Skip to content
BY-NC-ND 4.0 license Open Access Published by De Gruyter Open Access October 17, 2019

Cancer- and non-cancer related chronic pain: from the physiopathological basics to management

  • Antonello Sica EMAIL logo , Beniamino Casale , Maria Teresa Di Dato , Armando Calogero , Alessandro Spada , Caterina Sagnelli , Mario Santagata , Pietro Buonavolontà , Alfonso Fiorelli , Anna Salzano , Concetta Anna Dodaro , Erika Martinelli , Elisabetta Saracco , Teresa Troiani , Dario Tammaro , Fortunato Ciardiello and Alfonso Papa
From the journal Open Medicine

Abstract

The prevalence of chronic pain is between 33% to 64% and is due to cancer pain, but it has also been observed in non-cancer patients. Chronic pain is associated with lower quality of life and higher psychological distress and depressive/anxiety disorders in patients without a history of disorder.

In this study we evaluated in clinical practice the effectiveness of the intrathecal pump in 140 patients who underwent pain therapy at our Center. These patients were consecutively enrolled from January 2010 to July 2018.

Follow-up was carried out over these eight years regarding the infusion modalities. Pain relief was obtained in 71 (50,7%) patients out of the 140 that experienced satisfactory pain control globally.

Intrathecal therapy is one of the best options for chronic severe refractory pain. The greatest advantage of this therapy is due to the possibility of treating the pain with minimal dosages of the drug, avoiding the appearance of troublesome side effects.

1 Introduction

Pain is a very complex physiopathological entity. It can take on different aspects both from a clinical and biological/ physiological point of view, and present itself as an epiphenomenon of a pathophysiological process, until it becomes a real pathological entity in its own right. Pain is defined as chronic when it is continuous for more than three tosix months [1]. It is a condition reported in 20% of people worldwide, in 15-20% of all physician examinations [2], and should receive more attention because a proper pain therapy is a human right [2,3]. The trigger for the development of chronic pain may be different in different situations. Inflammation causes inflammatory pain, while nerve injuries as the result of mechanical trauma (iatrogenic or not), metabolic or autoimmune disorders, and cancer and chemotherapy may give neuropathic pain [4, 5, 6]. The excitation of the primary neurons due to prolonged inflammation induce a pathological response that persist beyond the period of recovery of the tissue, constantly stimulating the nociceptive pathways and thus generating chronic pain with changes in ion channels, receptors and nerve synapses. The distribution of neurotransmitters and neuromediators allows peripheral and central neurons to reach the depolarization threshold early to cause ectopic discharges to amplify and activate nearby cells, with chronic pain [7]. Neuronal pathophysiological mechanisms are integrated with immunological response, and neuropathic pain is considered a neuro-immune disorder [8, 9, 10, 11, 12, 13]. In fact, patients with complex regional pain, peripheral neuropathy and neuropathic pain associated with spinal cord injury syndrome have increased serum IL-4, IL-6 and TNF-α, as well as reduced serum IL-10 levels [14, 15]. The serum increases of IL-1β, IL-6, IL-2, TNF-α, and IFN-γ increase the intensity of chronic pain [16, 17, 18, 19]. Therefore, inflammatory and allogenic processes are supported by a complex balance between cells and cytokines (due to both pro and anti-inflammatory molecules) and the nervous system.

IL-6, TNF-α and IL1-β, produced by macrophages, are cytokines that amplify chronic pain. According to some studies, the administration of TNF-α is associated with thermal hyperalgesia, mechanical allodynia and hypersensitivity associated with pain. In the electrophysiological field, TNF-α can also increase the conductivity of K+ ions of the membrane in a non-voltage dependent manner, resulting in overall neuronal hyperexcitability and therefore neuropathic pain. TNF-α induce the release of glial mediators TNF-α-induced glial mediators cause endocytosis of GABA receptors with consequent reduction of inhibitory modulation of the GABAergic system. Longterm potentiation (LTP) is a physiological mechanism for strengthening a neuronal circuit and is involved in numerous nerve functions. Intrathecal therapy (IT) is a good choice to improve therapeutic results in chronic pain.

The aim of this study was to evaluate in clinical practice the effectiveness of the intrathecal pump in 140 patients consecutively enrolled from January 2010 to July 2018 who underwent pain therapy at our Center.

2 Materials and methods

One hundred and forty patients who underwent pain therapy at our Center were consecutively enrolled from January 2010 to July 2018. All procedures conducted in the study were in accordance with international guidelines, with the standards of human experimentation of the local Ethics Committees and with the Helsinki Declaration of 1975, revised in 1983.

At the baseline visit, each patient signed their informed consent for the use of their data in clinical research.

All 140 patients underwent complete physical examination. Their pain was evaluated with the McGill Pain Questionnaire (MPQ), recording the Numerical Rating Scale (NRS) of average pain, minimum pain, maximum pain and pain during exertion (scale 0–10: 0 = no pain, 10 = worst pain ever). All patients were evaluated for HBV, and HCV serum markers. Serum samples were tested for HBsAg, anti-HCV, total anti-HBc, and anti-hepatitis B surface antibody (HBs) using commercial immunoenzymatic assays (Abbott Laboratories, North Chicago, IL, USA: AxSYM® HBsAg (v2) M/S for HBsAg, AxSYM® HCV (v3) for anti-HCV, AxSYM® CORE™ (v2) for total anti-HBc, and AxSYM® AUSAB® for anti-HBs), as described in previous studies [20, 21, 22, 23, 24, 25, 26, 27, 28].

Epidemiological, clinical, and immunological data recorded at the baseline visit included patients’ age and sex and risk factors for the acquisition of pain as stated by the patients.

All cases were followed-up in these 8 years with complete physical examinations, laboratory tests were carried out and pain was re-evaluated.

3 Results

The demographic and clinical data obtained at enrolment are shown in Table 1.

Table 1

Demographic and clinical characteristics of the 140 enrolled patients

N. of patients 140
Males, N. (%) 98 (70.0%)
Age, years (M + SD) 51.2 ± 8.2
Caucasian, N (%) 140 (100%)
Etiology of the pain, N(%): chronic cancer pain 99 (71.0%)
chronic non-cancer pain 41 (29.0%)
Therapy, N (%):
Morphine in primary infusion 120 (85.6%)
Ziconotide in primary infusion 3 (0.2%)
Baclofen in primary infusion 11 (0.8%)
Ziconotide plus morphine 3 (0.2%)
Bupivacaine plus morphine 3 (0.2%)

The 140 patients were predominantly male: 98 (70%), with a media age of 51.2 years old (range: 42-63). Of 140 patients, 99 cases had been subjected to a system for the treatment of chronic cancer pain; 41 patients were diagnosed with non-cancer pain. In non-cancer pain patients, the main diagnoses were neuropathic or mixed pain, chronic, not responsive to conventional drug therapy and / or burdened by the incidence of significantly elevated side effects with high doses. Some patients were not responders to spinal cord stimulation (Table 1).

Morphine was administered as the first intrathecal continuous infusion in 120 cases, in three patients ziconotide was used alone as the primary infusion, and 11 patients received primary Baclofen infusion alone. Ziconotide and Bupivacaine are analgesic adjuvants to morphine. In three patients, Ziconotide had been used in association with morphine as a secondary infusion, and three patients had a secondary infusion of Bupivacaine used in association with morphine. The patient was then given an intrathecal continuous infusion of one of the following: ziconotide 2.5 mL (2.5 μg at 1 μg per mL) daily, morphine 0.1-8 mg daily, baclofen 250-1000 μg daily or bupivacaine 3-8 mg daily.

From the follow-up carried out over these 8 years regarding the infusion modalities and the titration of the described drugs, the following data concerning pain relief was obtained: 71 (50,7%) patients out of 140 experienced satisfactory pain control globally, settling on a percentage of pain relief variable from 50 to 70%.

Only one patient, followed by oncologist, showed pain relief less than 30%.

In patients on follow-up with baclofen infusion for a diagnosis other than chronic pain, a variable reduction of the painful physiological symptomatology in spasticity was observed.

4 Discussion

Intrathecal therapy is a good choice to improve therapeutic results in patients with chronic pain. In fact, the morbidity and mortality of IT opioids are lower than those of systemic administration of opioids. A trial on 6398 patients in over a period of ten years [29], showed that there weren’t deaths correlated with the IT opioid infusion. In the US, death from systemically administered opioids are about 19,000 a year. Intrathecal drug delivery (IDD) is potentially a safer option for the patient [30].The Food and Drug Administration (FDA) has approved two intrathecal pain relief drugs: morphine free of preservatives and ziconotide peptide. Intrathecal opioid administration may cause side effects (vomiting, nausea, itching, constipation, urinary retention and neuroendocrine dysfunction) [31]. The most serious adverse reaction reported is respiratory depression [32], which occurs with the administration of opioid IT therapy or restarting of IT opioids after stopping [33]. The risk may be higher with hydrophilic opioids, such as morphine, and in cases where other depressants of the central nervous system (CNS), such as benzodiazepines, are administered [34]. The Polyanalgesic Consensus Conference (PACC) has recently recommended testing opioid therapy in an outpatient setting with a conservative dose, as a single dose of 0.15 mg morphine produces respiratory depression (defined as a PaCO2 above 40 or a lower respiratory rate at 10 breaths per minute) [35, 36, 37]. Ziconotide is a non-opioid drug approved for IT [38, 39, 40, 41], with adverse events of the CNS reported (nausea, nystagmus, vertigo, dysmetria, ataxia, agitation, hallucinations and coma) [42]. Neuropsychiatric adverse effects can occur after many months of asymptomatic infusion and are the main reason for its suspension [43]. Although an increase in serum creatinine kinase is present in 40% of patients, this significant increase typically occurs in the first two months after initiation of ziconotide therapy and only three cases of rhabdomyolysis have been described. There is no antidote for the overdose of ziconotide IT. The use of intrathecal bupivacaine (usually in combination with morphine) is off-label and it has occasionally been associated with numbness and rarely with weakness [44]. The PACC of 2016 presented evidence-based recommendations on patient survival, pathological process and use of IT drugs. Regarding cancer pain, IT is frequently thought to offer reliable, safe and effective treatment [45, 46, 47, 48, 49, 50].

Regarding the algorithm for IT management in patients with cancer pain, the prognosis, the probable disease progression in anatomical regions, features of tumors and findings of peri-procedural imaging are useful considerations for the selection of the device. Patients with cancer pain deserve special attention, in terms of implementation of IT and selection of drugs, together with the sustainability of a regime. Therefore, patients are stratified into 3 main categories by stage of disease and life expectancy. In the last PACC update, IT is used to treat related/unrelated cancer pain, and chronic-severe refractory pain [51]. The focus was on the patient’s age, although age contributions are reflected in the sustainability of the therapy, which is addressed elsewhere in the recommendations [52,53]. The costs of this therapy and its safety are better than the use of systemic opioids [54, 55].

The need to resort to the continuous infusion of subarachnoid drugs occurs when pharmacological, neurostimulation or surgical therapies have been unsuccessful. The main indications are: chronic non-cancer pain with a strong neuropathic component; chronic cancer pain with more than 6 months of life expectancy, and spasticity of degenerative and traumatic origin.

The procedure is completely reversible, the catheter can be removed if the expectations in terms of reduction / disappearance of the symptomatology are not met; the greatest advantage is the possibility of treating the symptoms (pain, spasticity) with minimal doses of effective drug, avoiding the appearance of troublesome side effects. Fixed-flow pumps have an indefinite duration, while programmable flow pumps are constrained to when the battery runs out, so they must be replaced before the battery expires.

The implant procedure involves positioning the catheter in the subarachnoid space, connecting it to the pump through a second catheter (extension) that is tunneled to the abdomen where the subcutaneous pocket containing the pump is packaged. Both the first and second phases of the operation are performed under local anesthesia, possibly paired with a mild analgosedation. In addition to the risks associated with the completion of a surgical procedure, the implants and / or the use of IT devices may also entail the following risks and complications: the displacement or disconnection of the catheter may occur; insertion of a catheter into the subarachnoid space may result in epidural bleeding, headache, hematoma, infection, spinal cord compression and/ or paresis; loss of cerebrospinal fluid (fluid that bathes nerve structures), with possible headache; persistent pain at the site of implantation of the catheter or pump; serum collection at the pump site just under the skin, which can be evacuated with a simple puncture; catheter migration, which can cause changes in the analgesic effect; allergic reaction or rejection of implanted materials;and pain localized at the implant site.

Our study demonstrates the effectiveness of intrathecal therapy in reducing severe chronic refractory pain compared to intravenous (i.v.) therapies. The importance of specialized staff dedicated to these procedures can drastically reduce adverse events and complications. It is a safe and manageable solution in pain management and can be considered for chronic refractory pain from all sources. Our data shows a longer follow up and case study than the others in the literature [56, 57, 58, 59, 60, 61, 62] and this reinforces our obtained results.

5 Conclusion

Chronic pain is correlated with lower quality of life and depressive/anxiety disorders (15.5%) in patients without a history of these disorders. Depression and anxiety are associated with more severe pain.

Intrathecal therapy is one of the best options in the treatment of chronic/severe refractory pain. The costs associated with therapy in terms of safety are significantly better than those of systemic opioids.

The greatest advantage of this therapy is due to the possibility of treating the pain with minimal dosages of the drug, avoiding the appearance of troublesome side effects. Non-responsive patients represent the ideal category to be addressed with “niche” techniques such as cordotomy.

Our results confirm that intrathecal therapy is one of the best choices in the management and treatment of severe chronic refractory pain. This therapy, overall, is safer than systemic opioids, which often need higher dosages to be effective. With systemic opioid therapy, the higher dosages and possible interferences with other drugs used in this type of complex patient allow an increase in the possibility of serious adverse events. Intrathecal therapy has demonstrated superiority in the management of dosages of these drugs which can be reduced to the minimum effective with this technique.

The use of minimum effective dosages of opioids in pain therapy allows a reduction in serious adverse events, and for this reason also a considerable reduction in the overall costs of treatment of chronic patients.

The need to resort to the continuous infusion of subarachnoid drugs occurs when pharmacological, neurostimulation or surgical therapies have been unsuccessful. The catheter can be removed calmly, if the expectations in terms of symptomatic reduction are not met.


Tel: +393332253315, +3908119573375

  1. Conflict of interest

    Conflict-of-interest statement: All the authors of the manuscript declare that they have no conflicts of interest in connection with this paper.

References

[1] Merskey H, Bogduk N. Classification of chronic pain. 2nd ed. Seattle: IASP Press, 1994. p. 1Search in Google Scholar

[2] Treede RD, Rief W, Barke A, Aziz Q, Bennett MI, Benoliel R, et al. A classification of chronic pain for ICD-11. Pain. 2015;156(6):1003-7. doi: 10.1097/j. pain.0000000000000160.10.1097/j.pain.0000000000000160Search in Google Scholar PubMed PubMed Central

[3] Breivik H, Collett B, Ventafridda V, Cohen R, Gallacher D. Survey of chronic pain in Europe: prevalence, impact on daily life, and treatment. Eur J Pain 2006;10:287.10.1016/j.ejpain.2005.06.009Search in Google Scholar PubMed

[4] Vellucci R. Heterogeneity of chronic pain. Clin Drug Investig 2012;32(Suppl 1):3–10.10.2165/11630030-000000000-00000Search in Google Scholar PubMed

[5] Calogero A, Sagnelli C, Carlomagno N, Tammaro V, Candida M, Vernillo A, et al. Familial polyposis coli: the management of desmoid tumors bleeding. Open Med (Wars). 2019;14:572-576. doi: 10.1515/med-2019-0064.10.1515/med-2019-0064Search in Google Scholar PubMed PubMed Central

[6] Sica A, Vitiello P, Papa A, Sagnelli C, Calogero A, Casale D, et al. Use of rituximab in NHL MALT type affected pregnant woman, during the first trimester for two times. Open Medicine. 2019 in press.10.1515/med-2019-0087Search in Google Scholar PubMed PubMed Central

[7] de Miguel M, Kraychete DC, Meyer Nascimento RJ. Chronic pain: cytokines, lymphocytes and chemokines. Inflamm Allergy Drug Targets. 2014;13(5):339-49.10.2174/1871528114666150114170004Search in Google Scholar

[8] Scholz J; Woolf CJ. The neuropathic pain triad: neurons, immune cells and glia. Nature Neurosci. 2007;10(11):1361-1368.10.1038/nn1992Search in Google Scholar PubMed

[9] Zhang JM, An J. Cytokines, inflammation, and pain. Int Anesthesiol Clin 2007;45:27–37.10.1097/AIA.0b013e318034194eSearch in Google Scholar PubMed PubMed Central

[10] Basbaum AI, Bautista DM, Scherrer G, Julius D. Cellular and molecular mechanisms of pain. Cell 2009;139:267–84.10.1016/j.cell.2009.09.028Search in Google Scholar PubMed PubMed Central

[11] Hu P, Bembrick AL, Keay KA, McLachlan EM. Immune cell involvement in dorsal root ganglia and spinal cord after chronic constriction or transection of the rat sciatic nerve. Brain Behav Immun 2007;21:599–616.10.1016/j.bbi.2006.10.013Search in Google Scholar PubMed

[12] Kobayashi Y, Kiguchi N, Fukazawa Y, et al. Macrophage-T cell interactions mediate neuropathic pain through the glucocorticoid-induced tumor necrosis factor ligand system. J Biol Chem 2015;290:12603–13.10.1074/jbc.M115.636506Search in Google Scholar PubMed PubMed Central

[13] Uçeyler N, Rogausch JP, Toyka KV, Sommer C. Differential expression of cytokines in painful and painless neuropathies. Neurology. 2007;69(1):42-9.10.1212/01.wnl.0000265062.92340.a5Search in Google Scholar PubMed

[14] Uçeyler N, Eberle T, Rolke R, Birklein F, Sommer C. Differential expression patterns of cytokines in complex regional pain syndrome. Pain. 2007;132(1-2):195-205.10.1016/j.pain.2007.07.031Search in Google Scholar PubMed

[15] Koch A, Zacharowski K, Boehm O, Stevens M, Lipfert P, von Giesen HJ, et al. Nitric oxide and pro-inflammatory cytokines correlate with pain intensity in chronic pain patients. Inflamm Res. 2007;56(1):32-7.10.1007/s00011-007-6088-4Search in Google Scholar PubMed

[16] Sandkühler J, Gruber-Schoffnegger D. Hyperalgesia by synaptic long-term potentiation (LTP): an update. Curr. Opin. Pharmacol. 2012;12(1):18-27.10.1016/j.coph.2011.10.018Search in Google Scholar PubMed PubMed Central

[17] Gruber-Schoffnegger D, Drdla-Schutting R, Hönigsperger C, Wunderbaldinger G, Gassner M; Sandkühler J. Induction of thermal hyperalgesia and synaptic long-term potentiation in the spinal cord lamina I by TNF-α and IL-1β is mediated by glial cells. J. Neurosci. 2013;33(15):6540-6551.10.1523/JNEUROSCI.5087-12.2013Search in Google Scholar PubMed PubMed Central

[18] Thacker MA, Clark AK, Marchand F, McMahon SB. Pathophysiology of peripheral neuropathic pain: immune cells and molecules. Anesth Analg. 2007;105(3):838-47.10.1213/01.ane.0000275190.42912.37Search in Google Scholar PubMed

[19] Watkins LR, Maier SF. Glia: a novel drug discovery target for clinical pain. Nat. Rev. Drug Discov. 2003;2(12):973-985.10.1038/nrd1251Search in Google Scholar PubMed

[20] Pisaturo M, Guastafierro S, Filippini P, Tonziello G, Sica A, Di Martino F, et al. Absence of occult HCV infection in patients experiencing an immunodepression condition. Infez Med. 2013;21(4):296-301.Search in Google Scholar

[21] Sagnelli C, Pisaturo M, Calò F, Martini S, Sagnelli E. Reactivation of HBV infection in patients with hemo-lymphoproliferative diseases, and its prevention. World Journal of Gastroenterology 2019;25(26):3299-3312. doi: 10.3748/wjg. v25.i26.3299.10.3748/wjg.v25.i26.3299Search in Google Scholar PubMed PubMed Central

[22] Tonziello G, Pisaturo M, Sica A, Ferrara MG, Sagnelli C, Pasquale G, et al. Transient reactivation of occult hepatitis B virus infection despite lamivudine prophylaxis in a patient treated for non-Hodgkin lymphoma. Infection. 2013;41(1):225-9. doi: 10.1007/s15010-012-0305-y.10.1007/s15010-012-0305-ySearch in Google Scholar PubMed

[23] Sagnelli C, Sagnelli E. Towards the worldwide eradication of HBV infection; A combination of prophylactic and therapeutic factors. World Journal of Clinical Infectious Diseases 2019.10.5495/wjcid.v9.i2.11Search in Google Scholar

[24] Coppola N, Pisaturo M, Guastafierro S, Tonziello G, Sica A, Iodice V, et al. Increased hepatitis C viral load and reactivation of liver disease in HCV RNA-positive patients with onco-haematological disease undergoing chemotherapy. Dig Liver Dis. 2012;44(1):49-54. doi: 10.1016/j.dld.2011.07.016.10.1016/j.dld.2011.07.016Search in Google Scholar PubMed

[25] Coppola N, Pisaturo M, Guastafierro S, Tonziello G, Sica A, Sagnelli C, et al. Absence of occult hepatitis C virus infection in patients under immunosupressive therapy for oncohematological diseases.Hepatology. 2011;54(4):1487-9. doi: 10.1002/hep.24436.10.1002/hep.24436Search in Google Scholar PubMed

[26] Merli M, Frigeni M, Alric L, Visco C, Besson C, Mannelli L, et al. Direct-Acting Antivirals in Hepatitis C Virus-Associated Diffuse Large B-cell Lymphomas. Oncologist. 2018. pii: theoncologist.2018-0331. doi: 10.1634/theoncologist.2018-0331.10.1634/theoncologist.2018-0331Search in Google Scholar PubMed PubMed Central

[27] Calogero A, Sagnelli E, Creta M, Angeletti S, Peluso G, Incollingo P, et al. Eradication of HCV infection with the Direct-Acting Antiviral Therapy in renal allograft recipients. BioMed Research International. Vol. 2019, Article ID 4674560, 8 pages https://doi.org/10.1155/2019/4674560 Erratum to “Eradication of HCV Infection with the Direct-Acting Antiviral Therapy in Renal Allograft Recipients”,” BioMed Research International, vol. 2019, Article ID 8797329, 1 pages, 2019. https://doi.org/10.1155/2019/8797329https://doi.org/10.1155/2019/8797329Search in Google Scholar PubMed PubMed Central

[28] Sagnelli C, Uberti-Foppa C, Hasson H, Bellini G, Minichini C, Salpietro S, et al. IN VIVO evidence that the cannabinoid receptor 2-63-RR variant is associated with the acquisition and/or expansion of HIV infection. HIV Medicine 2018;19(9):597-604. doi: 10.1111/hiv.1263810.1111/hiv.12638Search in Google Scholar PubMed

[29] Medtronic. 2015 Product Performance Report. Targeted Drug Delivery Systems. http://professional.medtronic.com/ppr/intrathecal-drug-delivery-systems/index.htm#.WAY_sySkyzwSearch in Google Scholar

[30] Center for Disease Control and Prevention, National Center for Health Statistics, National Vital Statistics System, Mortality File. (2015). Number and Age-Adjusted Rates of Drug-poisoning Deaths Involving Opioid Analgesics and Heroin: United States, 2000–2014. Atlanta, GA: Center for Disease Control and Prevention. http://www.cdc.gov/nchs/data/health_policy/AADR_drug_poisoning_involving_OA_Heroin_US_2000-2014.pdfSearch in Google Scholar

[31] Ruan X. Drug-related side effects of long-term intrathecal morphine therapy. Pain Physician 2007;10:357–366.10.36076/ppj.2007/10/357Search in Google Scholar

[32] Wallace MS, Rauck R, Fisher R, Charapata SG, Ellis D, Dissanayake S. Intrathecal ziconotide for severe chronic pain: safety and tolerability results of an open-label, long-term trial. Anesth Analg 2008;106:628–637.10.1213/ane.0b013e3181606fadSearch in Google Scholar PubMed

[33] Coffey RJ, Owens ML, Broste SK, Dubois MY, Ferrante FM, Schultz DM, et al. Medical practice perspective: identification and mitigation of risk factors for mortality associated with intrathecal opioids for non-cancer pain. Pain Med. 2010;11(7):1001-9. doi: 10.1111/j.1526-4637.2010.00889.x.10.1111/j.1526-4637.2010.00889.xSearch in Google Scholar PubMed

[34] Prager J, Deer T, Levy R, Bruel B, Buchser E, Caraway D, et al. Best practices for intrathecal drug delivery for pain. Neuromodulation. 2014;17(4):354-72; discussion 372. doi: 10.1111/ner.1214610.1111/ner.12146Search in Google Scholar PubMed

[35] Deer TR, Pope J, Hayek SM. The Polyanalgesic Consensus Conference (PACC) recommendations on intrathecal drug infusion systems best practices and guidelines. Neuromodulation 2017;20:96–132.10.1111/ner.12538Search in Google Scholar PubMed

[36] Deer TR, Hayek SM, Pope JE, Lamer TJ, Hamza M, Grider JS, et al. The Polyanalgesic Consensus Conference (PACC): recommendations for trialing of intrathecal drug delivery infusion therapy. Neuromodulation 2017;20:133–154.10.1111/ner.12543Search in Google Scholar

[37] Pope JE, Deer TR, Amirdelfan K, McRoberts WP, Azeem N. The pharmacology of spinal opioids and ziconotide for the treatment of non-cancer pain. Curr Neuropharmacol. 2017;15(2):206-21610.2174/1570159X14666160210142339Search in Google Scholar

[38] Pope JE, Deer TR. Ziconotide: a clinical update and pharmacologic review. Expert Opin Pharmacother 2013;14:957–966.10.1517/14656566.2013.784269Search in Google Scholar

[39] Rauck R, Webster L, Wallace M et al. Effect of concomitant antidepressant and anticonvulsant use on adverse events in patients receiving intrathecal ziconotide in a long-term extension study. Poster presented a WVSIPP, Puerto Rico, 2014.Search in Google Scholar

[40] Ziconotide: new drug. Limited analgesic efficacy, too many adverse effects. Prescrire Int 2008;17:179–182.Search in Google Scholar

[41] Schmidtko A, Lotsch J, Freynhagen R, Geisslinger G. Ziconotide for treatment of severe chronic pain. Lancet 2010;375:1569–1577.10.1016/S0140-6736(10)60354-6Search in Google Scholar

[42] Penn RD, Paice JA. Adverse effects associated with the intrathecal administration of ziconotide. Pain 2000;85:291–296.10.1016/S0304-3959(99)00254-7Search in Google Scholar

[43] Hayek SM, Hanes MC, Wang C, Veizi IE. Ziconotide combination intrathecal therapy for noncancer pain is limited secondary to delayed adverse effects: a case series with a 24-month follow-up. Neuromodulation 2015;18:397–403.10.1111/ner.12270Search in Google Scholar PubMed

[44] Goucke CR, Dusci LJ, Van Leeuwen S, Fairclough D, Ilett KF. Stability and tolerability of high concentrations of intrathecal bupivacaine and opioid mixtures in chronic noncancer pain: an open-label pilot study. Pain Med 2010;11:1612–1618.10.1111/j.1526-4637.2010.00958.xSearch in Google Scholar PubMed

[45] Hayek SM, Deer TR, Pope JE, Panchal S, Patel V, Burton AW. Intrathecal therapy for cancer and non-cancer pain. Pain Physician 2011;14:219–248.10.36076/ppj.2011/14/219Search in Google Scholar

[46] Falco F, Patel VB, Hayek SM, Deer TR, Geffert S, Zhu J, et al. Intrathecal infusion systems for long-term management of chronic cancer pain: an update of assessment of evidence. Pain Physician 2013;16:SE185–SE216.10.36076/ppj.2013/16/SE185Search in Google Scholar

[47] Smith TJ, Staats PS, Deer T, Stearns LJ, Rauck RL, Boortz-Marx RL, et al. Randomized clinical trial of an implantable drug delivery system compared with comprehensive medical management for refractory cancer pain: impact on pain, drug-related toxicity, and survival. J Clin Oncol 2002;20:4040–4049.10.1200/JCO.2002.02.118Search in Google Scholar PubMed

[48] Staats PS, Yearwood T, Charapata SG, Presley RW, Wallace MS, Byas-Smith M, et al. Intrathecal ziconotide in the treatment of refractory pain in patients with cancer or AIDS: a randomized controlled trial. JAMA 2004;291:63–70.10.1001/jama.291.1.63Search in Google Scholar PubMed

[49] Lindblom U, Meyerson BA. Influence on touch, vibration and cutaneous pain of dorsal column stimulation in man. Pain 1975;1:257–270.10.1016/0304-3959(75)90042-1Search in Google Scholar

[50] Kloth D. President’s Message. NANS Newsletter 2015;11:1–2.Search in Google Scholar

[51] Deer TR, Pope JE, Hayek SM, Lamer TJ, Veizi IE, Erdek M, et al. The Polyanalgesic Consensus Conference (PACC): Recommendations for Intrathecal Drug Delivery: Guidance for Improving Safety and Mitigating Risks. Neuromodulation. 2017;20(2):155-176. doi: 10.1111/ner.12579.10.1111/ner.12579Search in Google Scholar PubMed

[52] Saulino M, Stearns L, (moderators). Intrathecal Therapies: Basic Challenges. Concurrent session presented at: North American Neuromodulation Society 19th Annual Meeting; Dec. 2015; Las Vegas, NV.Search in Google Scholar

[53] Deer TR, Prager J, Levy R, Rathmell J, Buchser E, Burton A, et al. Polyanalgesic Consensus Conference 2012: recommendations for the management of pain by intrathecal (intraspinal) drug delivery: report of an interdisciplinary expert panel. Neuromodulation. 2012;15(5):436-64; discussion 464-6. doi: 10.1111/j.1525-1403.2012.00476.x.10.1111/j.1525-1403.2012.00476.xSearch in Google Scholar PubMed

[54] Pope J. Presentation at the 9th Annual Scientific Meeting of the Canadian Neuromodulation Society. September 30–October 2, 2016; Regina, Saskatchewan, Canada.Search in Google Scholar

[55] Pope JE, Deer TR. Intrathecal pharmacology update: novel dosing strategy for intrathecal monotherapy ziconotide on efficacy and sustainability. Neuromodulation 2015;18:414–420.10.1111/ner.12274Search in Google Scholar PubMed

[56] van den Beuken-van Everdingen M, de Rijke J, Kessels A, Schouten H, van Kleef M, Patijn J. Prevalence of pain in patients with cancer: a systematic review of the past 40 years. Annals of Oncology. 2007;18(9):1437–1449. doi: 10.1093/annonc/mdm05610.1093/annonc/mdm056Search in Google Scholar PubMed

[57] Inoue S, Kobayashi F, Nishihara M, Arai YC, Ikemoto T, Kawai T, et al. Chronic pain in the Japanese community— prevalence, characteristics and impact on quality of life. PLoS One. 2015;10(6) doi: 10.1371/journal.pone.0129262.e012926210.1371/journal.pone.0129262.e0129262Search in Google Scholar

[58] Tsatali M, Papaliagkas V, Damigos D, Mavreas V, Gouva M, Tsolaki M. Depression and anxiety levels increase chronic musculoskeletal pain in patients with Alzheimer’s disease. Current Alzheimer Research. 2014;11(6):574–579. doi: 10.2174/1567205011666140618103406.10.2174/1567205011666140618103406Search in Google Scholar PubMed

[59] Kaczocha M, Azim S, Nicholson J, Rebecchi MJ, Lu Y, Feng T, et al. Intrathecal morphine administration reduces postoperative pain and peripheral endocannabinoid levels in total knee arthroplasty patients: a randomized clinical trial. BMC Anesthesiol. 2018;18(1):27. doi: 10.1186/s12871-018-0489-5.10.1186/s12871-018-0489-5Search in Google Scholar PubMed PubMed Central

[60] Reif I, Wincent A, Stiller CO. Intrathecal analgesia by bupivacaine is not enhanced by coadministration of morphine in patients with severe cancer-related pain: a randomized double-blind cross-over study.Int J Clin Pharmacol Ther. 2017;55(6):525-532. doi: 10.5414/CP202955.10.5414/CP202955Search in Google Scholar PubMed

[61] Slavković Z, Stamenković DM, Gerić V, Veljović M, Ivanoviić N, Tomić A, et al. Comparison of analgesic effect of intrathecal morphine alone or in combination with bupivacaine and fentanyl in patients undergoing total gastrectomy: a prospective randomized, double blind clinical trial. Vojnosanit Pregl. 2013;70(6):541-7.10.2298/VSP111011011SSearch in Google Scholar

[62] Kuchálik J, Granath B, Ljunggren A, Magnuson A, Lundin A, Gupta A. Postoperative pain relief after total hip arthroplasty: a randomized, double-blind comparison between intrathecal morphine and local infiltration analgesia. Br J Anaesth. 2013;111(5):793-9. doi: 10.1093/bja/aet248.10.1093/bja/aet248Search in Google Scholar PubMed

Received: 2019-05-13
Accepted: 2019-08-16
Published Online: 2019-10-17

© 2019 Antonello Sica et al. published by De Gruyter

This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.

Downloaded on 19.5.2024 from https://www.degruyter.com/document/doi/10.1515/med-2019-0088/html
Scroll to top button