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.

  • Article
  • Published:

An injectable and biodegradable zwitterionic gel for extending the longevity and performance of insulin infusion catheters

Abstract

Continuous subcutaneous insulin infusion (CSII) is an essential insulin replacement therapy in the management of diabetes. However, the longevity of clinical CSII is limited by skin complications, by impaired insulin absorption and by occlusions associated with the subcutaneous insertion of CSII catheters, which require replacement and rotation of the insertion site every few days. Here we show that a biodegradable zwitterionic gel covering the tip end of commercial off-the-shelf CSII catheters fully resolves early skin irritations, extends the longevity of catheters and improves the rate of insulin absorption (also with respect to conventional syringe-based subcutaneous injection) for longer than 6 months in diabetic mice, and by 11 days in diabetic minipigs (from 2 to 13 days, under standard CSII-wearing conditions of insulin pump therapy and in a continuous basal-plus-bolus-infusion setting). The implanted gel displayed anti-inflammatory and anti-foreign-body-reaction properties and promoted the local formation of new blood vessels. The gel is subcutaneously injected before the tip of catheter is inserted into it, and should be generally applicable to CSII catheters and other implantable devices.

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

Access options

Buy this article

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

Fig. 1: Schematic representation of injectable zwitterionic gel enabling the long-term use of infusion catheter and faster insulin absorption.
Fig. 2: Zwitterionic gel fully resolved early inflammatory responses and skin irritation.
Fig. 3: Zwitterionic gel enabled the long-term use of infusion catheters and faster insulin absorption in non-diabetic C57BL/6 mice.
Fig. 4: Zwitterionic gel enabled long-term use of an insulin infusion catheter and faster insulin absorption in fresh-induced diabetic C57BL/6 mice.
Fig. 5: Zwitterionic gel enabled the long-term use of insulin infusion catheters and faster insulin absorption in mice with a 3 month diabetic history.
Fig. 6: Zwitterionic gel retained both presence and function in diabetic mice under practical pump therapy conditions.
Fig. 7: Zwitterionic gel enabled the long-term use of subcutaneous infusion catheters and faster insulin absorption in diabetic pigs.
Fig. 8: Degradable gel enables the long-term use of SC infusion catheters and faster insulin absorption in diabetic pigs.

Similar content being viewed by others

Data availability

The main data supporting the results in this study are available within the paper and its Supplementary Information. All data generated or analysed during the study are available for research purposes from the corresponding author on reasonable request.

References

  1. Kopan, C. et al. Approaches in immunotherapy, regenerative medicine, and bioengineering for type 1 diabetes. Front. Immunol. 9, 1354 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Boldison, J. & Wong, F. S. Immune and pancreatic β cell interactions in type 1 diabetes. Trends Endocrinol. Metab. 27, 856–867 (2016).

    Article  PubMed  CAS  Google Scholar 

  3. Maikawa, C. L. et al. A co-formulation of supramolecularly stabilized insulin and pramlintide enhances mealtime glucagon suppression in diabetic pigs. Nat. Biomed. Eng. 4, 507–517 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Pathak, V., Pathak, N. M., O’Neill, C. L., Guduric-Fuchs, J. & Medina, R. J. Therapies for type 1 diabetes: current scenario and future perspectives. Clin. Med. Insights Endocrinol. Diabetes 12, 1179551419844521 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Han, X. et al. Zwitterionic micelles efficiently deliver oral insulin without opening tight junctions. Nat. Nanotechnol. 15, 605–614 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Jeitler, K. et al. Continuous subcutaneous insulin infusion versus multiple daily insulin injections in patients with diabetes mellitus: systematic review and meta-analysis. Diabetologia 51, 941–951 (2008).

    Article  PubMed  CAS  Google Scholar 

  7. Monami, M., Lamanna, C., Marchionni, N. & Mannucci, E. Continuous subcutaneous insulin infusion versus multiple daily insulin injections in type 1 diabetes: a meta-analysis. Acta Diabetol. 47, 77–81 (2010).

    Article  PubMed  CAS  Google Scholar 

  8. Birkebaek, N. et al. Quality of life in Danish children and adolescents with type 1 diabetes treated with continuous subcutaneous insulin infusion or multiple daily injections. Diabetes Res. Clin. Pract. 106, 474–480 (2014).

    Article  PubMed  CAS  Google Scholar 

  9. Tauschmann, M. & Hovorka, R. Technology in the management of type 1 diabetes mellitus—current status and future prospects. Nat. Rev. Endocrinol. 14, 464–475 (2018).

    Article  PubMed  Google Scholar 

  10. Pickup, J. C. Is insulin pump therapy effective in Type 1 diabetes? Diabet. Med. 36, 269–278 (2019).

    Article  PubMed  CAS  Google Scholar 

  11. Reznik, Y. et al. Insulin pump treatment compared with multiple daily injections for treatment of type 2 diabetes (OpT2mise): a randomised open-label controlled trial. Lancet 384, 1265–1272 (2014).

    Article  PubMed  CAS  Google Scholar 

  12. Pickup, J. C. Diabetes: insulin pump therapy for type 2 diabetes mellitus. Nat. Rev. Endocrinol. 10, 647–649 (2014).

    Article  PubMed  CAS  Google Scholar 

  13. Freckmann, G. et al. Insulin pump therapy for patients with type 2 diabetes mellitus: evidence, current barriers, and new technologies. J. Diabetes Sci. Technol. 15, 901–915 (2021).

  14. Heinemann, L. Insulin infusion sets: a critical reappraisal. Diabetes Technol. Ther. 18, 327–333 (2016).

    Article  PubMed  Google Scholar 

  15. Heinemann, L., Walsh, J. & Roberts, R. We need more research and better designs for insulin infusion sets. J. Diabetes Sci. Technol. 8, 199–202 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Pickup, J. C., Yemane, N., Brackenridge, A. & Pender, S. Nonmetabolic complications of continuous subcutaneous insulin infusion: a patient survey. Diabetes Technol. Ther. 16, 145–149 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Schober, E. & Rami, B. Dermatological side effects and complications of continuous subcutaneous insulin infusion in preschool-age and school-age children. Pediatr. Diabetes 10, 198–201 (2009).

    Article  PubMed  Google Scholar 

  18. Pickup, J. C. Insulin-pump therapy for type 1 diabetes mellitus. N. Engl. J. Med. 366, 1616–1624 (2012).

    Article  PubMed  CAS  Google Scholar 

  19. Thethi, T. K. et al. Consequences of delayed pump infusion line change in patients with type 1 diabetes mellitus treated with continuous subcutaneous insulin infusion. J. Diabetes Complications 24, 73–78 (2010).

    Article  PubMed  Google Scholar 

  20. Schmid, V., Hohberg, C., Borchert, M., Forst, T. & Pfützner, A. Pilot study for assessment of optimal frequency for changing catheters in insulin pump therapy—trouble starts on day 3. J. Diabetes Sci. Technol. 4, 976–982 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Karinka, S. A. et al. Improved accuracy of 14-day factory-calibrated FreeStyle Libre system with new glucose algorithm. Diabetes 68, 910 (2019).

    Article  Google Scholar 

  22. Bequette, B. W. Challenges and recent progress in the development of a closed-loop artificial pancreas. Annu. Rev. Control 36, 255–266 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Chug, M. K., Feit, C. & Brisbois, E. J. Increasing the lifetime of insulin cannula with antifouling and nitric oxide releasing properties. ACS Appl. Bio. Mater. 2, 5965–5975 (2019).

    Article  PubMed  CAS  Google Scholar 

  24. Hauzenberger, J. R. et al. Systematic in vivo evaluation of the time-dependent inflammatory response to steel and Teflon insulin infusion catheters. Sci. Rep. 8, 1132 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Zhang, E. & Cao, Z. Tissue response to subcutaneous infusion catheter. J. Diabetes Sci. Technol. 14, 226–232 (2020).

    Article  PubMed  Google Scholar 

  26. Mecklenburg, R. S. et al. Long-term metabolic control with insulin pump therapy: report of experience with 127 patients. N. Engl. J. Med. 313, 465–468 (1985).

    Article  PubMed  CAS  Google Scholar 

  27. Sampson Perrin, A. J. et al. A web-based study of the relationship of duration of insulin pump infusion set use and fasting blood glucose level in adults with type 1 diabetes. Diabetes Technol. Ther. 17, 307–310 (2015).

    Article  PubMed  CAS  Google Scholar 

  28. Eisler, G. et al. In vivo investigation of the tissue response to commercial Teflon insulin infusion sets in large swine for 14 days: the effect of angle of insertion on tissue histology and insulin spread within the subcutaneous tissue. BMJ Open Diabetes Res. Care 7, e000881 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Pozzilli, P. et al. Continuous subcutaneous insulin infusion in diabetes: patient populations, safety, efficacy, and pharmacoeconomics. Diabetes Metab. Res. Rev. 32, 21–39 (2016).

    Article  PubMed  CAS  Google Scholar 

  30. Home, P. The pharmacokinetics and pharmacodynamics of rapid‐acting insulin analogues and their clinical consequences. Diabetes Obes. Metab. 14, 780–788 (2012).

    Article  PubMed  CAS  Google Scholar 

  31. Haahr, H. & Heise, T. Fast-acting insulin aspart: a review of its pharmacokinetic and pharmacodynamic properties and the clinical consequences. Clin. Pharmacokinet. 59, 155–172 (2020).

    Article  PubMed  CAS  Google Scholar 

  32. Pepper, A. R. et al. A prevascularized subcutaneous device-less site for islet and cellular transplantation. Nat. Biotechnol. 33, 518–523 (2015).

    Article  PubMed  CAS  Google Scholar 

  33. Song, W. et al. Engineering transferrable microvascular meshes for subcutaneous islet transplantation. Nat. Commun. 10, 4602 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Vlahos, A. E., Cober, N. & Sefton, M. V. Modular tissue engineering for the vascularization of subcutaneously transplanted pancreatic islets. Proc. Natl Acad. Sci. USA 114, 9337–9342 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Leslie, D. C. et al. A bioinspired omniphobic surface coating on medical devices prevents thrombosis and biofouling. Nat. Biotechnol. 32, 1134–1140 (2014).

    Article  PubMed  CAS  Google Scholar 

  36. Hook, A. L. et al. Combinatorial discovery of polymers resistant to bacterial attachment. Nat. Biotechnol. 30, 868–875 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Smith, R. S. et al. Vascular catheters with a nonleaching poly-sulfobetaine surface modification reduce thrombus formation and microbial attachment. Sci. Transl. Med. 4, 153ra132 (2012).

    Article  PubMed  Google Scholar 

  38. Miao, X. et al. TNF-α/TNFR1 signaling is required for the full expression of acute and chronic itch in mice via peripheral and central mechanisms. Neurosci. Bull. 34, 42–53 (2018).

    Article  PubMed  CAS  Google Scholar 

  39. Rask-Madsen, C. & King, G. L. Vascular complications of diabetes: mechanisms of injury and protective factors. Cell Metab. 17, 20–33 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Rivard, A. et al. Rescue of diabetes-related impairment of angiogenesis by intramuscular gene therapy with adeno-VEGF. Am. J. Pathol. 154, 355–363 (1999).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Costa, P. Z. & Soares, R. Neovascularization in diabetes and its complications. Unraveling the angiogenic paradox. Life Sci. 92, 1037–1045 (2013).

    Article  PubMed  CAS  Google Scholar 

  42. Wong, S. L. et al. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat. Med. 21, 815–819 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Soumya, D. & Srilatha, B. Late stage complications of diabetes and insulin resistance. J. Diabetes Metab. 2, 1000167 (2011).

    Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  45. Liu, Q. et al. Zwitterionically modified alginates mitigate cellular overgrowth for cell encapsulation. Nat. Commun. 10, 5262 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Chen, X. & Yang, D. Functional zwitterionic biomaterials for administration of insulin. Biomater. Sci. 8, 4906–4919 (2020).

    Article  PubMed  CAS  Google Scholar 

  47. Evert, A. B. et al. Improving patient experience with insulin infusion sets: practical guidelines and future directions. Diabetes Educ. 42, 470–484 (2016).

    Article  PubMed  Google Scholar 

  48. Yu, J. et al. Glucose-responsive insulin patch for the regulation of blood glucose in mice and minipigs. Nat. Biomed. Eng. 4, 499–506 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Zhang, E. et al. Biodegradable zwitterionic cream gel for effective prevention of postoperative adhesion. Adv. Funct. Mater. 31, 2009431 (2021).

    Article  PubMed  CAS  Google Scholar 

  50. Zhang, E. et al. Fouling-resistant zwitterionic polymers for complete prevention of postoperative adhesion. Proc. Natl Acad. Sci. USA 117, 32046–32055 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Institute of Diabetes and Digestive and Kidney Diseases of the National Institutes of Health (grant no. DP2DK111910). This work was partially supported by the National Institute of Diabetes and Digestive and Kidney Diseases of the National Institutes of Health (grant no. R01DK123293), National Science Foundation (grant nos. 410853 and 1809229), Juvenile Diabetes Research Foundation (grant nos. 1-SRA-2015-41-A-N, 1-SRA-2016-270-A-N, 2-PNF-2016-324-S-B and 2-SRA-2017-429-S-B) and the faculty start-up fund at Wayne State University.

Author information

Authors and Affiliations

Authors

Contributions

E.Z. and Z.C. conceived the project and designed the experiments. E.Z. conducted all experiments. Y.S. helped with pig experiments. X.H. helped with the inflammatory cytokines assay. H.Z. helped with mouse experiments. B.S. helped with synthesis of CBAA. C.Y. helped with statistical analysis. E.Z. and Z.C. analysed experimental data and wrote the paper. Z.C. supervised the entire study.

Corresponding author

Correspondence to Zhiqiang Cao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks the anonymous reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Blood glucose and time to the lowest blood glucose at day 60 and 180 post-implantation in non-diabetic C57BL/6 mice.

Mice received insulin administration (2 IU/kg) through the lumen of the completely implanted catheter in treated and untreated groups and mice receiving syringe-based SC insulin injection at the same dosage were used as control (n = 6 biologically independent animals in each group, mean ± s.d.). A one-way ANOVA with Tukey multi-comparison was used for statistical analysis. For the untreated group at day 180 post-implantation, insulin administration through the lumen of the implanted catheter only can be performed in 2 of 6 mice due to the occlusion of the untreated catheter, and the valid blood glucose data are presented as mean (n = 2 biologically independent animals).

Extended Data Fig. 2 Representative blood vessel staining images in non-diabetic C57BL/6 mice.

Blood vessel staining (brown) using MECA-32 antibody on tissues surrounding SC implanted catheter from different groups at day 14, 30, 60, and 90 post-implantation in healthy C57BL/6 mice. Skin tissue samples collected from mice without implantation surgery were used as a control. Arrow indicates newly formed blood vessels around the zwitterionic gel treated catheter. C: catheter. Scale bar, 100 μm.

Extended Data Fig. 3 Blood glucose and time to the lowest blood glucose at day 60 and 180 post-implantation in fresh-diabetic mice.

Mice received insulin administration (4 IU/kg) through the lumen of the completely implanted catheter in treated and untreated groups and mice receiving syringe-based SC insulin injection at the same dosage were used as control (n = 6 biologically independent animals in each group, mean ± s.d.). A one-way ANOVA with Tukey multi-comparison was used for statistical analysis at day 60. A two-tailed t-test analysis was used for statistical analysis at day 180. For the untreated group at day 180 post-implantation, insulin administration through the lumen of the implanted catheter only can be performed in 1 mouse due to the occlusion of the untreated catheter.

Extended Data Fig. 4 Representative blood vessel staining images in fresh-diabetic C57BL/6 mice.

Blood vessel staining (brown) using MECA-32 antibody on tissues surrounding SC implanted catheter from different groups at day 14, 30, 60, and 90 post-implantation in fresh-diabetic C57BL/6 mice. Skin tissue samples collected from mice without implantation surgery were used as a control. Arrow indicates newly formed blood vessels around the zwitterionic gel treated catheter. C: catheter. Scale bar, 100 μm.

Extended Data Fig. 5 Blood glucose and time to the lowest blood glucose at day 60 and 180 post-implantation in three-month diabetic mice.

Mice received insulin administration (4 IU/kg) through the lumen of the completely implanted catheter in treated and untreated groups and mice receiving syringe-based SC insulin injection at the same dosage were used as control (n = 6 biologically independent animals in each group, mean ± s.d.). A one-way ANOVA with Tukey multi-comparison was used for statistical analysis at day 60. A two-tailed t-test analysis was used for statistical analysis at day 180. For the untreated group at day 180 post-implantation, no insulin administration through the lumen of the implanted catheter only can be performed due to the occlusion of the untreated catheter.

Extended Data Fig. 6 Representative blood vessel staining images in three-month diabetic C57BL/6 mice.

Blood vessel staining (brown) using MECA-32 antibody on tissues surrounding SC implanted catheter from different groups at day 14, 30, 60, and 90 post-implantation in three-month diabetic C57BL/6 mice. Skin tissue samples collected from mice without implantation surgery were used as a control. Arrow indicates newly formed blood vessels around the zwitterionic gel treated catheter. C: catheter. Scale bar, 100 μm.

Extended Data Fig. 7 Representative immunofluorescence staining images in three-month diabetic C57BL/6 mice.

Blood vessel staining using MECA-32 antibody and alpha-smooth muscle actin (α-SMA) antibody on tissues surrounding SC implanted catheter from mice with three-month diabetic history at day 14, 30, and 90 post-implantation. Skin tissue samples collected from mice with three-month diabetic history without receiving implantation surgery were used as a control. SC: subcutaneous; C: catheter. Arrow indicates newly formed blood vessels around the zwitterionic gel treated catheter. Scale bar, 100 μm.

Extended Data Fig. 8 The macrophage recruitment and accumulation in tissues surrounding SC implanted catheter from mice with three-month diabetic history at day 14 and 30 post-implantation.

a, Representative immunofluorescence staining using F4/80 antibody on tissues surrounding SC implanted catheter. Skin tissue samples collected from mice with three-month diabetic history without receiving implantation surgery were used as a control. C: catheter. Arrow indicates the zwitterionic gel around the treated catheter. Scale bar, 100 μm. b, c, Mean fluorescence intensity of F4/80 surrounding SC implanted catheter at day 14 (b) and 30 (c) post-implantation (n = 3 biologically independent samples, mean ± s.d.). Quantitative analysis was performed by ImageJ. A one-way ANOVA with Tukey multi-comparison was used for statistical analysis. Both treated and untreated catheters showed a remarkable macrophage accumulation at both time points relative to the control.

Extended Data Fig. 9 The pro-inflammatory M1 polarization and anti-inflammatory M2 polarization in tissues surrounding SC implanted catheter from mice with three-month diabetic history at day 14 and 30 post-implantation.

a, Representative immunofluorescence staining using CD86 antibody for M1 and CD206 antibody for M2 on tissues surrounding SC implanted catheter from mice with three-month diabetic history at day 14 and 30 post-implantation. Skin tissue samples collected from mice with three-month diabetic history without receiving implantation surgery were used as a control. C: catheter. Arrow indicates the zwitterionic gel around the treated catheter. Scale bar, 100 μm. b, c, The ratio of M2/M1 surrounding SC implanted catheter at day 14 (b) and 30 (c) post-implantation (n = 3 biologically independent samples, mean ± s.d.) was calculated by measuring the mean fluorescence intensity of CD206 over CD86. A one-way ANOVA with Tukey multi-comparison was used for statistical analysis.

Extended Data Fig. 10 The rheological properties, in vitro and in vivo degradability, and vascularizing function of degradable zwitterionic gel after SC injection in healthy C57BL/6 mice.

a, Photograph of the prepared degradable zwitterionic gel (100 uL). b, Representative micrograph of degradable zwitterionic gel after dispersed in DI water. c, d, Frequency-dependent (c, under 1% strain, 25 °C) and strain-dependent (d, 10 rad/s frequency, 25 °C) oscillatory sweeps of degradable zwitterionic gel. e, In vitro degradation of degradable zwitterionic gel in PBS containing 20 µM GSH (n = 3 independent samples, mean ± s.d.) f, The degradable zwitterionic gel (100 uL) was SC implanted into the mice (n = 5 for each group at each time point). g, h, Representative Masson trichrome staining images (g) and blood vessel staining (brown) using MECA-32 antibody (h) on tissues surrounding SC injected degradable zwitterionic gel at day 30, 60, and 90. Scale bar, 1000 μm in g and 200 μm in h. i, Blood vessel density at different time points post-injection (n = 5 biologically independent samples at each time point, mean ± s.d.). A one-way ANOVA with Tukey multi-comparison was used for statistical analysis.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, E., Shi, Y., Han, X. et al. An injectable and biodegradable zwitterionic gel for extending the longevity and performance of insulin infusion catheters. Nat. Biomed. Eng (2023). https://doi.org/10.1038/s41551-023-01108-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41551-023-01108-z

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