Skip to main content

Advertisement

Log in

Regenerative medicine for anal incontinence: a review of regenerative therapies beyond cells

  • Review Article
  • Published:
International Urogynecology Journal Aims and scope Submit manuscript

Abstract

Introduction and hypothesis

Current treatment modalities for anal sphincter injuries are ineffective for many patients, prompting research into restorative and regenerative therapies. Although cellular therapy with stem cells and progenitor cells show promise in animal models with short-term improvement, there are additional regenerative approaches that can augment or replace cellular therapies for anal sphincter injuries. The purpose of this article is to review the current knowledge of cellular therapies for anal sphincter injuries and discusses the use of other regenerative therapies including cytokine therapy with CXCL12.

Methods

A literature search was performed to search for articles on cellular therapy and cytokine therapy for anal sphincter injuries and anal incontinence.

Results

The article search identified 337 articles from which 33 articles were included. An additional 12 referenced articles were included as well as 23 articles providing background information. Cellular therapy has shown positive results for treating anal sphincter injuries and anal incontinence in vitro and in one clinical trial. However, cellular therapy has disadvantages such as the source and processing of stem cells and progenitor cells. CXCL12 does not have such issues while showing promising in vitro results for treating anal sphincter injuries. Additionally, electrical stimulation and extracorporeal shock wave therapy are potential regenerative medicine adjuncts for anal sphincter injuries. A vision for future research and clinical applications of regenerative medicine for anal sphincter deficiencies is provided.

Conclusion

There are viable regenerative medicine therapies for anal sphincter injuries beyond cellular therapy. CXCL12 shows promise as a focus of therapeutic research in this field.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

Abbreviations

OASI:

obstetrical anal sphincter injury

SDF-1:

stromal-derived factor 1

IM:

intramuscular

IV:

intravenous

GFP:

green fluorescent protein

ISD:

intrinsic sphincter deficiency

ESWT:

extracorporeal shock wave therapy

References

  1. Macmillan AK, et al. The prevalence of fecal incontinence in community-dwelling adults: a systematic review of the literature. Dis Colon Rectum. 2004;47(8):1341–9.

    Article  PubMed  Google Scholar 

  2. Bharucha AE, et al. Prevalence and burden of fecal incontinence: a population-based study in women. Gastroenterology. 2005;129(1):42–9.

    Article  PubMed  Google Scholar 

  3. Harvey MA, et al. Obstetrical anal sphincter injuries (OASIS): prevention, recognition, and repair. J Obstet Gynaecol Can. 2015;37(12):1131–48.

    Article  PubMed  Google Scholar 

  4. Snooks SJ, Henry MM, Swash M. Faecal incontinence due to external anal sphincter division in childbirth is associated with damage to the innervation of the pelvic floor musculature: a double pathology. Br J Obstet Gynaecol. 1985;92(8):824–8.

    Article  CAS  PubMed  Google Scholar 

  5. Allen RE, et al. Pelvic floor damage and childbirth: a neurophysiological study. Br J Obstet Gynaecol. 1990;97(9):770–9.

    Article  CAS  PubMed  Google Scholar 

  6. Sandridge DA, et al. Vaginal delivery is associated with occult disruption of the anal sphincter mechanism. Am J Perinatol. 1997;14(9):527–33.

    Article  CAS  PubMed  Google Scholar 

  7. Crawford LA, et al. Incontinence following rupture of the anal sphincter during delivery. Obstet Gynecol. 1993;82(4 Pt 1):527–31.

    CAS  PubMed  Google Scholar 

  8. Omar MI, Alexander CE. Drug treatment for faecal incontinence in adults. Cochrane Database Syst Rev. 2013;6:CD002116.

    Google Scholar 

  9. Brown SR, Wadhawan H, Nelson RL. Surgery for faecal incontinence in adults. Cochrane Database Syst Rev. 2013;7:CD001757.

    Google Scholar 

  10. Graf W, et al. Efficacy of dextranomer in stabilised hyaluronic acid for treatment of faecal incontinence: a randomised, sham-controlled trial. Lancet. 2011;377(9770):997–1003.

    Article  CAS  PubMed  Google Scholar 

  11. Wexner SD, et al. Sacral nerve stimulation for fecal incontinence: results of a 120-patient prospective multicenter study. Ann Surg. 2010;251(3):441–9.

    Article  PubMed  Google Scholar 

  12. Lam TJ, et al. Clinical response and sustainability of treatment with temperature-controlled radiofrequency energy (Secca) in patients with faecal incontinence: 3 years follow-up. Int J Color Dis. 2014;29(6):755–61.

    Article  CAS  Google Scholar 

  13. Majka M, et al. Concise review: mesenchymal stem cells in cardiovascular regeneration: emerging research directions and clinical applications. Stem Cells Transl Med. 2017;6(10):1859–67.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Bitar KN, Zakhem E. Bioengineering the gut: future prospects of regenerative medicine. Nat Rev Gastroenterol Hepatol. 2016;13(9):543–56.

    Article  CAS  PubMed  Google Scholar 

  15. Williams JK, et al. Regenerative medicine therapies for stress urinary incontinence. J Urol. 2016;196(6):1619–26.

    Article  PubMed  Google Scholar 

  16. Sun L, et al. Electrical stimulation followed by mesenchymal stem cells improves anal sphincter anatomy and function in a rat model at a time remote from injury. Dis Colon Rectum. 2016;59(5):434–42.

    Article  PubMed  Google Scholar 

  17. Fitzwater JL, et al. Effect of myogenic stem cells on the integrity and histomorphology of repaired transected external anal sphincter. Int Urogynecol J. 2015;26(2):251–6.

    Article  PubMed  Google Scholar 

  18. Lorenzi B, et al. Treatment of experimental injury of anal sphincters with primary surgical repair and injection of bone marrow-derived mesenchymal stem cells. Dis Colon Rectum. 2008;51(4):411–20.

    Article  PubMed  Google Scholar 

  19. Montoya TI, et al. Myogenic stem cell-laden hydrogel scaffold in wound healing of the disrupted external anal sphincter. Int Urogynecol J. 2015;26(6):893–904.

    Article  PubMed  Google Scholar 

  20. White AB, et al. Effect of myogenic stem cells on contractile properties of the repaired and unrepaired transected external anal sphincter in an animal model. Obstet Gynecol. 2010;115(4):815–23.

    Article  PubMed  Google Scholar 

  21. Cruz M, et al. Pelvic organ distribution of mesenchymal stem cells injected intravenously after simulated childbirth injury in female rats. Obstet Gynecol Int. 2012;2012:612946.

    Article  PubMed  CAS  Google Scholar 

  22. Pathi SD, et al. Recovery of the injured external anal sphincter after injection of local or intravenous mesenchymal stem cells. Obstet Gynecol. 2012;119(1):134–44.

    Article  CAS  PubMed  Google Scholar 

  23. Salcedo L, et al. Mesenchymal stem cells can improve anal pressures after anal sphincter injury. Stem Cell Res. 2013;10(1):95–102.

    Article  PubMed  Google Scholar 

  24. Salcedo L, et al. Functional outcome after anal sphincter injury and treatment with mesenchymal stem cells. Stem Cells Transl Med. 2014;3(6):760–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Salcedo L, et al. Chemokine upregulation in response to anal sphincter and pudendal nerve injury: potential signals for stem cell homing. Int J Color Dis. 2011;26(12):1577–81.

    Article  Google Scholar 

  26. Lane FL, et al. In vivo recovery of the injured anal sphincter after repair and injection of myogenic stem cells: an experimental model. Dis Colon Rectum. 2013;56(11):1290–7.

    Article  PubMed  Google Scholar 

  27. Aghaee-Afshar M, et al. Potential of human umbilical cord matrix and rabbit bone marrow-derived mesenchymal stem cells in repair of surgically incised rabbit external anal sphincter. Dis Colon Rectum. 2009;52(10):1753–61.

    Article  PubMed  Google Scholar 

  28. Kajbafzadeh AM, et al. Functional external anal sphincter reconstruction for treatment of anal incontinence using muscle progenitor cell auto grafting. Dis Colon Rectum. 2010;53(10):1415–21.

    Article  PubMed  Google Scholar 

  29. Oh HK, et al. Functional and histological evidence for the targeted therapy using biocompatible polycaprolactone beads and autologous myoblasts in a dog model of fecal incontinence. Dis Colon Rectum. 2015;58(5):517–25.

    Article  PubMed  Google Scholar 

  30. Frudinger A, et al. Muscle-derived cell injection to treat anal incontinence due to obstetric trauma: pilot study with 1 year follow-up. Gut. 2010;59(1):55–61.

    Article  CAS  PubMed  Google Scholar 

  31. Frudinger A, et al. Autologous skeletal-muscle-derived cell injection for anal incontinence due to obstetric trauma: a 5-year follow-up of an initial study of 10 patients. Color Dis. 2015;17(9):794–801.

    Article  CAS  Google Scholar 

  32. Gras S, Tolstrup CK, Lose G. Regenerative medicine provides alternative strategies for the treatment of anal incontinence. Int Urogynecol J. 2017;28(3):341–50.

    Article  PubMed  Google Scholar 

  33. Cottler-Fox MH, et al. Stem cell mobilization. Hematology Am Soc Hematol Educ Program. 2003;2003:419–37.

    Article  Google Scholar 

  34. Lapidot T, Petit I. Current understanding of stem cell mobilization: the roles of chemokines, proteolytic enzymes, adhesion molecules, cytokines, and stromal cells. Exp Hematol. 2002;30(9):973–81.

    Article  CAS  PubMed  Google Scholar 

  35. Zhou S, et al. Stem cell therapy for treatment of stress urinary incontinence: the current status and challenges. Stem Cells Int. 2016;2016:7060975.

    PubMed  PubMed Central  Google Scholar 

  36. Williams JK, et al. Local versus intravenous injections of skeletal muscle precursor cells in nonhuman primates with acute or chronic intrinsic urinary sphincter deficiency. Stem Cell Res Ther. 2016;7(1):147.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Williams JK, et al. Cell versus chemokine therapy in a nonhuman primate model of chronic intrinsic urinary sphincter deficiency. J Urol. 2016;196(6):1809–15.

    Article  CAS  PubMed  Google Scholar 

  38. Sun L, et al. Regenerating the anal sphincter: cytokines, stem cells, or both? Dis Colon Rectum. 2017;60(4):416–25.

    Article  PubMed  Google Scholar 

  39. Koudy Williams J, et al. Efficacy and initial safety profile of CXCL12 treatment in a rodent model of urinary sphincter deficiency. Stem Cells Transl Med. 2017;6(8):1740–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Deng K, et al. Mesenchymal stem cells and their secretome partially restore nerve and urethral function in a dual muscle and nerve injury stress urinary incontinence model. Am J Physiol Renal Physiol. 2015;308(2):F92–F100.

    Article  CAS  PubMed  Google Scholar 

  41. Kang SB, et al. Sphincter contractility after muscle-derived stem cells autograft into the cryoinjured anal sphincters of rats. Dis Colon Rectum. 2008;51(9):1367–73.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Aref-Adib M, et al. Stem cell therapy for stress urinary incontinence: a systematic review in human subjects. Arch Gynecol Obstet. 2013;288(6):1213–21.

    Article  CAS  PubMed  Google Scholar 

  43. Hijaz AK, et al. Stem cell homing factor, CCL7, expression in mouse models of stress urinary incontinence. Female Pelvic Med Reconstr Surg. 2013;19(6):356–61.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Lenis AT, et al. Impact of parturition on chemokine homing factor expression in the vaginal distention model of stress urinary incontinence. J Urol. 2013;189(4):1588–94.

    Article  CAS  PubMed  Google Scholar 

  45. Couri BM, et al. Effect of pregnancy and delivery on cytokine expression in a mouse model of pelvic organ prolapse. Female Pelvic Med Reconstr Surg. 2017;23(6):449–56.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Chatterjee M, Gawaz M. Platelet-derived CXCL12 (SDF-1alpha): basic mechanisms and clinical implications. J Thromb Haemost. 2013;11(11):1954–67.

    Article  CAS  PubMed  Google Scholar 

  47. Duda DG, et al. CXCL12 (SDF1alpha)-CXCR4/CXCR7 pathway inhibition: an emerging sensitizer for anticancer therapies? Clin Cancer Res. 2011;17(8):2074–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Wurth R, et al. CXCL12 modulation of CXCR4 and CXCR7 activity in human glioblastoma stem-like cells and regulation of the tumor microenvironment. Front Cell Neurosci. 2014;8:144.

    PubMed  PubMed Central  Google Scholar 

  49. Teicher BA, Fricker SP. CXCL12 (SDF-1)/CXCR4 pathway in cancer. Clin Cancer Res. 2010;16(11):2927–31.

    Article  CAS  PubMed  Google Scholar 

  50. Pawig L, et al. Diversity and inter-connections in the CXCR4 chemokine receptor/ligand family: molecular perspectives. Front Immunol. 2015;6:429.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Scala S. Molecular pathways: targeting the CXCR4-CXCL12 Axis--untapped potential in the tumor microenvironment. Clin Cancer Res. 2015;21(19):4278–85.

    Article  CAS  PubMed  Google Scholar 

  52. Peled A, et al. Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science. 1999;283(5403):845–8.

    Article  CAS  PubMed  Google Scholar 

  53. Aiuti A, et al. The chemokine SDF-1 is a chemoattractant for human CD34+ hematopoietic progenitor cells and provides a new mechanism to explain the mobilization of CD34+ progenitors to peripheral blood. J Exp Med. 1997;185(1):111–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Peled A, et al. The chemokine SDF-1 activates the integrins LFA-1, VLA-4, and VLA-5 on immature human CD34(+) cells: role in transendothelial/stromal migration and engraftment of NOD/SCID mice. Blood. 2000;95(11):3289–96.

    Article  CAS  PubMed  Google Scholar 

  55. MacArthur JW Jr, et al. Mathematically engineered stromal cell-derived factor-1alpha stem cell cytokine analog enhances mechanical properties of infarcted myocardium. J Thorac Cardiovasc Surg. 2013;145(1):278–84.

    Article  CAS  PubMed  Google Scholar 

  56. Penn MS, et al. An open-label dose escalation study to evaluate the safety of administration of nonviral stromal cell-derived factor-1 plasmid to treat symptomatic ischemic heart failure. Circ Res. 2013;112(5):816–25.

    Article  CAS  PubMed  Google Scholar 

  57. Kuraitis D, et al. A stromal cell-derived factor-1 releasing matrix enhances the progenitor cell response and blood vessel growth in ischaemic skeletal muscle. Eur Cell Mater. 2011;22:109–23.

    Article  CAS  PubMed  Google Scholar 

  58. Balog BM, et al. Electrical stimulation for neuroregeneration in urology: a new therapeutic paradigm. Curr Opin Urol. 2019;29(4):458–65.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Jiang HH, et al. Electrical stimulation of the pudendal nerve promotes neuroregeneration and functional recovery from stress urinary incontinence in a rat model. Am J Physiol Renal Physiol. 2018;315(6):F1555–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Salcedo L, et al. Low current electrical stimulation upregulates cytokine expression in the anal sphincter. Int J Color Dis. 2012;27(2):221–5.

    Article  Google Scholar 

  61. Romeo P, et al. Extracorporeal shock wave therapy in musculoskeletal disorders: a review. Med Princ Pract. 2014;23(1):7–13.

    Article  PubMed  Google Scholar 

  62. Fukumoto Y, et al. Extracorporeal cardiac shock wave therapy ameliorates myocardial ischemia in patients with severe coronary artery disease. Coron Artery Dis. 2006;17(1):63–70.

    Article  PubMed  Google Scholar 

  63. Lu Z, et al. Low-intensity extracorporeal shock wave treatment improves erectile function: a systematic review and meta-analysis. Eur Urol. 2017;71(2):223–33.

    Article  PubMed  Google Scholar 

  64. Lin G, et al. In situ activation of penile progenitor cells with low-intensity extracorporeal shockwave therapy. J Sex Med. 2017;14(4):493–501.

    Article  PubMed  Google Scholar 

  65. Bauer G, Elsallab M, Abou-El-Enein M. Concise review: a comprehensive analysis of reported adverse events in patients receiving unproven stem cell-based interventions. Stem Cells Transl Med. 2018;7(9):676–85.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Berkowitz AL, et al. Glioproliferative lesion of the spinal cord as a complication of "stem-cell tourism". N Engl J Med. 2016;375(2):196–8.

    Article  PubMed  Google Scholar 

  67. Amariglio N, et al. Donor-derived brain tumor following neural stem cell transplantation in an ataxia telangiectasia patient. PLoS Med. 2009;6(2):e1000029.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Fischer UM, et al. Pulmonary passage is a major obstacle for intravenous stem cell delivery: the pulmonary first-pass effect. Stem Cells Dev. 2009;18(5):683–92.

    Article  CAS  PubMed  Google Scholar 

  69. Ott HC, McCue J, Taylor DA. Cell-based cardiovascular repair--the hurdles and the opportunities. Basic Res Cardiol. 2005;100(6):504–17.

    Article  CAS  PubMed  Google Scholar 

  70. Baldo BA. Side effects of cytokines approved for therapy. Drug Saf. 2014;37(11):921–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Song M, et al. Regeneration of chronic myocardial infarction by injectable hydrogels containing stem cell homing factor SDF-1 and angiogenic peptide ac-SDKP. Biomaterials. 2014;35(8):2436–45.

    Article  CAS  PubMed  Google Scholar 

  72. Liu H, et al. CXCR4 antagonist delivery on decellularized skin scaffold facilitates impaired wound healing in diabetic mice by increasing expression of SDF-1 and enhancing migration of CXCR4-positive cells. Wound Repair Regen. 2017;25(4):652–64.

    Article  PubMed  Google Scholar 

  73. Shafiq M, Kong D, Kim SH. SDF-1alpha peptide tethered polyester facilitates tissue repair by endogenous cell mobilization and recruitment. J Biomed Mater Res A. 2017;105(10):2670–84.

    Article  CAS  PubMed  Google Scholar 

  74. Bromage DI, Davidson SM, Yellon DM. Stromal derived factor 1alpha: a chemokine that delivers a two-pronged defence of the myocardium. Pharmacol Ther. 2014;143(3):305–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Kanki S, et al. Stromal cell-derived factor-1 retention and cardioprotection for ischemic myocardium. Circ Heart Fail. 2011;4(4):509–18.

    Article  CAS  PubMed  Google Scholar 

  76. Kowalski K, et al. Stromal derived factor-1 and granulocyte-colony stimulating factor treatment improves regeneration of Pax7−/− mice skeletal muscles. J Cachexia Sarcopenia Muscle. 2016;7(4):483–96.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Janine Tillett MSLS AHIP from the Wake Forest School of Medicine Carpenter Library was instrumental in performing the academic article searches for this article.

Author information

Authors and Affiliations

Authors

Contributions

Andre Plair: Protocol/project development, data collection, manuscript writing.

Julie Bennington: Manuscript editing.

James Koudy Williams: Manuscript editing.

Candace Parker-Autry: Manuscript editing.

Catherine Ann Matthews: Protocol/project development, manuscript editing.

Gopal Badlani: Protocol/project development, manuscript editing.

Corresponding author

Correspondence to Andre Plair.

Ethics declarations

Conflict of interest

Andre Plair, grant support from Neomedic.

Julie Bennington, supported by NIH Post-doctoral T32 Institutional Training Grant “Laboratory Animal and Comparative Medicine Research” (NIH T32 OD010957).

James Koudy Williams, None.

Candace Parker-Autry, None.

Catherine Ann Matthews, consultant for Boston Scientific and Pelvalon; grant support from Boston Scientific and Neomedic; expert defense for Johnson and Johnson.

Gopal Badlani, advisory panelist for Olympus and Neotract; board of directors of AAGUS, and Urology Care Foundation.

Additional information

Publisher’s note

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Plair, A., Bennington, J., Williams, J.K. et al. Regenerative medicine for anal incontinence: a review of regenerative therapies beyond cells. Int Urogynecol J 32, 2337–2347 (2021). https://doi.org/10.1007/s00192-020-04620-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00192-020-04620-x

Keywords

Navigation