop a technology to integrate all components of viable endocrine tissue for clinical application.
Collapse
Affiliation(s)
- Antonio Citro
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Harald C Ott
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 4700, Boston, MA, 02114, USA.
- Harvard Medical School, Boston, MA, USA.
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
37
|
Perez-Basterrechea M, Esteban MM, Vega JA, Obaya AJ. Tissue-engineering approaches in pancreatic islet transplantation. Biotechnol Bioeng 2018; 115:3009-3029. [PMID: 30144310 DOI: 10.1002/bit.26821] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/08/2018] [Accepted: 08/14/2018] [Indexed: 12/15/2022]
Abstract
Pancreatic islet transplantation is a promising alternative to whole-pancreas transplantation as a treatment of type 1 diabetes mellitus. This technique has been extensively developed during the past few years, with the main purpose of minimizing the complications arising from the standard protocols used in organ transplantation. By using a variety of strategies used in tissue engineering and regenerative medicine, pancreatic islets have been successfully introduced in host patients with different outcomes in terms of islet survival and functionality, as well as the desired normoglycemic control. Here, we describe and discuss those strategies to transplant islets together with different scaffolds, in combination with various cell types and diffusible factors, and always with the aim of reducing host immune response and achieving islet survival, regardless of the site of transplantation.
Collapse
Affiliation(s)
- Marcos Perez-Basterrechea
- Unidad de Terapia Celular y Medicina Regenerativa, Servicio de Hematología y Hemoterapia, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain.,Plataforma de Terapias Avanzadas, Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Manuel M Esteban
- Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
| | - Jose A Vega
- Departamento de Morfología y Biología Celular, Universidad de Oviedo, Oviedo, Spain.,Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Alvaro J Obaya
- Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
38
|
Abstract
β cell replacement with either pancreas or islet transplantation has progressed immensely over the last decades with current 1- and 5-year insulin independence rates of approximately 85% and 50%, respectively. Recent advances are largely attributed to improvements in immunosuppressive regimen, donor selection, and surgical technique. However, both strategies are compromised by a scarce donor source. Xenotransplantation offers a potential solution by providing a theoretically unlimited supply of islets, but clinical application has been limited by concerns for a potent immune response against xenogeneic tissue. β cell clusters derived from embryonic or induced pluripotent stem cells represent another promising unlimited source of insulin producing cells, but clinical application is pending further advances in the function of the β cell like clusters. Exciting developments and rapid progress in all areas of β cell replacement prompted a lively debate by members of the young investigator committee of the International Pancreas and Islet Transplant Association at the 15th International Pancreas and Islet Transplant Association Congress in Melbourne and at the 26th international congress of The Transplant Society in Hong Kong. This international group of young investigators debated which modality of β cell replacement would predominate the landscape in 10 years, and their arguments are summarized here.
Collapse
|
39
|
Safley SA, Kenyon NS, Berman DM, Barber GF, Willman M, Duncanson S, Iwakoshi N, Holdcraft R, Gazda L, Thompson P, Badell IR, Sambanis A, Ricordi C, Weber CJ. Microencapsulated adult porcine islets transplanted intraperitoneally in streptozotocin-diabetic non-human primates. Xenotransplantation 2018; 25:e12450. [PMID: 30117193 DOI: 10.1111/xen.12450] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 05/18/2018] [Accepted: 06/26/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Xenogeneic donors would provide an unlimited source of islets for the treatment of type 1 diabetes (T1D). The goal of this study was to assess the function of microencapsulated adult porcine islets (APIs) transplanted ip in streptozotocin (STZ)-diabetic non-human primates (NHPs) given targeted immunosuppression. METHODS APIs were encapsulated in: (a) single barium-gelled alginate capsules or (b) double alginate capsules with an inner, islet-containing compartment and a durable, biocompatible outer alginate layer. Immunosuppressed, streptozotocin-diabetic NHPs were transplanted ip with encapsulated APIs, and graft function was monitored by measuring blood glucose, %HbA1c, and porcine C-peptide. At graft failure, explanted capsules were assessed for biocompatibility and durability plus islet viability and functionality. Host immune responses were evaluated by phenotyping peritoneal cell populations, quantitation of peritoneal cytokines and chemokines, and measurement of anti-porcine IgG and IgM plus anti-Gal IgG. RESULTS NHP recipients had reduced hyperglycemia, decreased exogenous insulin requirements, and lower percent hemoglobin A1c (%HbA1c) levels. Porcine C-peptide was detected in plasma of all recipients, but these levels diminished with time. However, relatively high levels of porcine C-peptide were detected locally in the peritoneal graft site of some recipients at sacrifice. IV glucose tolerance tests demonstrated metabolic function, but the grafts eventually failed in all diabetic NHPs regardless of the type of encapsulation or the host immunosuppression regimen. Explanted microcapsules were intact, "clean," and free-floating without evidence of fibrosis at graft failure, and some reversed diabetes when re-implanted ip in diabetic immunoincompetent mice. Histology of explanted capsules showed scant evidence of a host cellular response, and viable islets could be found. Flow cytometric analyses of peritoneal cells and peripheral blood showed similarly minimal evidence of a host immune response. Preformed anti-porcine IgG and IgM antibodies were present in recipient plasma, but these levels did not rise post-transplant. Peritoneal graft site cytokine or chemokine levels were equivalent to normal controls, with the exception of minimal elevation observed for IL-6 or IL-1β, GRO-α, I-309, IP-10, and MCP-1. However, we found central necrosis in many of the encapsulated islets after graft failure, and explanted islets expressed endogenous markers of hypoxia (HIF-1α, osteopontin, and GLUT-1), suggesting a role for non-immunologic factors, likely hypoxia, in graft failure. CONCLUSIONS With donor xenoislet microencapsulation and host immunosuppression, APIs corrected hyperglycemia after ip transplantation in STZ-diabetic NHPs in the short term. The islet xenografts lost efficacy gradually, but at graft failure, some viable islets remained, substantial porcine C-peptide was detected in the peritoneal graft site, and there was very little evidence of a host immune response. We postulate that chronic effects of non-immunologic factors, such as in vivo hypoxic and hyperglycemic conditions, damaged the encapsulated islet xenografts. To achieve long-term function, new approaches must be developed to prevent this damage, for example, by increasing the oxygen supply to microencapsulated islets in the ip space.
Collapse
Affiliation(s)
- Susan A Safley
- Department of Surgery, Emory University, Atlanta, Georgia
| | - Norma S Kenyon
- Diabetes Research Institute, Miami, Florida.,Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida.,Department of Microbiology & Immunology, Miller School of Medicine, University of Miami, Miami, Florida.,Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida
| | - Dora M Berman
- Diabetes Research Institute, Miami, Florida.,Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida.,Department of Microbiology & Immunology, Miller School of Medicine, University of Miami, Miami, Florida.,Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida
| | | | | | - Stephanie Duncanson
- Department of Biomedical Engineering, School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Neal Iwakoshi
- Department of Surgery, Emory University, Atlanta, Georgia
| | | | | | - Peter Thompson
- Department of Surgery, Emory University, Atlanta, Georgia
| | - I Raul Badell
- Department of Surgery, Emory University, Atlanta, Georgia
| | - Athanassios Sambanis
- Department of Biomedical Engineering, School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia
| | - Camillo Ricordi
- Diabetes Research Institute, Miami, Florida.,Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida.,Department of Microbiology & Immunology, Miller School of Medicine, University of Miami, Miami, Florida.,Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida.,Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
| | - Collin J Weber
- Department of Surgery, Emory University, Atlanta, Georgia
| |
Collapse
|
40
|
Smith KE, Purvis WG, Davis MA, Min CG, Cooksey AM, Weber CS, Jandova J, Price ND, Molano DS, Stanton JB, Kelly AC, Steyn LV, Lynch RM, Limesand SW, Alexander M, Lakey JRT, Seeberger K, Korbutt GS, Mueller KR, Hering BJ, McCarthy FM, Papas KK. In vitro characterization of neonatal, juvenile, and adult porcine islet oxygen demand, β-cell function, and transcriptomes. Xenotransplantation 2018; 25:e12432. [PMID: 30052287 DOI: 10.1111/xen.12432] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/20/2018] [Accepted: 05/24/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND There is currently a shortage of human donor pancreata which limits the broad application of islet transplantation as a treatment for type 1 diabetes. Porcine islets have demonstrated potential as an alternative source, but a study evaluating islets from different donor ages under unified protocols has yet to be conducted. METHODS Neonatal porcine islets (NPI; 1-3 days), juvenile porcine islets (JPI; 18-21 days), and adult porcine islets (API; 2+ years) were compared in vitro, including assessments of oxygen consumption rate, membrane integrity determined by FDA/PI staining, β-cell proliferation, dynamic glucose-stimulated insulin secretion, and RNA sequencing. RESULTS Oxygen consumption rate normalized to DNA was not significantly different between ages. Membrane integrity was age dependent, and API had the highest percentage of intact cells. API also had the highest glucose-stimulated insulin secretion response during a dynamic insulin secretion assay and had 50-fold higher total insulin content compared to NPI and JPI. NPI and JPI had similar glucose responsiveness, β-cell percentage, and β-cell proliferation rate. Transcriptome analysis was consistent with physiological assessments. API transcriptomes were enriched for cellular metabolic and insulin secretory pathways, while NPI exhibited higher expression of genes associated with proliferation. CONCLUSIONS The oxygen demand, membrane integrity, β-cell function and proliferation, and transcriptomes of islets from API, JPI, and NPI provide a comprehensive physiological comparison for future studies. These assessments will inform the optimal application of each age of porcine islet to expand the availability of islet transplantation.
Collapse
Affiliation(s)
- Kate E Smith
- Department of Physiological Sciences, University of Arizona, Tucson, AZ, USA.,Department of Surgery, University of Arizona, Tucson, AZ, USA
| | | | - Melissa A Davis
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Catherine G Min
- Department of Physiological Sciences, University of Arizona, Tucson, AZ, USA.,Department of Surgery, University of Arizona, Tucson, AZ, USA
| | - Amanda M Cooksey
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Craig S Weber
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | - Jana Jandova
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | | | - Diana S Molano
- Department of Surgery, University of Arizona, Tucson, AZ, USA
| | | | - Amy C Kelly
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Leah V Steyn
- Department of Surgery, University of Arizona, Tucson, AZ, USA
| | - Ronald M Lynch
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | - Sean W Limesand
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Michael Alexander
- Department of Surgery, University of California-Irvine, Orange, CA, USA
| | | | - Karen Seeberger
- Department of Surgery, Alberta Diabetes Institute, University of Alberta, Edmonton, AL, Canada
| | - Gregory S Korbutt
- Department of Surgery, Alberta Diabetes Institute, University of Alberta, Edmonton, AL, Canada
| | - Kate R Mueller
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Bernhard J Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Fiona M McCarthy
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | | |
Collapse
|
41
|
Abstract
The pancreas is made from two distinct components: the exocrine pancreas, a reservoir of digestive enzymes, and the endocrine islets, the source of the vital metabolic hormone insulin. Human islets possess limited regenerative ability; loss of islet β-cells in diseases such as type 1 diabetes requires therapeutic intervention. The leading strategy for restoration of β-cell mass is through the generation and transplantation of new β-cells derived from human pluripotent stem cells. Other approaches include stimulating endogenous β-cell proliferation, reprogramming non-β-cells to β-like cells, and harvesting islets from genetically engineered animals. Together these approaches form a rich pipeline of therapeutic development for pancreatic regeneration.
Collapse
|
42
|
Moroni L, Burdick JA, Highley C, Lee SJ, Morimoto Y, Takeuchi S, Yoo JJ. Biofabrication strategies for 3D in vitro models and regenerative medicine. NATURE REVIEWS. MATERIALS 2018; 3:21-37. [PMID: 31223488 PMCID: PMC6586020 DOI: 10.1038/s41578-018-0006-y] [Citation(s) in RCA: 384] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Organs are complex systems composed of different cells, proteins and signalling molecules that are arranged in a highly ordered structure to orchestrate a myriad of functions in our body. Biofabrication strategies can be applied to engineer 3D tissue models in vitro by mimicking the structure and function of native tissue through the precise deposition and assembly of materials and cells. This approach allows the spatiotemporal control over cell-cell and cell-extracellular matrix communication and thus the recreation of tissue-like structures. In this Review, we examine biofabrication strategies for the construction of functional tissue replacements and organ models, focusing on the development of biomaterials, such as supramolecular and photosensitive materials, that can be processed using biofabrication techniques. We highlight bioprinted and bioassembled tissue models and survey biofabrication techniques for their potential to recreate complex tissue properties, such as shape, vasculature and specific functionalities. Finally, we discuss challenges, such as scalability and the foreign body response, and opportunities in the field and provide an outlook to the future of biofabrication in regenerative medicine.
Collapse
Affiliation(s)
- Lorenzo Moroni
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Complex Tissue Regeneration, Maastricht University, Maastricht, Netherlands
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher Highley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yuya Morimoto
- Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| | - Shoji Takeuchi
- Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| | - James J. Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
43
|
Orive G, Emerich D, Khademhosseini A, Matsumoto S, Hernández RM, Pedraz JL, Desai T, Calafiore R, de Vos P. Engineering a Clinically Translatable Bioartificial Pancreas to Treat Type I Diabetes. Trends Biotechnol 2018; 36:445-456. [PMID: 29455936 DOI: 10.1016/j.tibtech.2018.01.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/11/2018] [Accepted: 01/16/2018] [Indexed: 02/06/2023]
Abstract
Encapsulating, or immunoisolating, insulin-secreting cells within implantable, semipermeable membranes is an emerging treatment for type 1 diabetes. This approach can eliminate the need for immunosuppressive drug treatments to prevent transplant rejection and overcome the shortage of donor tissues by utilizing cells derived from allogeneic or xenogeneic sources. Encapsulation device designs are being optimized alongside the development of clinically viable, replenishable, insulin-producing stem cells, for the first time creating the possibility of widespread therapeutic use of this technology. Here, we highlight the status of the most advanced and widely explored implementations of cell encapsulation with an eye toward translating the potential of this technological approach to medical reality.
Collapse
Affiliation(s)
- Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain; BTI Biotechnology Institute, Vitoria, Spain; Joint first authors and contributed equally to the paper.
| | - Dwaine Emerich
- NsGene,225 Chapman Street, Providence, RI, USA; Joint first authors and contributed equally to the paper
| | - Ali Khademhosseini
- Department of Bioengineering, Department of Chemical and Biomolecular Engineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, USA; Department of Radiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA; Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, USA; California NanoSystems Institute (CNSI), University of California-Los Angeles, Los Angeles, CA, USA. http://twitter.com/@khademh
| | - Shinichi Matsumoto
- Research and Development Center, Otsuka Pharmaceutical Factory, 115 Kuguhara, Tateiwa, Muya-cho, Naruto, Tokushima 772-8601, Japan
| | - R M Hernández
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - J L Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - Tejal Desai
- University of California, San Francisco, Department of Bioengineering and Therapeutic Sciences, Byers Hall Room 203C, MC 2520, 1700 4th Street, San Francisco, CA, USA
| | - Riccardo Calafiore
- Department of Medicine, Section of Cardiovascular, Endocrine and Metabolic Clinical Physiology, Laboratory for Endocrine Cell Transplants and Biohybrid Organs, University of Perugia, Piazzale Gambuli, Perugia, Italy; Joint first authors and contributed equally to the paper
| | - Paul de Vos
- University of Groningen, Pathology and Medical Biology Section, Immunoendocrinology, Groningen, The Netherlands.
| |
Collapse
|
44
|
Richardson TC, Mathew S, Candiello JE, Goh SK, Kumta PN, Banerjee I. Development of an Alginate Array Platform to Decouple the Effect of Multiparametric Perturbations on Human Pluripotent Stem Cells During Pancreatic Differentiation. Biotechnol J 2018; 13. [DOI: 10.1002/biot.201700099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 12/09/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Thomas C. Richardson
- Department of Chemical and Petroleum Engineering; University of Pittsburgh; Pittsburgh USA
| | - Shibin Mathew
- Department of Chemical and Petroleum Engineering; University of Pittsburgh; Pittsburgh USA
| | | | - Saik K. Goh
- Department of Bioengineering; University of Pittsburgh; Pittsburgh USA
| | - Prashant N. Kumta
- Department of Bioengineering, Chemical and Petroleum Engineering; Mechanical Engineering and Materials Science; University of Pittsburgh; Pittsburgh USA
| | - Ipsita Banerjee
- Department of Chemical and Petroleum Engineering; University of Pittsburgh; Pittsburgh USA
| |
Collapse
|
45
|
Zhu H, Li W, Liu Z, Li W, Chen N, Lu L, Zhang W, Wang Z, Wang B, Pan K, Zhang X, Chen G. Selection of Implantation Sites for Transplantation of Encapsulated Pancreatic Islets. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:191-214. [PMID: 29048258 DOI: 10.1089/ten.teb.2017.0311] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pancreatic islet transplantation has been validated as a valuable therapy for type 1 diabetes mellitus patients with exhausted insulin treatment. However, this therapy remains limited by the shortage of donor and the requirement of lifelong immunosuppression. Islet encapsulation, as an available bioartificial pancreas (BAP), represents a promising approach to enable protecting islet grafts without or with minimal immunosuppression and possibly expanding the donor pool. To develop a clinically implantable BAP, some key aspects need to be taken into account: encapsulation material, capsule design, and implant site. Among them, the implant site exerts an important influence on the engraftment, stability, and biocompatibility of implanted BAP. Currently, an optimal site for encapsulated islet transplantation may include sufficient capacity to host large graft volumes, portal drainage, ease of access using safe and reproducible procedure, adequate blood/oxygen supply, minimal immune/inflammatory reaction, pliable for noninvasive imaging and biopsy, and potential of local microenvironment manipulation or bioengineering. Varying degrees of success have been confirmed with the utilization of liver or extrahepatic sites in an experimental or preclinical setting. However, the ideal implant site remains to be further engineered or selected for the widespread application of encapsulated islet transplantation.
Collapse
Affiliation(s)
- Haitao Zhu
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China .,2 Department of Hepatobiliary Surgery, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University , Xi'an, China
| | - Wenjing Li
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Zhongwei Liu
- 3 Department of Cardiology, Shaanxi Provincial People's Hospital , Xi'an, China
| | - Wenliang Li
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Niuniu Chen
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Linlin Lu
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Wei Zhang
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Zhen Wang
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Bo Wang
- 2 Department of Hepatobiliary Surgery, the First Affiliated Hospital, Medical School of Xi'an Jiaotong University , Xi'an, China .,4 Institute of Advanced Surgical Technology and Engineering, Xi'an Jiaotong University , Xi'an, China
| | - Kaili Pan
- 5 Department of Pediatrics (No. 2 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Xiaoge Zhang
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| | - Guoqiang Chen
- 1 Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital , Xi'an, China
| |
Collapse
|
46
|
Lee SJ, Lee JB, Park YW, Lee DY. 3D Bioprinting for Artificial Pancreas Organ. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:355-374. [PMID: 30471043 DOI: 10.1007/978-981-13-0445-3_21] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Type 1 diabetes mellitus (T1DM) results from an autoimmune destruction of insulin-producing beta cells in the islet of the endocrine pancreas. Although islet transplantation has been regarded as an ideal strategy for T1D, transplanted islets are rejected from host immune system. To immunologically protect them, islet encapsulation technology with biocompatible materials is emerged as an immuno-barrier. However, this technology has been limited for clinical trial such as hypoxia in the central core of islet bead, impurity of islet bead and retrievability from the body. Recently, 3D bioprinting has been emerged as an alternative approach to make the artificial pancreas. It can be used to position live cells in a desired location with real scale of human organ. Furthermore, constructing a vascularization of the artificial pancreas is actualized with 3D bioprinting. Therefore, it is possible to create real pancreas-mimic artificial organ for clinical application. In conclusion, 3D bioprinting can become a new leader in the development of the artificial pancreas to overcome the existed islet.
Collapse
Affiliation(s)
- Seon Jae Lee
- Department of Bioengineering, College of Engineering, BK21 PLUS Future Biopharmaceutical Human Resource Training and Research Team, Hanyang University, Seoul, South Korea
| | - Jae Bin Lee
- Department of Bioengineering, College of Engineering, BK21 PLUS Future Biopharmaceutical Human Resource Training and Research Team, Hanyang University, Seoul, South Korea
| | - Young-Woo Park
- Department of Bioengineering, College of Engineering, BK21 PLUS Future Biopharmaceutical Human Resource Training and Research Team, Hanyang University, Seoul, South Korea
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, BK21 PLUS Future Biopharmaceutical Human Resource Training and Research Team, Hanyang University, Seoul, South Korea. .,Institute of Nano Science & Technology (INST), Hanyang University, Seoul, South Korea.
| |
Collapse
|
47
|
Abstract
The principle of immunoisolation of cells is based on encapsulation of cells in immunoprotective but semipermeable membranes that protect cells from hazardous effects of the host immune system but allows ingress of nutrients and outgress of therapeutic molecules. The technology was introduced in 1933 but has only received its deserved attention for its therapeutic application for three decades now.In the past decade important advances have been made in creating capsules that provoke minimal or no inflammatory responses. There are however new emerging challenges. These challenges relate to optimal nutrition and oxygen supply as well as standardization and documentation of capsule properties.It is concluded that the proof of principle of applicability of encapsulated grafts for treatment of human disease has been demonstrated and merits optimism about its clinical potential. Further innovation requires a much more systematic approach in identifying crucial properties of capsules and cellular grafts to allow sound interpretations of the results.
Collapse
Affiliation(s)
- Paul de Vos
- Division of Immuno-Endocrinology, Departments of Pathology and Laboratory Medicine, University of Groningen, Groningen, Groningen, The Netherlands.
| |
Collapse
|
48
|
Krishnan R, Ko D, Foster CE, Liu W, Smink AM, de Haan B, De Vos P, Lakey JRT. Immunological Challenges Facing Translation of Alginate Encapsulated Porcine Islet Xenotransplantation to Human Clinical Trials. Methods Mol Biol 2017; 1479:305-333. [PMID: 27738946 DOI: 10.1007/978-1-4939-6364-5_24] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Transplantation of alginate-encapsulated islets has the potential to treat patients suffering from type I diabetes, a condition characterized by an autoimmune attack against insulin-secreting beta cells. However, there are multiple immunological challenges associated with this procedure, all of which must be adequately addressed prior to translation from trials in small animal and nonhuman primate models to human clinical trials. Principal threats to graft viability include immune-mediated destruction triggered by immunogenic alginate impurities, unfavorable polymer composition and surface characteristics, and release of membrane-permeable antigens, as well as damage associated molecular patterns (DAMPs) by the encapsulated islets themselves. The lack of standardization of significant parameters of bioencapsulation device design and manufacture (i.e., purification protocols, surface-modification grafting techniques, alginate composition modifications) between labs is yet another obstacle that must be overcome before a clinically effective and applicable protocol for encapsulating islets can be implemented. Nonetheless, substantial progress is being made, as is evident from prolonged graft survival times and improved protection from immune-mediated graft destruction reported by various research groups, but also with regard to discoveries of specific pathways involved in explaining observed outcomes. Progress in the latter is essential for a comprehensive understanding of the mechanisms responsible for the varying levels of immunogenicity of certain alginate devices. Successful translation of encapsulated islet transplantation from in vitro and animal model testing to human clinical trials hinges on application of this knowledge of the pathways and interactions which comprise immune-mediated rejection. Thus, this review not only focuses on the different factors contributing to provocation of the immune reaction by encapsulated islets, but also on the defining characteristics of the response itself.
Collapse
Affiliation(s)
- Rahul Krishnan
- Department of Surgery, University of California Irvine, 333 City Blvd West, Suite 1600, Orange, CA, 92868, USA
| | - David Ko
- Department of Surgery, University of California Irvine, 333 City Blvd West, Suite 1600, Orange, CA, 92868, USA
| | - Clarence E Foster
- Department of Surgery, University of California Irvine, 333 City Blvd West, Suite 1600, Orange, CA, 92868, USA.,Department of Transplantation, University of California Irvine, Orange, CA, USA
| | - Wendy Liu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - A M Smink
- Division of Immuno-Endocrinology, Departments of Pathology and Laboratory Medicine, University of Groningen, Groningen, The Netherlands
| | - Bart de Haan
- Division of Immuno-Endocrinology, Departments of Pathology and Laboratory Medicine, University of Groningen, Groningen, The Netherlands
| | - Paul De Vos
- Division of Immuno-Endocrinology, Departments of Pathology and Laboratory Medicine, University of Groningen, Groningen, The Netherlands
| | - Jonathan R T Lakey
- Department of Surgery, University of California Irvine, 333 City Blvd West, Suite 1600, Orange, CA, 92868, USA. .,Department of Transplantation, University of California Irvine, Orange, CA, USA. .,Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
49
|
|
50
|
Abstract
The pancreas is a complex organ with exocrine and endocrine components. Many pathologies impair exocrine function, including chronic pancreatitis, cystic fibrosis and pancreatic ductal adenocarcinoma. Conversely, when the endocrine pancreas fails to secrete sufficient insulin, patients develop diabetes mellitus. Pathology in either the endocrine or exocrine pancreas results in devastating economic and personal consequences. The current standard therapy for treating patients with type 1 diabetes mellitus is daily exogenous insulin injections, but cell sources of insulin provide superior glycaemic regulation and research is now focused on the goal of regenerating or replacing β cells. Stem-cell-based models might be useful to study exocrine pancreatic disorders, and mesenchymal stem cells or secreted factors might delay disease progression. Although the standards that bioengineered cells must meet before being considered as a viable therapy are not yet established, any potential therapy must be acceptably safe and functionally superior to current therapies. Here, we describe progress and challenges in cell-based methods to restore pancreatic function, with a focus on optimizing the site for cell delivery and decreasing requirements for immunosuppression through encapsulation. We also discuss the tools and strategies being used to generate exocrine pancreas and insulin-producing β-cell surrogates in situ and highlight obstacles to clinical application.
Collapse
|