Skip to main content

Advertisement

Log in

New Advances in Neuromodulation

  • Pain Medicine (GJ Meredith, Section Editor)
  • Published:
Current Anesthesiology Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

The purpose of this article is to illustrate the recent advances made in the field of neuromodulation for chronic pain management and it provides a brief review of the literature supporting their safe and efficacious use.

Recent Findings

Spinal cord stimulation (SCS) has been an effective therapy for the management of intractable pain conditions including complex regional pain syndrome (CRPS), peripheral neuropathy, and failed back surgery syndrome (FBSS). With the introduction of high-frequency and burst SCS waveforms, there has been a noted improvement in the treatment outcomes of these pain syndromes. Dorsal root ganglion stimulation and peripheral nerve stimulation have made it possible to achieve pain relief in the focal areas of pain, which was difficult before with the use of conventional SCS.

Summary

Chronic pain conditions afflict a large population and are the leading causes of disability among industrialized nations. Conditions such as CRPS, peripheral neuropathy, and FBSS are often difficult to treat with conservative medical measures. This has been one of the factors that have led to increased opioid prescribing and has contributed to the current opioid crisis. Neuromodulation offers a relatively safe and effective therapy for the chronic pain patients that have failed conservative pain measures or are unwilling to undergo higher risk options such as spine surgery or opioid therapy.

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.

Institutional subscriptions

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Collaborators USBoD, Mokdad AH, Ballestros K, Echko M, Glenn S, Olsen HE, et al. The state of US health, 1990–2016: burden of diseases, injuries, and risk factors among US states. JAMA. 2018;319(14):1444–72.

    Google Scholar 

  2. Dieleman JL, Baral R, Birger M, Bui AL, Bulchis A, Chapin A, et al. US spending on personal health care and public health, 1996–2013. JAMA. 2016;316(24):2627–46.

    PubMed  PubMed Central  Google Scholar 

  3. Dworkin RH, O’Connor AB, Backonja M, Farrar JT, Finnerup NB, Jensen TS, et al. Pharmacologic management of neuropathic pain: evidence-based recommendations. Pain. 2007;132(3):237–51.

    CAS  PubMed  Google Scholar 

  4. Moulin DE, Clark AJ, Gilron I, Ware MA, Watson CP, Sessle BJ, et al. Pharmacological management of chronic neuropathic pain - consensus statement and guidelines from the Canadian Pain Society. Pain Res Manag. 2007;12(1):13–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Daubresse M, Chang HY, Yu Y, Viswanathan S, Shah ND, Stafford RS, et al. Ambulatory diagnosis and treatment of nonmalignant pain in the United States, 2000–2010. Med Care. 2013;51(10):870–8.

    PubMed  Google Scholar 

  6. Annual surveillance report of drug-related risks and outcomes—United States. https://www.cdc.gov/drugoverdose/pdf/pubs/2017-cdc-drug-surveillance-report.pdf. 2017.

  7. Melzack R, Wall PD. Pain mechanisms: a new theory. Science. 1965;150(3699):971–9.

    CAS  PubMed  Google Scholar 

  8. Shealy CN, Mortimer JT, Reswick JB. Electrical inhibition of pain by stimulation of the dorsal columns: preliminary clinical report. Anesth Analg. 1967;46(4):489–91.

    CAS  PubMed  Google Scholar 

  9. Gildenberg PL. Treatment of spasmodic torticollis with dorsal column stimulation. Acta Neurochir (Wien). 1977;(Suppl 24):65–6.

  10. Cook AW. Electrical stimulation in multiple sclerosis. Hosp Pract. 1976;11(4):51–8.

    CAS  PubMed  Google Scholar 

  11. Dooley DM, Kasprak M. Modification of blood flow to the extremities by electrical stimulation of the nervous system. South Med J. 1976;69(10):1309–11.

    CAS  PubMed  Google Scholar 

  12. Augustinsson LE, Carlsson CA, Holm J, Jivegard L. Epidural electrical stimulation in severe limb ischemia. Pain relief, increased blood flow, and a possible limb-saving effect. Ann Surg. 1985;202(1):104–10.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Murphy DF, Giles KE. Dorsal column stimulation for pain relief from intractable angina pectoris. Pain. 1987;28(3):365–8.

    CAS  PubMed  Google Scholar 

  14. Zhang TC, Janik JJ, Grill WM. Mechanisms and models of spinal cord stimulation for the treatment of neuropathic pain. Brain Res. 2014;1569:19–31.

    CAS  PubMed  Google Scholar 

  15. Zhang TC, Janik JJ, Peters RV, Chen G, Ji RR, Grill WM. Spinal sensory projection neuron responses to spinal cord stimulation are mediated by circuits beyond gate control. J Neurophysiol. 2015;114(1):284–300.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Yakhnitsa V, Linderoth B, Meyerson BA. Spinal cord stimulation attenuates dorsal horn neuronal hyperexcitability in a rat model of mononeuropathy. Pain. 1999;79(2–3):223–33.

    CAS  PubMed  Google Scholar 

  17. Stiller CO, Cui JG, O’Connor WT, Brodin E, Meyerson BA, Linderoth B. Release of gamma-aminobutyric acid in the dorsal horn and suppression of tactile allodynia by spinal cord stimulation in mononeuropathic rats. Neurosurgery. 1996;39(2):367–74 discussion 74-5.

    CAS  PubMed  Google Scholar 

  18. Schechtmann G, Song Z, Ultenius C, Meyerson BA, Linderoth B. Cholinergic mechanisms involved in the pain relieving effect of spinal cord stimulation in a model of neuropathy. Pain. 2008;139(1):136–45.

    CAS  PubMed  Google Scholar 

  19. Cui JG, Meyerson BA, Sollevi A, Linderoth B. Effect of spinal cord stimulation on tactile hypersensitivity in mononeuropathic rats is potentiated by simultaneous GABA(B) and adenosine receptor activation. Neurosci Lett. 1998;247(2–3):183–6.

    CAS  PubMed  Google Scholar 

  20. Linderoth B, Gazelius B, Franck J, Brodin E. Dorsal column stimulation induces release of serotonin and substance P in the cat dorsal horn. Neurosurgery. 1992;31(2):289–96 discussion 96–7.

    CAS  PubMed  Google Scholar 

  21. Barchini J, Tchachaghian S, Shamaa F, Jabbur SJ, Meyerson BA, Song Z, et al. Spinal segmental and supraspinal mechanisms underlying the pain-relieving effects of spinal cord stimulation: an experimental study in a rat model of neuropathy. Neuroscience. 2012;215:196–208.

    CAS  PubMed  Google Scholar 

  22. Al-Kaisy A, Palmisani S, Smith TE, Carganillo R, Houghton R, Pang D, et al. Long-term improvements in chronic axial low back pain patients without previous spinal surgery: a cohort analysis of 10-kHz high-frequency spinal cord stimulation over 36 months. Pain Med. 2018;19(6):1219–26.

    PubMed  Google Scholar 

  23. • Amirdelfan K, Yu C, Doust MW, Gliner BE, Morgan DM, Kapural L, et al. Long-term quality of life improvement for chronic intractable back and leg pain patients using spinal cord stimulation: 12-month results from the SENZA-RCT. Qual Life Res. 2018;27(8):2035–44 A landmark study demonstrating the success of high-frequency SCS.

    PubMed  Google Scholar 

  24. Kapural L, Yu C, Doust MW, Gliner BE, Vallejo R, Sitzman BT, et al. Novel 10-kHz high-frequency therapy (HF10 therapy) is superior to traditional low-frequency spinal cord stimulation for the treatment of chronic back and leg pain: the SENZA-RCT randomized controlled trial. Anesthesiology. 2015;123(4):851–60.

    PubMed  Google Scholar 

  25. Kilgore KL, Bhadra N. Reversible nerve conduction block using kilohertz frequency alternating current. Neuromodulation. 2014;17(3):242–54 discussion 54–5.

    PubMed  Google Scholar 

  26. Litvak LM, Smith ZM, Delgutte B, Eddington DK. Desynchronization of electrically evoked auditory-nerve activity by high-frequency pulse trains of long duration. J Acoust Soc Am. 2003;114(4 Pt 1):2066–78.

    PubMed  Google Scholar 

  27. Reilly JP, Freeman VT, Larkin WD. Sensory effects of transient electrical stimulation--evaluation with a neuroelectric model. IEEE Trans Biomed Eng. 1985;32(12):1001–11.

    CAS  PubMed  Google Scholar 

  28. De Ridder D, Lenders MW, De Vos CC, Dijkstra-Scholten C, Wolters R, Vancamp T, et al. A 2-center comparative study on tonic versus burst spinal cord stimulation: amount of responders and amount of pain suppression. Clin J Pain. 2015;31(5):433–7.

    PubMed  Google Scholar 

  29. Muhammad S, Roeske S, Chaudhry SR, Kinfe TM. Burst or high-frequency (10 kHz) spinal cord stimulation in failed back surgery syndrome patients with predominant back pain: one year comparative data. Neuromodulation. 2017;20(7):661–7.

    PubMed  Google Scholar 

  30. De Ridder D, van der Loo E, Van der Kelen K, Menovsky T, van de Heyning P, Moller A. Do tonic and burst TMS modulate the lemniscal and extralemniscal system differentially? Int J Med Sci. 2007;4(5):242–6.

    PubMed  PubMed Central  Google Scholar 

  31. De Ridder D, Vanneste S. Burst and tonic spinal cord stimulation: different and common brain mechanisms. Neuromodulation. 2016;19(1):47–59.

    PubMed  Google Scholar 

  32. Kemler MA, Barendse GA, van Kleef M, de Vet HC, Rijks CP, Furnee CA, et al. Spinal cord stimulation in patients with chronic reflex sympathetic dystrophy. N Engl J Med. 2000;343(9):618–24.

    CAS  PubMed  Google Scholar 

  33. Kumar K, Rizvi S, Nguyen R, Abbas M, Bishop S, Murthy V. Impact of wait times on spinal cord stimulation therapy outcomes. Pain Pract. 2014;14(8):709–20.

    PubMed  Google Scholar 

  34. Rizvi S, Kumar K. Spinal cord stimulation for chronic pain: the importance of early referral. Pain Manag. 2014;4(5):329–31.

    PubMed  Google Scholar 

  35. • Russo M, Cousins MJ, Brooker C, Taylor N, Boesel T, Sullivan R, et al. Effective relief of pain and associated symptoms with closed-loop spinal cord stimulation system: preliminary results of the Avalon study. Neuromodulation. 2018;21(1):38–47 An important study demonstrating effectiveness of closed-loop SCS.

    PubMed  Google Scholar 

  36. Pope JE, Deer TR, Falowski S, Provenzano D, Hanes M, Hayek SM, et al. Multicenter retrospective study of neurostimulation with exit of therapy by explant. Neuromodulation. 2017;20(6):543–52.

    PubMed  Google Scholar 

  37. Bevan S, Yeats J. Protons activate a cation conductance in a sub-population of rat dorsal root ganglion neurones. J Physiol. 1991;433:145–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Deer TR, Grigsby E, Weiner RL, Wilcosky B, Kramer JM. A prospective study of dorsal root ganglion stimulation for the relief of chronic pain. Neuromodulation. 2013;16(1):67–71 discussion −2.

    PubMed  Google Scholar 

  39. Liem L, Russo M, Huygen FJ, Van Buyten JP, Smet I, Verrills P, et al. A multicenter, prospective trial to assess the safety and performance of the spinal modulation dorsal root ganglion neurostimulator system in the treatment of chronic pain. Neuromodulation. 2013;16(5):471–82 discussion 82.

    PubMed  Google Scholar 

  40. Sheng SR, Wang XY, Xu HZ, Zhu GQ, Zhou YF. Anatomy of large animal spines and its comparison to the human spine: a systematic review. Eur Spine J. 2010;19(1):46–56.

    PubMed  Google Scholar 

  41. Gemes G, Koopmeiners A, Rigaud M, Lirk P, Sapunar D, Bangaru ML, et al. Failure of action potential propagation in sensory neurons: mechanisms and loss of afferent filtering in C-type units after painful nerve injury. J Physiol. 2013;591(4):1111–31.

    CAS  PubMed  Google Scholar 

  42. Wang W, Gu J, Li YQ, Tao YX. Are voltage-gated sodium channels on the dorsal root ganglion involved in the development of neuropathic pain? Mol Pain. 2011;7:16.

    PubMed  PubMed Central  Google Scholar 

  43. Li JY, Xie W, Strong JA, Guo QL, Zhang JM. Mechanical hypersensitivity, sympathetic sprouting, and glial activation are attenuated by local injection of corticosteroid near the lumbar ganglion in a rat model of neuropathic pain. Reg Anesth Pain Med. 2011;36(1):56–62.

    PubMed  PubMed Central  Google Scholar 

  44. Xie WR, Deng H, Li H, Bowen TL, Strong JA, Zhang JM. Robust increase of cutaneous sensitivity, cytokine production and sympathetic sprouting in rats with localized inflammatory irritation of the spinal ganglia. Neuroscience. 2006;142(3):809–22.

    CAS  PubMed  Google Scholar 

  45. •• Deer TR, Levy RM, Kramer J, Poree L, Amirdelfan K, Grigsby E, et al. Dorsal root ganglion stimulation yielded higher treatment success rate for complex regional pain syndrome and causalgia at 3 and 12 months: a randomized comparative trial. Pain. 2017;158(4):669–81 A landmark clinical trial showing efficacy of DRG stimulation over conventional SCS and the only available RCT till date for DRG stimulation.

    PubMed  Google Scholar 

  46. •• Deer TR, Pope JE, Lamer TJ, Grider JS, Provenzano D, Lubenow TR, et al. The neuromodulation appropriateness consensus committee on best practices for dorsal root ganglion stimulation. Neuromodulation. 2018; An extremely important practice guidelines for physicians performing DRG stimulation published by the neuromodulation appropriateness concensus committee.

  47. Schu SMH, Kapur S, Guru R, Leljevahl E, Wahlstedt A, Eldabe S. Sustained pain relief in painful diabetic neuropathy (pdn) achieved through targeted spinal cord stimulation (SCS): a retrospective case series. Neuromodulation. 2015;18(2):82.

    Google Scholar 

  48. Eldabe SEA, Kang P. Dorsal root ganglia stimulation for painful diabetic neuropathy: a preliminary report. Neuromodulation. 2017;20:e51.

    Google Scholar 

  49. Julius A. A treatise on medical electricity, theoretical and practical; and its use in the treatment of paralysis, neuralgia, and other diseases. London: Longmans, Green, AND CO. 1873.

  50. Campbell JN, Long DM. Peripheral nerve stimulation in the treatment of intractable pain. J Neurosurg. 1976;45(6):692–9.

    CAS  PubMed  Google Scholar 

  51. Weiner RL, Reed KL. Peripheral neurostimulation for control of intractable occipital neuralgia. Neuromodulation. 1999;2(3):217–21.

    CAS  PubMed  Google Scholar 

  52. Radhakrishnan R, King EW, Dickman JK, Herold CA, Johnston NF, Spurgin ML, et al. Spinal 5-HT(2) and 5-HT(3) receptors mediate low, but not high, frequency TENS-induced antihyperalgesia in rats. Pain. 2003;105(1–2):205–13.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Radhakrishnan R, Sluka KA. Spinal muscarinic receptors are activated during low or high frequency TENS-induced antihyperalgesia in rats. Neuropharmacology. 2003;45(8):1111–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Sluka KA, Deacon M, Stibal A, Strissel S, Terpstra A. Spinal blockade of opioid receptors prevents the analgesia produced by TENS in arthritic rats. J Pharmacol Exp Ther. 1999;289(2):840–6.

    CAS  PubMed  Google Scholar 

  55. Saper JR, Dodick DW, Silberstein SD, McCarville S, Sun M, Goadsby PJ, et al. Occipital nerve stimulation for the treatment of intractable chronic migraine headache: ONSTIM feasibility study. Cephalalgia. 2011;31(3):271–85.

    PubMed  PubMed Central  Google Scholar 

  56. Silberstein SD, Dodick DW, Saper J, Huh B, Slavin KV, Sharan A, et al. Safety and efficacy of peripheral nerve stimulation of the occipital nerves for the management of chronic migraine: results from a randomized, multicenter, double-blinded, controlled study. Cephalalgia. 2012;32(16):1165–79.

    PubMed  Google Scholar 

  57. Schoenen J, Jensen RH, Lanteri-Minet M, Lainez MJ, Gaul C, Goodman AM, et al. Stimulation of the sphenopalatine ganglion (SPG) for cluster headache treatment. Pathway CH-1: a randomized, sham-controlled study. Cephalalgia. 2013;33(10):816–30.

    PubMed  PubMed Central  Google Scholar 

  58. Young RF. Electrical stimulation of the trigeminal nerve root for the treatment of chronic facial pain. J Neurosurg. 1995;83(1):72–8.

    CAS  PubMed  Google Scholar 

  59. Taub E, Munz M, Tasker RR. Chronic electrical stimulation of the Gasserian ganglion for the relief of pain in a series of 34 patients. J Neurosurg. 1997;86(2):197–202.

    CAS  PubMed  Google Scholar 

  60. Wilson RD, Harris MA, Gunzler DD, Bennett ME, Chae J. Percutaneous peripheral nerve stimulation for chronic pain in subacromial impingement syndrome: a case series. Neuromodulation. 2014;17(8):771–6 discussion 6.

    PubMed  PubMed Central  Google Scholar 

  61. Wilson RD, Gunzler DD, Bennett ME, Chae J. Peripheral nerve stimulation compared with usual care for pain relief of hemiplegic shoulder pain: a randomized controlled trial. Am J Phys Med Rehabil. 2014;93(1):17–28.

    PubMed  PubMed Central  Google Scholar 

  62. Guentchev M, Preuss C, Rink R, Peter L, Wocker EL, Tuettenberg J. Technical note: treatment of sacroiliac joint pain with peripheral nerve stimulation. Neuromodulation. 2015;18(5):392–6.

    PubMed  Google Scholar 

  63. Deer TR, Levy RM, Rosenfeld EL. Prospective clinical study of a new implantable peripheral nerve stimulation device to treat chronic pain. Clin J Pain. 2010;26(5):359–72.

    PubMed  Google Scholar 

  64. Stinson LW Jr, Roderer GT, Cross NE, Davis BE. Peripheral subcutaneous electrostimulation for control of intractable post-operative inguinal pain: a case report series. Neuromodulation. 2001;4(3):99–104.

    PubMed  Google Scholar 

  65. Kothari SGT. Percutaneous permanent electrode implantation to ulnar nerves for upper extremity chronic pain: 6-year follow-up: 201. Reg Anesth Pain Med. 2006;31(5):16.

    Google Scholar 

  66. Bouche BEE, Meignier M, et al. Facilitation of diagnostic and percutaneous trial lead placement with ultrasound guidance for peripheral nerve stimulation ilioinguinal neuralgia. Abstracts from the 10th World Congress of the International Neuromodulation Society. Neuromodulation. 2011;14:563.

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sameer Jain.

Ethics declarations

Conflict of Interest

Sameer Jain declares that he has no conflict of interest. Timothy R. Deer is a consultant for Abbott, Axonics, Bioness, Flowonix, Jazz Pharm, Saluda Medical, Vertos, SpineThera, Nalu, and Vertiflex; is a member of the advisory board for Abbott, Flowonix, Jazz Pharm, Nalu, and Vertiflex; has equity options in Bioness, Vertiflex, Axonic, Vertos, SpineThera, Saluda Medical, Nalu, Cornerloc, and Flowonix; is a research consultant for Abbott, Mainstay Medical, Saluda, and Vertiflex; formerly had equity positions in Spinal Modulation, and Nevro; and has a patent pending for the DRG paddle lead with Abbott.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

This article is part of the Topical Collection on Pain Medicine

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jain, S., Deer, T.R. New Advances in Neuromodulation. Curr Anesthesiol Rep 8, 329–336 (2018). https://doi.org/10.1007/s40140-018-0298-x

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40140-018-0298-x

Keywords

Navigation