Insulin expression and C-peptide in type 1 diabetes subjects implanted with stem cell-derived pancreatic endoderm cells in an encapsulation device

A M James Shapiro, David Thompson, Thomas W Donner, Melena D Bellin, Willa Hsueh, Jeremy Pettus, Jon Wilensky, Mark Daniels, Richard M Wang, Eugene P Brandon, Manasi S Jaiman, Evert J Kroon, Kevin A D'Amour, Howard L Foyt, A M James Shapiro, David Thompson, Thomas W Donner, Melena D Bellin, Willa Hsueh, Jeremy Pettus, Jon Wilensky, Mark Daniels, Richard M Wang, Eugene P Brandon, Manasi S Jaiman, Evert J Kroon, Kevin A D'Amour, Howard L Foyt

Abstract

These preliminary data from an ongoing first-in-human phase 1/2, open-label study provide proof-of-concept that pluripotent stem cell-derived pancreatic endoderm cells (PEC-01) engrafted in type 1 diabetes patients become islet cells releasing insulin in a physiologically regulated fashion. In this study of 17 subjects aged 22-57 with type 1 diabetes, PEC-01 cells were implanted subcutaneously in VC-02 macroencapsulation devices, allowing for direct vascularization of the cells. Engraftment and insulin expression were observed in 63% of VC-02 units explanted from subjects at 3-12 months post-implant. Six of 17 subjects (35.3%) demonstrated positive C-peptide as early as 6 months post-implant. Most reported adverse events were related to surgical implant or explant procedures (27.9%) or to side-effects of immunosuppression (33.7%). Initial data suggest that pluripotent stem cells, which can be propagated to the desired biomass and differentiated into pancreatic islet-like tissue, may offer a scalable, renewable alternative to pancreatic islet transplants.

Trial registration: ClinicalTrials.gov NCT03163511.

Conflict of interest statement

M.D., R.M.W., E.J.K., E.P.B., K.A.D., M.J., and H.L.F. are employees of ViaCyte and may have equity interest in ViaCyte, Inc., a privately held company. J.S. and J.W. are consultants for ViaCyte and may have equity interest in ViaCyte. ViaCyte has 850 global and US patents. The remaining authors declare no conflicts of interest. Funding sources for this work included the California Institute for Regenerative Medicine, JDRF, and the Stem Cell Network of Canada. These groups were not involved in the research, the writing, or the decision to submit the manuscript for publication. Authors have not been paid by a pharmaceutical company or other organization to write this article.

© 2021 The Authors.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
The two different types of units used in the present study (Upper photo) Sentinels (VC-02-20), which are smaller in size (1.5 cm x 1 cm) and have fewer cells, are explanted and examined periodically to evaluate status of the dose-finding units. For size comparison a US quarter is shown. (Lower photo) The larger dose-finding units (VC-02-300, 9 cm x 3 cm)) are used to develop the procedures and to assess the safety and efficacy of various quanta of cell doses.
Figure 2
Figure 2
Trial Design of Cohort 2
Figure 3
Figure 3
Histology of cross-sections of a sentinel explanted from “Subject D-001” with glucose-responsive C-peptide illustrates both the cellular and non-cellular regions of a mature graft (A–C) Three serial cross-sections (B) of approximately 4 mm apart within the explant stained with hematoxylin and eosin (H&E) reveal cellular regions (purple, hematoxylin-stained, enlarged in C) and non-cellular regions (pink, eosin-stained, enlarged in A). The non-cellular regions consist of collagenous fibers corresponding to regions of infiltrating host myofibroblasts observed at earlier time points (see Figure S2). (D–G) Panels (D)–(G) show immunohistochemistry of the cellular region identified in (C). Chromogranin A-staining shows the vast majority of the graft consists of endocrine cells (D). Staining for alpha smooth muscle actin (αSMA) identifies host-derived myofibroblasts (E), especially in the bottom left within the device lumen, and in larger blood vessels, but not or only weakly so smaller capillaries. Glucagon (brown) and insulin (magenta) staining account for the majority of graft-derived endocrine cells, glucagon being more abundant (F). Staining for CD34 identifies small capillaries as well as larger blood vessels (G). Note paucity of CD34 staining in the region devoid of graft-derived cells (bottom left within device lumen). Counterstain in all IHC panels is hematoxylin. Scale bars, 2.5 mm (top center) and 250 μm (all other images).
Figure 4
Figure 4
Histology of cross-sections from an explanted dose-finding unit at approximately 11 months from “Subject E-002” Several panels illustrate the histological features routinely observed in the cellular regions of VC-02 grafts. Shown are “nearly adjacent” sections. Gray strips seen at the top and bottom of each image are the delivery device membranes. (A) H&E staining reveals densely packed cells reminiscent of human endocrine islet morphologies, albeit without the macro-anatomical “islet” nature. (B) Nearly all graft-derived cells are endodermal in nature as demonstrated by nuclear staining to the transcription factors FOXA2 and CDX2. The open structure lined with endodermal cells, exemplified in the lower left of each image, is not a prominent feature but observed occasionally in the cellular regions of the grafts. (C) A subset of graft-derived cells coincident with insulin-expressing cells show nuclear immunoreactivity for NKX6-1, a transcription factor specific to β cells in mature human islets. (D) The cellular regions are well vascularized with microcapillary structure, demonstrated with the endothelial cell marker CD34 (magenta). CD3 staining (brown) marks T cells, not abundant but observed occasionally outside the device lumen, but rarely inside the lumen among graft-derived cells. (E) Many cells show glucagon (GCG) immunoreactivity. (F) Many cells show insulin (INS) immunoreactivity in regions coincident with NKX6-1 expression. Scale bars, 250 μm. Counterstain is hematoxylin.

References

    1. Shapiro A.M., Lakey J.R., Ryan E.A., Korbutt G.S., Toth E., Warnock G.L., Kneteman N.M., Rajotte R.V. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N. Engl. J. Med. 2000;343:230–238.
    1. Hering B.J., Clarke W.R., Bridges N.D., Eggerman T.L., Alejandro R., Bellin M.D., Chaloner K., Czarniecki C.W., Goldstein J.S., Hunsicker L.G., et al. Clinical Islet Transplantation Consortium Phase 3 Trial of Transplantation of Human Islets in Type 1 Diabetes Complicated by Severe Hypoglycemia. Diabetes Care. 2016;39:1230–1240.
    1. Lablanche S., Vantyghem M.C., Kessler L., Wojtusciszyn A., Borot S., Thivolet C., Girerd S., Bosco D., Bosson J.L., Colin C., et al. TRIMECO trial investigators Islet transplantation versus insulin therapy in patients with type 1 diabetes with severe hypoglycaemia or poorly controlled glycaemia after kidney transplantation (TRIMECO): a multicentre, randomised controlled trial. Lancet Diabetes Endocrinol. 2018;6:527–537.
    1. Thomson J.A., Itskovitz-Eldor J., Shapiro S.S., Waknitz M.A., Swiergiel J.J., Marshall V.S., Jones J.M. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282:1145–1147.
    1. D’Amour K.A., Agulnick A.D., Eliazer S., Kelly O.G., Kroon E., Baetge E.E. Efficient differentiation of human embryonic stem cells to definitive endoderm. Nat. Biotechnol. 2005;23:1534–1541.
    1. D’Amour K.A., Bang A.G., Eliazer S., Kelly O.G., Agulnick A.D., Smart N.G., Moorman M.A., Kroon E., Carpenter M.K., Baetge E.E. Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat. Biotechnol. 2006;24:1392–1401.
    1. Schulz T.C., Young H.Y., Agulnick A.D., Babin M.J., Baetge E.E., Bang A.G., Bhoumik A., Cepa I., Cesario R.M., Haakmeester C., et al. A scalable system for production of functional pancreatic progenitors from human embryonic stem cells. PLoS ONE. 2012;7:e37004.
    1. Schulz T.C. Concise Review: Manufacturing of Pancreatic Endoderm Cells for Clinical Trials in Type 1 Diabetes. Stem Cells Transl. Med. 2015;4:927–931.
    1. Brandon E., Scott M., Zimmerman M., D’Amour K. In: Second Generation Cell and Gene-based Therapies: Biological Advances, Clinical Outcomes, and Strategies for Capitalisation. Vertès Alain A., Smith Devyn M., Qureshi Nasib, Dowden Nathan J., editors. Elsevier; 2020. Stem cell-derived islet replacement: Which cells & how to protect them from the immune system?
    1. Kroon E., Martinson L.A., Kadoya K., Bang A.G., Kelly O.G., Eliazer S., Young H., Richardson M., Smart N.G., Cunningham J., et al. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat. Biotechnol. 2008;26:443–452.
    1. Kelly O.G., Chan M.Y., Martinson L.A., Kadoya K., Ostertag T.M., Ross K.G., Richardson M., Carpenter M.K., D’Amour K.A., Kroon E., et al. Cell-surface markers for the isolation of pancreatic cell types derived from human embryonic stem cells. Nat. Biotechnol. 2011;29:750–756.
    1. Rezania A., Bruin J.E., Riedel M.J., Mojibian M., Asadi A., Xu J., Gauvin R., Narayan K., Karanu F., O’Neil J.J., et al. Maturation of human embryonic stem cell-derived pancreatic progenitors into functional islets capable of treating pre-existing diabetes in mice. Diabetes. 2012;61:2016–2029.
    1. Motté E., Szepessy E., Suenens K., Stangé G., Bomans M., Jacobs-Tulleneers-Thevissen D., Ling Z., Kroon E., Pipeleers D., Beta Cell Therapy Consortium EU-FP7 Composition and function of macroencapsulated human embryonic stem cell-derived implants: comparison with clinical human islet cell grafts. Am. J. Physiol. Endocrinol. Metab. 2014;307:E838–E846.
    1. Agulnick A.D., Ambruzs D.M., Moorman M.A., Bhoumik A., Cesario R.M., Payne J.K., Kelly J.R., Haakmeester C., Srijemac R., Wilson A.Z., et al. Insulin-Producing Endocrine Cells Differentiated In Vitro From Human Embryonic Stem Cells Function in Macroencapsulation Devices In Vivo. Stem Cells Transl. Med. 2015;4:1214–1222.
    1. Bruin J.E., Rezania A., Xu J., Narayan K., Fox J.K., O’Neil J.J., Kieffer T.J. Maturation and function of human embryonic stem cell-derived pancreatic progenitors in macroencapsulation devices following transplant into mice. Diabetologia. 2013;56:1987–1998.
    1. Haller C., Piccand J., De Franceschi F., Ohi Y., Bhoumik A., Boss C., De Marchi U., Jacot G., Metairon S., Descombes P., et al. Macroencapsulated Human iPSC-Derived Pancreatic Progenitors Protect against STZ-Induced Hyperglycemia in Mice. Stem Cell Reports. 2019;12:787–800.
    1. Ramzy A., Thompson D.M., Ward-Hartsonge K.A., Ivison S., Cook L., Garcia R.V., Loyal J., Kim P.T.W., Warnock G.L., Levings M.K., Kieffer T.J. Implanted pluripotent stem cell-derived pancreatic endoderm cells secrete glucose-responsive C-peptide in subjects with type 1 diabetes. Cell Stem Cell. 2021;28:2047–2061. doi: 10.1016/j.stem.2021.10.003.
    1. Pusztaszeri M.P., Seelentag W., Bosman F.T. Immunohistochemical expression of endothelial markers CD31, CD34, von Willebrand factor, and Fli-1 in normal human tissues. J. Histochem. Cytochem. 2006;54:385–395.
    1. Keymeulen B., Jacobs-Tulleneers-Thevissen D., Sr., Kroon E.J., Jaiman M.S., Daniels M., Wang R., Pipeleers D., D’Amour K., Foyt H.L. 196-LB: Stem Cell–Derived Islet Replacement Therapy (VC-02) Demonstrates Production of C-Peptide in Patients with Type 1 Diabetes (T1D) and Hypoglycemia Unawareness. Diabetes. 2021 doi: 10.2337/db21-196-LB.
    1. Henry R.R., Pettus J.H., Wilensky J., Shapiro A.M., Senior P.A., Roep B.O., Wang R.S., Kroon E.J., Scott M.J., D’Amour K.A., Foyt H.L. Initial Clinical Evaluation of VC-01TM Combination Product: A Stem Cell-Derived Islet Replacement for Type 1 Diabetes (T1D) Diabetes. 2018 doi: 10.2337/db18-138-OR.

Source: PubMed

3
Předplatit