We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Colonic delivery of vasoactive intestinal peptide nanomedicine alleviates colitis and shows promise as an oral capsule

    Dulari Jayawardena

    Department of Medicine, College of Medicine, University of Illinois at Chicago, IL 60612, USA

    Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, IL 60612, USA

    ,
    Arivarasu N Anbazhagan

    Department of Medicine, College of Medicine, University of Illinois at Chicago, IL 60612, USA

    ,
    Shubha Priyamvada

    Department of Medicine, College of Medicine, University of Illinois at Chicago, IL 60612, USA

    ,
    Anoop Kumar

    Department of Medicine, College of Medicine, University of Illinois at Chicago, IL 60612, USA

    Jesse Brown VA Medical Center, Chicago, IL 60612, USA

    ,
    Seema Saksena

    Department of Medicine, College of Medicine, University of Illinois at Chicago, IL 60612, USA

    Jesse Brown VA Medical Center, Chicago, IL 60612, USA

    ,
    Hayat Onyuksel

    Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, IL 60612, USA

    &
    Pradeep K Dudeja

    *Author for correspondence: Tel.: +1 (312) 569 7434;

    E-mail Address: pkdudeja@uic.edu

    Department of Medicine, College of Medicine, University of Illinois at Chicago, IL 60612, USA

    Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, IL 60612, USA

    Jesse Brown VA Medical Center, Chicago, IL 60612, USA

    Published Online:https://doi.org/10.2217/nnm-2020-0280

    Aim: To evaluate the efficacy of locally delivered nanomedicine, vasoactive intestinal peptide in sterically stabilized micelles (VIP-SSM) to the colon and conduct in vitro release studies of a potential oral formulation. Materials & methods: Intracolonic instillation of VIP-SSM was tested in a mouse model of dextran sulfate sodium-induced colitis. Based on the effective mouse dose, human equivalent dose containing nanomedicine powder was filled into enteric coated capsules for in vitro release testing. Results: Colonic delivery of VIP-SSM significantly alleviated colitis. VIP-SSM containing capsules completely dissolved at colonic pH allowing micelles to reform with active VIP. Capsule formulations exhibited reproducible release profiles when stored up to 6 weeks demonstrating stability. Conclusion: VIP-SSM is an effective nanomedicine formulation which appears to have potential for oral treatment of colitis in humans.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Abad C, Waschek JA. Immunomodulatory roles of VIP and PACAP in models of multiple sclerosis. Curr. Pharm. Des. 17(10), 1025–1035 (2011).
    • 2. Delgado M, Martinez C, Pozo D et al. Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activation polypeptide (PACAP) protect mice from lethal endotoxemia through the inhibition of TNF-α and IL-6. J. Immunol. 162(2), 1200–1205 (1999).
    • 3. Gomariz R, Martinez C, Abad C, Leceta J, Delgado M. Immunology of VIP: a review and therapeutical perspectives. Curr. Pharm. Des. 7(2), 89–111 (2001).
    • 4. Smalley S, Barrow P, Foster N. Immunomodulation of innate immune responses by vasoactive intestinal peptide (VIP): its therapeutic potential in inflammatory disease. Clin. Exp. Immunol. 157(2), 225–234 (2009). • Highlights the immunomodulatory properties of vasoactive intestinal peptide (VIP) and therefore forms the basis of its use in inflammatory diseases.
    • 5. Seo S, Miyake H, Alganabi M et al. Vasoactive intestinal peptide decreases inflammation and tight junction disruption in experimental necrotizing enterocolitis. J. Pediatr. Surg. 54(12), 2520–2523 (2019).
    • 6. Campbell J, Berry J, Liang Y. Anatomy and physiology of the small intestine. In: Shackelford's Surgery of the Alimentary Tract - 2 Volume Set. Elsevier, PA, USA, 817–841 (2019).
    • 7. Seillet C, Luong K, Tellier J et al. The neuropeptide VIP confers anticipatory mucosal immunity by regulating ILC3 activity. Nat. Immunol. 21, 168–177 (2019).
    • 8. Jönsson M, Norrgård Ö, Hansson M, Forsgren S. Decrease in binding for the neuropeptide VIP in response to marked inflammation of the mucosa in ulcerative colitis. Ann. NY Acad. Sci. 1107(1), 280–289 (2007).
    • 9. Mazumdar S, Das KM. Immunocytochemical localization of vasoactive intestinal peptide and substance P in the colon from normal subjects and patients with inflammatory bowel disease. Am. J. Gastroenterol. 87(2), 176–181 (1992).
    • 10. Cassuto J, Fahrenkrug J, Jodal M, Tuttle R, Lundgren O. Release of vasoactive intestinal polypeptide from the cat small intestine exposed to cholera toxin. Gut 22(11), 958–963 (1981).
    • 11. Guan X, Karpen HE, Stephens J et al. GLP-2 receptor localizes to enteric neurons and endocrine cells expressing vasoactive peptides and mediates increased blood flow. Gastroenterology 130(1), 150–164 (2006).
    • 12. Dharmsathaphorn K, Harms V, Yamashiro DJ, Hughes R, Binder H, Wright E. Preferential binding of vasoactive intestinal polypeptide to basolateral membrane of rat and rabbit enterocytes. J. Clin. Invest. 71(1), 27–35 (1983).
    • 13. Jayawardena D, Guzman G, Gill RK, Alrefai WA, Onyuksel H, Dudeja PK. Expression and localization of VPAC1, the major receptor of vasoactive intestinal peptide along the length of the intestine. Am. J. Physiol. Gastrointest. Liver Physiol. 313(1), G16–G25 (2017). • The presence of VPAC1 receptors in human and mouse colonic epithelium is characterized. This is the receptor which we target in this manuscript.
    • 14. Jayawardena D, Anbazhagan AN, Guzman G, Dudeja PK, Onyuksel H. Vasoactive intestinal peptide nanomedicine for the management of inflammatory bowel disease. Mol. Pharm. 14(11), 3698–3708 (2017). •• This is the first time vasoactive intestinal peptide in sterically stabilized micelles (VIP-SSM) was tested in colitis and the optimal dose for the current study was decided upon.
    • 15. Abad C, Martinez C, Juarranz MG et al. Therapeutic effects of vasoactive intestinal peptide in the trinitrobenzene sulfonic acid mice model of Crohn's disease. Gastroenterology 124(4), 961–971 (2003). • Describes the beneficial effect of VIP for the first time in Inflammatory bowel disease. Frequent high doses were used for alleviating inflammation.
    • 16. Motlekar NA, Youan B-BC. The quest for non-invasive delivery of bioactive macromolecules: a focus on heparins. J. Control. Rel. 113(2), 91–101 (2006).
    • 17. Lim SB, Rubinstein I, Önyüksel H. Freeze drying of peptide drugs self-associated with long-circulating, biocompatible and biodegradable sterically stabilized phospholipid nanomicelles. Int. J. Pharm. 356(1–2), 345–350 (2008). • VIP-SSM freeze dried product is characterized, thereby demonstrating the stability of this peptide nanomedicine during the freeze drying process.
    • 18. Nair AB, Jacob S. A simple practice guide for dose conversion between animals and human. J. Basic Clin. Pharm. 7(2), 27 (2016). • Interspecies dosage conversion is discussed which was employed in the calculation of the human equivalent dose.
    • 19. Sethi V, Rubinstein I, Kuzmis A, Kastrissios H, Artwohl J, Onyuksel H. Novel, biocompatible, and disease modifying VIP nanomedicine for rheumatoid arthritis. Mol. Pharm. 10(2), 728–738 (2013). •• VIP-SSM nanomedicine was characterized for its physico-chemical properties and demonstrated the stability of this formulation in vivo.
    • 20. Banerjee A, Onyuksel H. Peptide delivery using phospholipid micelles. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 4(5), 562–574 (2012).
    • 21. Lim SB, Banerjee A, Önyüksel H. Improvement of drug safety by the use of lipid-based nanocarriers. J. Control. Rel. 163(1), 34–45 (2012).
    • 22. Vukovic L, Madriaga A, Kuzmis A et al. Solubilization of therapeutic agents in micellar nanomedicines. Langmuir 29(51), 15747–15754 (2013). • VIP-SSM structure was determined by molecular dynamic simulation studies.
    • 23. Krawisz J, Sharon P, Stenson W. Quantitative assay for acute intestinal inflammation based on myeloperoxidase activity: assessment of inflammation in rat and hamster models. Gastroenterology 87(6), 1344–1350 (1984).
    • 24. Viennois E, Chen F, Laroui H, Baker MT, Merlin D. Dextran sodium sulfate inhibits the activities of both polymerase and reverse transcriptase: lithium chloride purification, a rapid and efficient technique to purify RNA. BMC Res. Notes 6(1), 360 (2013).
    • 25. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative CT method. Nat. Protoc. 3(6), 1101–1108 (2008).
    • 26. Lindén SK, Florin TH, Mcguckin MA. Mucin dynamics in intestinal bacterial infection. PLoS ONE 3(12), e3952 (2008).
    • 27. McConnell EL, Fadda HM, Basit AW. Gut instincts: explorations in intestinal physiology and drug delivery. Int. J. Pharm. 364(2), 213–226 (2008).
    • 28. Önyüksel H, Bodalia B, Sethi V, Dagar S, Rubinsteina I. Surface-active properties of vasoactive intestinal peptide*. Peptides 21(3), 419–423 (2000).
    • 29. Kaplan GG. The global burden of IBD: from 2015 to 2025. Nat. Rev. Gastroenterol. Hepatol. 12(12), 720–727 (2015).
    • 30. Frokjaer S, Otzen DE. Protein drug stability: a formulation challenge. Nat. Rev. Drug Discov. 4(4), 298 (2005).
    • 31. Lewis AL, Richard J. Challenges in the delivery of peptide drugs: an industry perspective. Ther. Deliv. 6(2), 149–163 (2015).
    • 32. Richard J. Challenges in oral peptide delivery: lessons learnt from the clinic and future prospects. Ther. Deliv. 8(8), 663–684 (2017).
    • 33. Sarmento B, Ribeiro A, Veiga F, Sampaio P, Neufeld R, Ferreira D. Alginate/chitosan nanoparticles are effective for oral insulin delivery. Pharm. Res. 24(12), 2198–2206 (2007).
    • 34. Sarmento B, Martins S, Ferreira D, Souto EB. Oral insulin delivery by means of solid lipid nanoparticles. Int. J. Nanomedicine 2(4), 743 (2007).
    • 35. Araújo F, Shrestha N, Shahbazi M-A et al. The impact of nanoparticles on the mucosal translocation and transport of GLP-1 across the intestinal epithelium. Biomaterials 35(33), 9199–9207 (2014).
    • 36. Li C, Zhao Y, Cheng J et al. A proresolving peptide nanotherapy for site-specific treatment of inflammatory bowel disease by regulating proinflammatory microenvironment and gut microbiota. Adv. Sci. 6(18), 1900610 (2019).
    • 37. Laroui H, Dalmasso G, Nguyen HTT, Yan Y, Sitaraman SV, Merlin D. Drug-loaded nanoparticles targeted to the colon with polysaccharide hydrogel reduce colitis in a mouse model. Gastroenterology 138(3), 843–853 (2010).
    • 38. Ensign LM, Cone R, Hanes J. Oral drug delivery with polymeric nanoparticles: the gastrointestinal mucus barriers. Adv. Drug Deliv. Rev. 64(6), 557–570 (2012).
    • 39. Des Rieux A, Fievez V, Garinot M, Schneider Y-J, Préat V. Nanoparticles as potential oral delivery systems of proteins and vaccines: a mechanistic approach. J. Control. Rel. 116(1), 1–27 (2006).
    • 40. Hamman JH, Enslin GM, Kotzé AF. Oral delivery of peptide drugs. BioDrugs 19(3), 165–177 (2005).
    • 41. Hua S, Marks E, Schneider JJ, Keely S. Advances in oral nano-delivery systems for colon targeted drug delivery in inflammatory bowel disease: selective targeting to diseased versus healthy tissue. Nanomedicine 11(5), 1117–1132 (2015).
    • 42. Xiao B, Merlin D. Oral colon-specific therapeutic approaches toward treatment of inflammatory bowel disease. Expert Opin. Drug Deliv. 9(11), 1393–1407 (2012).
    • 43. Taipaleenmäki EM, Mouritzen SA, Schattling PS, Zhang Y, Städler B. Mucopenetrating micelles with a PEG corona. Nanoscale 9(46), 18438–18448 (2017).
    • 44. Chapman NJ, Brown ML, Phillips SF et al. Distribution of mesalamine enemas in patients with active distal ulcerative colitis. Mayo Clin. Proc. 67(3), 245–248 (1992).
    • 45. Sandborn W, Tremaine W, Leighton JA et al. Nicotine tartrate liquid enemas for mildly to moderately active left-sided ulcerative colitis unresponsive to first-line therapy: a pilot study. Aliment. Pharmacol. Ther. 11(4), 663–671 (1997).
    • 46. Greenfield NJ. Using circular dichroism spectra to estimate protein secondary structure. Nat. Protoc. 1(6), 2876 (2006).
    • 47. Mody R, Tramontano A, Paul S. Spontaneous hydrolysis of vasoactive intestinal peptide in neutral aqueous solution. Chem. Biol. Drug Des. 44(5), 441–447 (1994).
    • 48. Cui X, Cao D, Qu C, Zhang X, Zheng A. A study of the chemical and biological stability of vasoactive intestinal peptide. Drug Dev. Ind. Pharm. 39(12), 1907–1910 (2013).