Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Combinatorial hydrogel library enables identification of materials that mitigate the foreign body response in primates

A Corrigendum to this article was published on 09 June 2016

This article has been updated

Abstract

The foreign body response is an immune-mediated reaction that can lead to the failure of implanted medical devices and discomfort for the recipient1,2,3,4,5,6. There is a critical need for biomaterials that overcome this key challenge in the development of medical devices. Here we use a combinatorial approach for covalent chemical modification to generate a large library of variants of one of the most widely used hydrogel biomaterials, alginate. We evaluated the materials in vivo and identified three triazole-containing analogs that substantially reduce foreign body reactions in both rodents and, for at least 6 months, in non-human primates. The distribution of the triazole modification creates a unique hydrogel surface that inhibits recognition by macrophages and fibrous deposition. In addition to the utility of the compounds reported here, our approach may enable the discovery of other materials that mitigate the foreign body response.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Combinatorially modified hydrogels with reduced subcutaneous inflammation and fibrosis.
Figure 2: Three lead hydrogels show reduced fibrosis intraperitoneally in C57BL/6J mice.
Figure 3: Lead hydrogels show low immune cell recruitment in vivo with covalently modified surfaces.
Figure 4: Lead hydrogels mitigate the foreign body response in non-human primates.

Similar content being viewed by others

Change history

  • 18 April 2016

    In the version of this article initially published, one author, Adam C. Graham, his affiliation, and his contribution were omitted. In addition, two acknowledgments, to W. Salmon and J. Wyckoff, were omitted. The errors have been corrected in the HTML and PDF versions of the article.

References

  1. Anderson, J.M., Rodriguez, A. & Chang, D.T. Foreign body reaction to biomaterials. Semin. Immunol. 20, 86–100 (2008).

    Article  CAS  PubMed  Google Scholar 

  2. Langer, R. Perspectives and challenges in tissue engineering and regenerative medicine. Adv. Mater. 21, 3235–3236 (2009).

    Article  CAS  PubMed  Google Scholar 

  3. Ward, W.K. A review of the foreign-body response to subcutaneously-implanted devices: the role of macrophages and cytokines in biofouling and fibrosis. J. Diabetes Sci. Technol. 2, 768–777 (2008).

    Article  Google Scholar 

  4. Harding, J.L. & Reynolds, M.M. Combating medical device fouling. Trends Biotechnol. 32, 140–146 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Grainger, D.W. All charged up about implanted biomaterials. Nat. Biotechnol. 31, 507–509 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Williams, D.F. On the mechanisms of biocompatibility. Biomaterials 29, 2941–2953 (2008).

    Article  CAS  PubMed  Google Scholar 

  7. Wick, G. et al. The immunology of fibrosis. Annu. Rev. Immunol. 31, 107–135 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Wynn, T.A. & Ramalingam, T.R. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat. Med. 18, 1028–1040 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Zhang, L. et al. Zwitterionic hydrogels implanted in mice resist the foreign-body reaction. Nat. Biotechnol. 31, 553–556 (2013).

    Article  CAS  PubMed  Google Scholar 

  10. Sussman, E.M., Halpin, M.C., Muster, J., Moon, R.T. & Ratner, B.D. Porous implants modulate healing and induce shifts in local macrophage polarization in the foreign body reaction. Ann. Biomed. Eng. 42, 1508–1516 (2013).

    Article  PubMed  Google Scholar 

  11. Rodriguez, A., Meyerson, H. & Anderson, J.M. Quantitative in vivo cytokine analysis at synthetic biomaterial implant sites. J. Biomed. Mater. Res. A 89, 152–159 (2009).

    PubMed  Google Scholar 

  12. Hetrick, E.M., Prichard, H.L., Klitzman, B. & Schoenfisch, M.H. Reduced foreign body response at nitric oxide-releasing subcutaneous implants. Biomaterials 28, 4571–4580 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ratner, B.D. Reducing capsular thickness and enhancing angiogenesis around implant drug release systems. J. Control. Release 78, 211–218 (2002).

    Article  CAS  PubMed  Google Scholar 

  14. Lee, K.Y. & Mooney, D.J. Alginate: properties and biomedical applications. Prog. Polym. Sci. 37, 106–126 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Kearney, C.J. & Mooney, D.J. Macroscale delivery systems for molecular and cellular payloads. Nat. Mater. 12, 1004–1017 (2013).

    Article  CAS  PubMed  Google Scholar 

  16. Lim, F. & Sun, A.M. Microencapsulated islets as bioartificial endocrine pancreas. Science 210, 908–910 (1980).

    Article  CAS  PubMed  Google Scholar 

  17. Duvivier-Kali, V.F., Omer, A., Parent, R.J., O'Neil, J.J. & Weir, G.C. Complete protection of islets against allorejection and autoimmunity by a simple barium-alginate membrane. Diabetes 50, 1698–1705 (2001).

    Article  CAS  PubMed  Google Scholar 

  18. de Vos, P., Faas, M.M., Strand, B. & Calafiore, R. Alginate-based microcapsules for immunoisolation of pancreatic islets. Biomaterials 27, 5603–5617 (2006).

    Article  CAS  PubMed  Google Scholar 

  19. Tuch, B.E. et al. Safety and viability of microencapsulated human islets transplanted into diabetic humans. Diabetes Care 32, 1887–1889 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Weir, G.C. Islet encapsulation: advances and obstacles. Diabetologia 56, 1458–1461 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Jacobs-Tulleneers-Thevissen, D. et al. Beta Cell Therapy Consortium EU-FP7. Sustained function of alginate-encapsulated human islet cell implants in the peritoneal cavity of mice leading to a pilot study in a type 1 diabetic patient. Diabetologia 56, 1605–1614 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Scharp, D.W. & Marchetti, P. Encapsulated islets for diabetes therapy: history, current progress, and critical issues requiring solution. Adv. Drug Deliv. Rev. 67-68, 35–73 (2014).

    Article  CAS  PubMed  Google Scholar 

  23. Robitaille, R. et al. Inflammatory response to peritoneal implantation of alginate-poly-L-lysine microcapsules. Biomaterials 26, 4119–4127 (2005).

    Article  CAS  PubMed  Google Scholar 

  24. Dang, T.T. et al. Enhanced function of immuno-isolated islets in diabetes therapy by co-encapsulation with an anti-inflammatory drug. Biomaterials 34, 5792–5801 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Rokstad, A.M. et al. Alginate microbeads are complement compatible, in contrast to polycation containing microcapsules, as revealed in a human whole blood model. Acta Biomater. 7, 2566–2578 (2011).

    Article  CAS  PubMed  Google Scholar 

  26. King, A., Sandler, S. & Andersson, A. The effect of host factors and capsule composition on the cellular overgrowth on implanted alginate capsules. J. Biomed. Mater. Res. 57, 374–383 (2001).

    Article  CAS  PubMed  Google Scholar 

  27. Manoury, B., Caulet-Maugendre, S., Guénon, I., Lagente, V. & Boichot, E. TIMP-1 is a key factor of fibrogenic response to bleomycin in mouse lung. Int. J. Immunopathol. Pharmacol. 19, 471–487 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Brocchini, S., James, K., Tangpasuthadol, V. & Kohn, J. Structure-property correlations in a combinatorial library of degradable biomaterials. J. Biomed. Mater. Res. 42, 66–75 (1998).

    Article  CAS  PubMed  Google Scholar 

  29. Gu, M. et al. Combinatorial synthesis with high throughput discovery of protein-resistant membrane surfaces. Biomaterials 34, 6133–6138 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bratlie, K.M. et al. Rapid biocompatibility analysis of materials via in vivo fluorescence imaging of mouse models. PLoS One 5, e10032 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Christen, T. et al. Molecular imaging of innate immune cell function in transplant rejection. Circulation 119, 1925–1932 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Haller, J. et al. Visualization of pulmonary inflammation using noninvasive fluorescence molecular imaging. J. Appl. Physiol. 104, 795–802 (2008).

    Article  CAS  PubMed  Google Scholar 

  33. Omer, A. et al. Survival and maturation of microencapsulated porcine neonatal pancreatic cell clusters transplanted into immunocompetent diabetic mice. Diabetes 52, 69–75 (2003).

    Article  CAS  PubMed  Google Scholar 

  34. Omer, A. et al. Long-term normoglycemia in rats receiving transplants with encapsulated islets. Transplantation 79, 52–58 (2005).

    Article  CAS  PubMed  Google Scholar 

  35. Veiseh, O. et al. Size- and shape-dependent foreign body immune response to materials implanted in rodents and non-human primates. Nat. Mater. 14, 643–651 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Hofman, K., Hall, B., Cleaver, H. & Marshall, S. High-throughput quantification of hydroxyproline for determination of collagen. Anal. Biochem. 417, 289–291 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. Paredes-Juarez, G.A., de Haan, B.J., Faas, M.M. & de Vos, P. A technology platform to test the efficacy of purification of alginate. Materials (Basel) 7, 2087–2103 (2014).

    Article  CAS  Google Scholar 

  38. Paredes-Juarez, G.A., de Haan, B.J., Faas, M.M. & de Vos, P. The role of pathogen-associated molecular patterns in inflammatory responses against alginate based microcapsules. J. Control. Release 172, 983–992 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Sato, M. et al. Direct binding of Toll-like receptor 2 to zymosan, and zymosan-induced NF-kappa B activation and TNF-alpha secretion are down-regulated by lung collectin surfactant protein A. J. Immunol. 171, 417–425 (2003).

    Article  CAS  PubMed  Google Scholar 

  40. de Vos, P. et al. Multiscale requirements for bioencapsulation in medicine and biotechnology. Biomaterials 30, 2559–2570 (2009).

    Article  CAS  PubMed  Google Scholar 

  41. Orive, G., Tam, S.K., Pedraz, J.L. & Hallé, J.P. Biocompatibility of alginate-poly-L-lysine microcapsules for cell therapy. Biomaterials 27, 3691–3700 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Dusseault, J. et al. Evaluation of alginate purification methods: effect on polyphenol, endotoxin, and protein contamination. J. Biomed. Mater. Res. A 76, 243–251 (2006).

    Article  PubMed  Google Scholar 

  43. Ménard, M. et al. Role of protein contaminants in the immunogenicity of alginates. J. Biomed. Mater. Res. B Appl. Biomater. 93, 333–340 (2010).

    Article  PubMed  Google Scholar 

  44. Madden, L.R. et al. Proangiogenic scaffolds as functional templates for cardiac tissue engineering. Proc. Natl. Acad. Sci. USA 107, 15211–15216 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Brauker, J.H. et al. Neovascularization of synthetic membranes directed by membrane microarchitecture. J. Biomed. Mater. Res. 29, 1517–1524 (1995).

    Article  CAS  PubMed  Google Scholar 

  46. Liu, M. et al. Stabilized hemocompatible coating of nitinol devices based on photo-cross-linked alginate/heparin multilayer. Langmuir 23, 9378–9385 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Kim, K., Cheng, J., Liu, Q., Wu, X.Y. & Sun, Y. Investigation of mechanical properties of soft hydrogel microcapsules in relation to protein delivery using a MEMS force sensor. J. Biomed. Mater. Res. A 92, 103–113 (2010).

    Article  PubMed  Google Scholar 

  48. de Haan, B.J. et al. Structural surface changes and inflammatory responses against alginate-based microcapsules after exposure to human peritoneal fluid. J. Biomed. Mater. Res. A 98, 394–403 (2011).

    Article  PubMed  Google Scholar 

  49. Qi, M. et al. A recommended laparoscopic procedure for implantation of microcapsules in the peritoneal cavity of non-human primates. J. Surg. Res. 168, e117–e123 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Kharb, R., Sharma, P.C. & Yar, M.S. Pharmacological significance of triazole scaffold. J. Enzyme Inhib. Med. Chem. 26, 1–21 (2011).

    Article  CAS  PubMed  Google Scholar 

  51. Lindstedt, R. et al. The immunosuppressor st1959, a 3,5-diaryl-s-triazole derivative, inhibits T cell activation by reducing NFAT nuclear residency. Int. J. Immunopathol. Pharmacol. 22, 29–42 (2009).

    Article  CAS  PubMed  Google Scholar 

  52. Vegas, A.J. et al. Long term glycemic control using polymer-encapsulated human stem cell–derived beta cells in immune-competent mice. Nat. Med. 10.1038/nm.4030 (25 January 2016).

  53. Stoppel, W.L. et al. Terminal sterilization of alginate hydrogels: efficacy and impact on mechanical properties. J. Biomed. Mater. Res. B Appl. Biomater. 102, 877–884 (2014).

    Article  PubMed  Google Scholar 

  54. Bernhardt, A. et al. Improved sterilization of sensitive biomaterials with supercritical carbon dioxide at low temperature. PLoS One 10, e0129205 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Kang, J.W. et al. Combined confocal Raman and quantitative phase microscopy system for biomedical diagnosis. Biomed. Opt. Express 2, 2484–2492 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Kang, J.W., Nguyen, F.T., Lue, N., Dasari, R.R. & Heller, D.A. Measuring uptake dynamics of multiple identifiable carbon nanotube species via high-speed confocal Raman imaging of live cells. Nano Lett. 12, 6170–6174 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported jointly by the JDRF and Leona M. and the Harry B. Helmsley Charitable Trust (grant 3-SRA-2014-285-M-R), National Institutes of Health (NIH grants EB000244, EB000351, DE013023 and CA151884), NIH NIBIB (P41EB015871-27), MIT SkolTech initiative (J.W.K.), JDRF and the Department of Defense/Congressionally Directed Medical Research Programs (DOD/CDMRP postdoctoral fellowships 3-2013-178 and W81XWH-13-1-0215 for O.V.) and through a generous gift from the Tayebati Family Foundation. G.C.W. is supported by National Institutes of Health (NIH grants R01DK093909 and P30DK036836, the Joslin Diabetes Research Center and its Advanced Microscopy Core), as well as the Diabetes Research and Wellness Foundation. J.O. is supported by the National Institutes of Health (NIH/NIDDK) R01DK091526 and the Chicago Diabetes Project. This work was also supported in part by the Koch Institute Support (core) grant P30-CA14051 from the National Cancer Institute. We also thank the Koch Institute Swanson Biotechnology Center for technical support, specifically Tang Histology Facility, Microscopy, Flow Cytometry, Nanotechnology Materials, and Applied Therapeutics and Whole Animal Imaging. The authors would like to acknowledge the use of resources at the Harvard University Center for Nanoscale Systems and W.M. Keck Biological Imaging Facility (Whitehead Institute). The authors would also like to thank W. Salmon and J. Wyckoff for their assistance.

Author information

Authors and Affiliations

Authors

Contributions

A.J.V., O.V., J.C.D., M.M., K.B., J.L., A.R.B., E.L., K.O., P.F., J.W.K., J.H.-L., M.A.B., A.C., S.S., K.T., S.J., S.A.-D., N.D., R.T., T.V., M.C., J.C., K.S., M.Q. and J.M. designed and performed experiments, and analyzed data. H.H.T. assisted with data processing and data presentation. A.C.G. assisted with SEM imaging. S.L. assisted with histology. D.M.H., D.L.G., J.O. and G.C.W. provided conceptual advice and technical support. A.J.V. and D.G.A. wrote the paper. R.L. and D.G.A. supervised the study. All authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Daniel G Anderson.

Ethics declarations

Competing interests

A.J.V., O.V., J.C.D., M.M., K.B., R.L. and D.G.A. declare financial interest in patents filed by MIT on the material and hydrogel capsule technology.

Integrated supplementary information

Supplementary Figure 1 Reagents used for polymer modification.

Amines, alcohols, azides, and alkynes used for the chemical modification of alginate.

Supplementary Figure 2 Additional data for lead materials tested in C57BL/6J mice.

(a) Evaluation of enzyme activity of cathespins B and L in the absence and presence of 20 mM barium chloride. No reduction in enzymatic activity is observed (n = 6, mean values ± SD). (b) Representative hematoxylin and eosin (HE) stained subcutaneous 28 day histology of the top ten alginate analogue microcapsules and control alginate microcapsules (SLG20, V/S) that were implanted in Figure 2, n = 3 mice per group. Abnormal microcapsule morphology is caused by histological processing (dehydration) of the tissue. Scale bar = 400 µm. (c) PCA functional group analysis of the entire modified alginate library, with top performing materials indicated. Triazole-containing modifications are enriched for favorable in vivo performance. (d) Western-blot images used for SMA quantification in Figure 2c. Protein was extracted from retrieved microcapsules (n = 5 mice per group) of the top three alginate analogues and controls after 14 days IP in C57BL/6J mice. (e) Cellular viability of RAW 264.7 cells exposed to formulated lead microcapsules (n = 12, mean values ± SD). (f) Cytokine panel analysis of protein extracted from retrieved microcapsules (n = 5 mice per group) in Figure 2a. Scale shows average fold signal above background.

Supplementary Figure 3 Nanostring gene expression analysis.

Gene expression analysis of seventy-nine known inflammatory factors and immune cell markers from retrieved IP fluid, fat pad, and microcapsules 14 days post-implantation in C57BL/6J mice. Gene expression from IP and fat pad was normalized to mock implant controls, while gene expression from microcapsule-associated tissue was normalized to the SLG20 sample.

Supplementary Figure 4 Determination of potential contaminants in controls and lead materials.

(a) Determination of potential contaminant levels of alginate microcapsules prior to implantation. Endotoxin and glucan levels were measured and reported by Charles River Laboratories. The standard curves used for quantification of (b) flagellin and (c) LTA levels are also shown.

Supplementary Figure 5 Additional FACS data.

FACS analysis of (a) macrophages and (b) neutrophils isolated from retrieved 300 µm microcapsules after 14 days IP in C57BL/6J mice, n = 5 mice per group. One-way ANOVA with Bonferroni correction was utilized to allow for statistical comparison of multiple means, # = p < 0.05, ** = p < 0.001, *** = p < 0.0001, ns = not signficant. The data express the number of cells as a percentage of total cells measured. (c) Representative dot plots of FACS analysis for macrophages and neutrophils presented in (a) and (b).

Supplementary Figure 6 Additional confocal raman and cryo-SEM imaging of lead materials.

(a) Confocal raman cross-section mapping of 300 µm Z1-Y15 microcapsules. The raman peak at 857 cm-1 (shown in red) is indicative of the thiomorpholine dioxide end group of Z1-Y15, and peak intensity is enriched at the surface of the microcapsules than at the core. The peak at 884 cm-1 is mapped in green as a reference to the alginate backbone structure. (b) Confocal raman cross-section mapping of 300 µm Z1-Y19 microcapsules. The raman peak at 1563 cm-1 (shown in red) is indicative of the aniline end group of Z1-Y19, and peak intensity appears more uniform at both the surface of the microcapsules and at the core. The peak at 884 cm-1 is mapped in green as a reference to the alginate backbone structure. (c) Representative freeze-fracture cryo-SEM images of the core (scale bar = 10 µm) and fractured surface (scale bar = 10 µm) of 300 µm SLG20, V/S, Z2-Y12, Z1-Y15, and Z1-Y19 microcapsules.

Supplementary Figure 7 NHP omental histology and western blot images of lead material implants.

(a) Representative MT and HE stained histology of biopsied omental tissue 4 weeks post-implantation in cynomolgus macaque, n = 3. (b) Western-blot images used for SMA quantification of protein extracted from the top three alginate analogue spheres and control spheres (n = 3).

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vegas, A., Veiseh, O., Doloff, J. et al. Combinatorial hydrogel library enables identification of materials that mitigate the foreign body response in primates. Nat Biotechnol 34, 345–352 (2016). https://doi.org/10.1038/nbt.3462

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3462

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing