IJGII Inernational Journal of Gastrointestinal Intervention

pISSN 2636-0004 eISSN 2636-0012
ESCI
scopus

Article

home All Articles View

Review Article

Int J Gastrointest Interv 2022; 11(3): 126-131

Published online July 31, 2022 https://doi.org/10.18528/ijgii220026

Copyright © International Journal of Gastrointestinal Intervention.

Endoscopic ultrasound-guided celiac plexus neurolysis for managing abdominal pain related with advanced cancer

Ryota Nakano , Hideyuki Shiomi* , Shogo Ota , and Hiroko Iijima

Division of Gastroenterology and Hepato-Biliary-Pancreatology, Department of Internal Medicine, Hyogo Medical University, Nishinomiya, Japan

Correspondence to:*Division of Gastroenterology and Hepato-Biliary-Pancreatology, Department of Internal Medicine, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya 663-8501, Japan.
E-mail address: hideshio0403@gmail.com (H. Shiomi).

Received: June 6, 2022; Revised: July 12, 2022; Accepted: July 20, 2022

This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/bync/4.0) which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Cancer associated abdominal pain, as typified by pancreatic cancer, has conventionally been treated with narcotic drugs, but often the severe abdominal pain is difficult to control. Endoscopic ultrasound-guided celiac plexus neurolysis (EUS-CPN) is commonly performed for abdominal pain that is difficult to control. EUS-CPN can be performed more safely and reliably than other conventional procedures on the celiac plexus, and good outcomes of pain relief have been reported. Although a variety of endoscopic techniques are available for EUS-CPN, the choice varies among institutions, and evidence on the efficacy and safety of each procedure is limited. In this review, we summarize the indications for treatment, specific endoscopic techniques, therapeutic efficacy for pain relief, and complications from previous reports on EUS-CPN.

Keywords: Chronic pancreatitis, Endoscopic ultrasound-guided celiac ganglia neurolysis, Endoscopic ultrasound-guided celiac plexus neurolysis, Pancreatic cancer

Cancer pain associated with advanced cancer of the abdominal organs, such as unresectable pancreatic cancer, can often be difficult to control, significantly reducing the patient’s quality of life and limiting daily activities.1,2 Thus, it is obvious that adequate pain control is an extremely important issue. Pancreatic cancer remains one of the carcinomas with the poorest prognosis, with a 5-year survival rate of less than 10%.3 Most pancreatic cancers are detected at an advanced stage and are therefore unresectable.4 In pancreatic cancer, it has been reported that approximately 60% to 80% of patients suffer from abdominal pain due to plexus invasion of the tumor.2 Most pancreatic cancer patients require continuing chemotherapy, but their treatment is often interrupted by uncontrollable cancer-related pain. Traditionally, drug therapy centered on non-steroidal anti-inflammatory drugs (NSAIDS), such as loxoprofen, and narcotic analgesics, such as morphine, have been widely used to treat cancer pain associated with abdominal malignancies. However, especially in unresectable pancreatic cancer, severe tumor pain frequently occurs, and pain control with pharmacotherapy is often inadequate. When neoplastic abdominal pain treatment by NSAIDS or opioid analgesics fail, a celiac plexus neurolysis (CPN) can be used. CPN is guided by fluoroscopy, computed tomography (CT) or echo, and their usefulness has been reported.58

The usefulness of CPN using various modalities has been reported in many cases, with effective therapeutic response in more than 80% of patients.5,6 Pain originating in intraperitoneal organs such as the pancreas is transmitted by afferent nerve fibers through the celiac plexus and reaches the central nervous system through the dorsal root of the spinal cord at the T12-L2 level. The celiac plexus is a group of nerve fibers that converge on the celiac ganglion in the retroperitoneum, immediately adjacent to the anterior external wall of the aorta at the origin of the celiac trunk. CPNs were commonly performed percutaneously under echo-guided or CT-guided techniques. However, these treatments are invasive as they puncture multiple obstacles between the ventral or dorsal skin and the celiac plexus and carry the risk of various complications. Recently, endoscopic ultrasound-guided CPN (EUS-CPN) has been performed and reported as safe and effective.9 EUS-guided method allows for a safer approach from a site closer to the celiac plexus. However, evidence on the effectiveness and safety of EUS-CPN is limited, and the details of endoscopic technique often vary among institutions. This review will summarize previous reports on the therapeutic efficacy and complications of EUS-CPN, as well as specific endoscopic techniques.

The indication for a CPN is visceral pain originating in the upper abdominal organs. This includes the liver, biliary tract, and gastrointestinal tract, though it is most often performed for pain originating from pancreatic cancer.

Good indications for EUS-CPN are cases that are difficult to control with NSAIDS or opioid analgesics, or cases in which the side effects of narcotic analgesics make discontinuation of medication or reduce quality of life. While cases poorly controlled with medication are considered good indications for EUS-CPN, it has also been suggested that performing EUS-CPN early in the diagnosis may lead to better analgesia and reduced opioid consumption,10 and further studies are expected in the future.

On the other hand, the efficacy of EUS-CPN for upper abdominal pain related to chronic pancreatitis has also been reported and may be indicated not only for malignant tumors such as pancreatic cancer but also for chronic pancreatitis.11

Before performing EUS-CPN, contrast-enhanced CT or other imaging modality should be performed to evaluate the site of the lesion, assess involvement of the celiac plexus, and confirm vascular anatomy. It is also necessary to rule out other causes of upper abdominal pain besides tumor pain, such as acute cholangitis, cholecystitis, gastrointestinal ulcers, and ileus. In assessing the safety of the puncture, it is important to check the patient’s medical history and to confirm the risk of bleeding tendency. Most reports indicate that EUS-CPN is contraindicated in cases of impaired coagulation (international normalized ratio ≥ 1.6), thrombocytopenia (< 50,000/L), and patients with gastric and esophageal varices.12,13

Neurolytic agent

Generally, 98% ethanol is used as a neurolytic agent in EUS-CPN, although phenol is sometimes used.14 However, alcohol injection alone may cause postoperative pain due to perineural swelling and tissue damage. To reduce post-procedural pain and discomfort, local anesthetics such as bupivacaine are used.15 As a local anesthetic, 0.25% bupivacaine is most commonly used, although 0.75% bupivacaine, lidocaine and ropivacaine have also been used.1517 In a multicenter, retrospective study of 150 patients who underwent EUS-CPN, Zhao et al16 reported that 0.75% bupivacaine, as a local anesthetic, when compared with 0.375% bupivacaine and 0.5% ropivacaine, significantly reduced pain within three days after EUS-CPN. In practice, drug selection and its concentration are often determined by the endoscopist and the amount of alcohol injected as a neurolytic agent varies by reports, but 10 to 20 mL is typically used.16 Within EUS-CPN, the use of neurolytic agents is described as “EUS-CPN” in the narrow sense, while the use of only local anesthetics or steroids without neurolytic agents is sometimes described as “abdominal plexus block (EUS-celiac plexus block).”

Central and bilateral techniques

EUS-CPN is performed by a convex type EUS scope. By scanning just under the esophagogastric junction, the long axis image of the abdominal aorta and the branches of the celiac artery and superior mesenteric artery can be easily visualized (Fig. 1A). The puncture line can be clearly delineated, and the puncture can be performed while confirming intervening organs and blood vessels, making it a safer and more accurate procedure than percutaneous echo-guided or CT-guided puncture. There are two main techniques for EUS-CPN: the central technique and the bilateral technique. In the central technique, a longitudinal section of the abdominal aorta is drawn from the posterior wall of the upper gastric body to identify the branches of the celiac artery. The puncture needle is advanced to the trunk of the celiac artery branch, and the agent is injected until the high-echoic area expands (Fig. 1B). In the bilateral technique, after drawing the celiac artery branch from the longitudinal section of the aorta, the scope is rotated slightly clockwise or counterclockwise to inject the drug into both sides of the celiac artery branch trunk. Usually, the average injection volume of local anesthetic is 2 to 10 mL, followed by 10 to 20 mL of alcohol.

Figure 1. (A) Convex endoscopic ultrasonography depiction of the celiac plexus and puncture sites for the central and broad techniques. (B) Practical use of the central technique. The puncture needle is advanced to the trunk of the CA branch, and the agent is injected until the high-echoic area expands. CA, celiac artery; SMA, superior mesenteric artery; Ao, aorta.

Efficacy of EUS-CPN

Efficacy of EUS-CPN for pancreatic cancer

The efficacy of EUS-CPN in reducing pain has been reported to be 45%–85%, although it varies from report to report.1826 Details from previous reports on the treatment efficacy and complications of EUS-CPN are shown in Table 1. In a meta-analysis by Puli et al20 the therapeutic efficacy of EUS-CPN was reported to be 80.1% in patients with pancreatic cancer and 59.4% in patients with chronic pancreatitis and another meta-analysis by Asif et al21 reported that the efficacy of EUS-CPN in patients with pancreatic cancer was 71%. Although EUS-CPN is most commonly performed when opioid medications are ineffective, early EUS-CPN intervention has been indicated to provide better analgesia and lesser opioid consumption. Wyse et al10 reported that in a randomized trial, the early EUS-CPN group showed significant pain reduction and a trend toward lower morphine use after 3 months of treatment. On the other hand, some reports show no difference in pain relief between EUS-CPN and opioid-based medication. Kanno et al27 conducted a randomized control study of EUS-CPN versus medication alone and found no significant differences in treatment efficacy or the amount of opioid use, concluding that EUS-CPN is an effective treatment option but should not be performed routinely.

Table 1 . Characteristics of the Studies on Endoscopic Ultrasound-Guided Celiac Plexus Neurolysis.

ReferenceYearDesignNumberTechniquePain relief
(follow-up time)
Complication
Wiersema and Wiersema91996Retrospective30Bilateral88 (10 week)Diarrhea 13.3%
Gunaratnam et al262001Prospective58Bilateral78 (24 week)Pain 8.6%
Wyse et al102011RCT48Bilateral vs medicationNANone
Sahai et al352009Prospective160Central vs Bilateral45% vs 70% (1 week)Bleeding 0.7%
Iwata et al132011Retrospective47Central/Bilateral68% (1 week)Hypotension 17%, diarrhea 23%, inebriation 8%
Wiechowska-Kozłowska et al232012Retrospective29Central/Bilateral86%Hypotension 3.4%, severe pain 6.8%, diarrhea 10.2%
Téllez-Ávila et al122013Retrospective53Central vs Bilateral50% vs 60% (4 week)Transitory pain 3%
Seicean et al222013Retrospective32Central75% (2 week)None
Facciorusso et al452020Retrospective215Central86%24%–44%
Diarrhea, fever, abdominal pain, pancreatitis
Kanno et al272020RCT48Central, Bilateral, Broad vs medicationNANA
Han et al182021Retrospective58Central and bilateral74% (1 week)
67% (4 week)
Hypotension 1.7%, pain 5.2%, transient loose stools 3.4%

RCT, randomized controlled trials; NA, not available..



Efficacy of EUS-CPN for chronic pancreatitis

While EUS-CPN is highly effective in treating tumor-related pain in patients with pancreatic cancer, the treatment efficacy of EUS-CPN for abdominal pain caused by chronic pancreatitis is limited.11,2830 A meta-analysis by Kaufman et al31 found the efficacy of EUS-CPN in reducing pain was 72.4% for pancreatic cancer compared to 51.4% for chronic pancreatitis. Based on many reports of the poor treatment efficacy of EUS-CPN for chronic pancreatitis, Teoh et al32 suggest that EUS-CPN is not recommended for chronic pancreatitis in the consensus guidelines.

Efficacy of central vs bilateral technique

Several articles have compared the difference in pain reduction between the two techniques in randomized prospective trials. Many studies have reported no difference in pain reduction or safety between central and bilateral techniques. Bhatnagar et al33 reported that in a randomized trial, the bilateral and central technique were comparable in pain relief and side effects. Téllez-Ávila et al12 compared the central and bilateral technique of EUS-CPN and reported that the median pain reduction after 4 weeks of treatment had no significant difference between them. In a prospective randomized study, LeBlanc et al34 randomized EUS-CPN to central versus bilateral methods and reported that pain relief time, duration of pain relief, and complications were not significantly different between the two groups. On the other hand, some reported that bilateral technique was significantly more effective in reducing pain than central technique. Sahai et al35 compared the central and bilateral technique in 184 patients undergoing EUS-CPN and found that the reduction in visual analogue scale (VAS) score at day 7 was significantly higher in the bilateral group (mean pain reduction 70.4% [61.0%–80.0%] vs 45.9% [32.7%–57.4%]; P = 0.0016), and they reported that the bilateral method was the only predictor of > 50% pain reduction. However, they also reported that adrenal artery bleeding was observed with the bilateral technique.

EUS-guided celiac ganglia neurolysis

In 2006, it became clear that the celiac ganglion could be directly delineated by EUS.36,37

EUS-guided celiac ganglia neurolysis (EUS-CGN), in which the celiac ganglia are directly punctured and blocked under EUS guided technique, was reported in 2008.38 EUS-CGN is a technique for injecting a drug through a direct puncture into the visualized celiac ganglion. The celiac ganglia are commonly found at a height between the aorta and left adrenal gland and between the celiac artery and left renal artery by rotating the scope clockwise from the trunk of the celiac artery.17 The celiac ganglion is often visualized as a hypoechoic nodular structure (Fig. 2).

Figure 2. The celiac ganglia are commonly found at a height between the Ao and left adrenal gland and between the CA and left renal artery by rotating the scope clockwise from the trunk of the CA. The celiac ganglion is visualized as a hypoechoic nodular structure. CA, celiac artery; Ao, aorta.

Efficacy of EUS-CGN

Based on these findings, EUS-CPN is a safe treatment technique that greatly reduces pain associated with pancreatic cancer. However, a certain number of cases with inadequate response to treatment must be taken into consideration. Levy et al38 reported the first use of EUS-CGN in patients with pancreatic cancer and obtained pain relief in 94% (16/17) of them. In some comparative studies with EUS-CPN, CGN has been reported to provide pain relief to 45%–80% of patients and many reports indicate that CGN was significantly more effective in reducing pain than EUS-CPN.1,17,24,38,39 Details from previous reports on the treatment efficacy and complications of EUS-CGN are shown in Table 2.

Table 2 . Characteristics of the Studies on EUS-CGN and EUS-BPN.

ReferenceYearDesignNumberTechniquePain relief
(follow-up time)
Complication
Levy et al382008Retrospective17CGN94% (4 week)Diarrhea 16%, hypotension 35%, transient pain 41%
Ascunce et al12011Retrospective64CGN vs CPN65% vs 25% (1 week)Diarrhea 23%, hypotension 2%, transient pain 2%
Doi et al172013RCT68CGN vs CPN73% vs 45% (1 week)Diarrhea 5.9% vs 9.1%, hypotension 2.9% vs 6%, pain 29.4% vs 21.2%
Ishiwatari et al142014Retrospective22CGN/CPN83% (fenol) vs 69% (ethanol) (1 week)Diarrhea 9%, hypotension 4.5%, burning pain 4.5%, inebriation 4.5%
Si-Jie et al392014Retrospective41CGN vs CPN80% vs 80% (3–90 day)Hypotension 4.9%
Levy et al242019RCT110CGN + CPN vs CPN46% vs 40% (12 week)Transient pain 8.3% vs 44.9%, neurolytic effect 23.3% vs 55.1%
Sakamoto et al412010Retrospective67BPN vs CPNNANone
Minaga et al422016Retrospective64BPN77% (1 week), 67% (4 week)Diarrhea 3.6%, hypotension 4.5%, pain 3.6%, inebriation 8%

EUS-CGN, endoscopic ultrasound-guided celiac ganglia neurolysis; EUS-BPN, endoscopic ultrasound-guided broad plexus neurolysis; CPN, celiac plexus neurolysis; RCT, randomized controlled trials; NA, not available..



Several studies have reported on the treatment efficacy of EUS-CGN compared to EUS-CPN. EUS-CGN is reported to be superior to EUS-CPN, especially in the short term pain relief. In a review by Li et al,40 EUS-CGN had a significantly better short-term pain response rate than EUS-CPN with no difference in adverse events. Doi et al17 evaluated EUS-CPN and EUS-CGN in a randomized prospective trial where they evaluated treatment response on day 7 after treatment and found that both positive response and complete response were significantly higher in the EUS-CGN group. On the other hand, Levy et al24 performed a randomized, double-blind study of EUS-CPN versus EUS-CGN in 110 patients with unresectable pancreatic cancer and reported that the treatment efficacy of EUS-CPN and EUS-CGN was comparable, but patients who received EUS-CGN had a significantly shorter median survival. Although it was unclear from the multivariate analysis whether the shorter survival of CGN compared to CPN was due to the procedure or tumor-related characteristics, the authors concluded that the indication for EUS-CGN should be carefully reexamined. Koulouris et al19 reported in a meta-analysis no significant difference in response rates between central injection, bilateral injection, and CGN at 2 and 4 weeks postoperatively. Based on these findings, EUS-CGN shows good efficacy in short-term pain reduction. However, the possibility of a shorter prognosis should be taken into account, and treatment indications may need to be more carefully evaluated.

EUS-guided broad plexus neurolysis

EUS-guided broad plexus neurolysis (EUS-BPN) was reported by Sakamoto et al41 in 2010. Broadly, the EUS scope is first rotated clockwise from the upper gastric body to delineate the longitudinal aortic cross section and the celiac artery trunk. The scope is advanced 1 to 2 cm toward the anal side of the celiac artery trunk to visualize the branch of the superior mesenteric artery, and a 25-gauge puncture needle is inserted from the lateral side of the aorta to the level of the trunk of the superior mesenteric artery (the Broad technique; Fig. 1A).

Efficacy of EUS-BPN

Broad Technique was first reported in 2010 by Sakamoto et al.41 They reported that EUS-BPN, which punctures the root of the superior mesenteric artery, significantly improved VAS pain scores after 7 and 30 days of treatment compared to EUS-CPN. Minaga et al42 reported that a good pain response in 112 patients who underwent EUS-BPN for pancreatic cancer were 77.7% after 1 week of treatment and 67.9% after 4 weeks of treatment. However, reports on the usefulness of EUS-BPN are limited (Table 2), and further studies are needed.

New techniques for EUS-CPN

In 2021, Saleh et al43 performed EUS-CPN for pancreatic cancer with a mixture of 0.5% bupivacaine (0.5%) and dexmedetomidine, a selective agonist of alpha 2 adrenergic receptors.

They reported that the dexmedetomidine group showed an improvement in pain scores at 24 weeks post-procedure compared to the alcohol group. Recently, Bang et al44 reported the usefulness of EUS-guided radiofrequency ablation (EUS-RFA) for advanced pancreatic cancer by targeting the celiac plexus or visualized ganglia using a 19-gauge fine needle aspiration needle with a 1F monopolar probe. When patients treated with EUS-RFA were compared to patients treated with EUS-CPN in the randomized controlled trial, RFA showed more improvement in pain scores and caused significantly fewer post-procedural gastrointestinal problems than EUS-CPN. However, further study is needed to determine the usefulness of EUS-RFA.

Predictors of treatment effectiveness

Although relatively high treatment response rates have been reported for EUS-CPN (roughly 60%–80%), there are some patients for whom EUS-CPN provides little pain relief. Several reports have examined predictors of treatment response to EUS-CPN and noted that tumor invasion into the celiac plexus is a negative factor for the response to treatment with EUS-CPN.13,18

In a multivariate analysis, Iwata et al13 evaluated the effect of pain reduction in 47 patients who underwent EUS-CPN with a pain score 7 days after treatment. They reported that direct invasion of the celiac plexus and ethanol distribution on the left side only of the celiac artery on postoperative CT were significant factors for the negative effect of EUS-CPN. Han et al18 evaluated the factors involved in treatment response to EUS-CPN for pancreatic cancer and found that distant metastasis, no visible ganglia, and invasion of the celiac plexus were significant factors for negative response. Facciorusso et al45 noted a relationship with sarcopenia. They report that in patients with sarcopenia, the median duration of pain relief was significantly shorter, and complete response rates and VAS score reductions were also significantly lower. Some reports indicate that CT is performed after EUS-CPN treatment and that the greater the contrast spread, the better the pain relief.41,42 The findings of these studies suggest that there are two factors that influence the treatment efficacy of EUS-CPN: procedure-related factors and tumor progression-related factors. Injections only on the left side of the celiac artery or invisible ganglia may technically not provide enough neurolytic agent. It is also suggested that extensive cancer plexus invasion and pathologies associated with tumor progression, such as distant metastases and sarcopenia, may cause somatic pain and local pain control with EUS-CPN may not be sufficient.

EUS-CPN has very clear visibility of the approach route to the celiac plexus, making it easy to avoid surrounding organs and vessels to secure the puncture route. Therefore, it is considered a safer technique than CT-guided or other percutaneous drainage techniques.

As a result, complications are often transient and minor. Diarrhea, transient hypotension, and transient pain aggravation resulting from ethanol injection are commonly reported.19,46

On the other hand, serious complications have been reported in less than 1% of cases, including retroperitoneal abscess, hemorrhage, paraplegia, and ischemia.4750 These complications are often attributed to incorrect injection sites, but some fatal outcomes have been reported,48,50 and EUS-CPN should be performed by an expert endoscopist.

EUS-CPN can be performed more safely and reliably than percutaneous treatment. It is an effective treatment for reducing tumor pain, mainly from pancreatic cancer. It is highly expected that EUS-CPN will improve patients’ quality of life and provide stable and continuous treatment in patients with pancreatic cancer. In recent years, new techniques such as EUS-CGN and EUS-BPN have also been reported, which may be an effective option for difficult-to-control tumor pain. However, EUS-CPN should not be performed routinely and its indications should be carefully evaluated, because there are some reports of insufficient pain reduction and serious complications.

No potential conflict of interest relevant to this article was reported.

  1. Ascunce G, Ribeiro A, Reis I, Rocha-Lima C, Sleeman D, Merchan J, et al. EUS visualization and direct celiac ganglia neurolysis predicts better pain relief in patients with pancreatic malignancy (with video). Gastrointest Endosc. 2011;73:267-74.
    Pubmed CrossRef
  2. Caraceni A, Portenoy RK. Pain management in patients with pancreatic carcinoma. Cancer. 1996;78(3 Suppl):639-53.
    CrossRef
  3. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66: 7-30.
    Pubmed CrossRef
  4. Wray CJ, Ahmad SA, Matthews JB, Lowy AM. Surgery for pancreatic cancer: recent controversies and current practice. Gastroenterology. 2005;128:1626-41.
    Pubmed CrossRef
  5. Eisenberg E, Carr DB, Chalmers TC. Neurolytic celiac plexus block for treatment of cancer pain: a meta-analysis. Anesth Analg. 1995;80:290-5. Erratum in: Anesth Analg. 1995;81:213.
    CrossRef
  6. Nagels W, Pease N, Bekkering G, Cools F, Dobbels P. Celiac plexus neurolysis for abdominal cancer pain: a systematic review. Pain Med. 2013;14:1140-63.
    Pubmed CrossRef
  7. Zhang CL, Zhang TJ, Guo YN, Yang LQ, He MW, Shi JZ, et al. Effect of neurolytic celiac plexus block guided by computerized tomography on pancreatic cancer pain. Dig Dis Sci. 2008;53:856-60.
    Pubmed CrossRef
  8. Wong GY, Schroeder DR, Carns PE, Wilson JL, Martin DP, Kinney MO, et al. Effect of neurolytic celiac plexus block on pain relief, quality of life, and survival in patients with unresectable pancreatic cancer: a randomized controlled trial. JAMA. 2004;291:1092-9.
    Pubmed CrossRef
  9. Wiersema MJ, Wiersema LM. Endosonography-guided celiac plexus neurolysis. Gastrointest Endosc. 1996;44:656-62.
    CrossRef
  10. Wyse JM, Carone M, Paquin SC, Usatii M, Sahai AV. Randomized, double-blind, controlled trial of early endoscopic ultrasound-guided celiac plexus neurolysis to prevent pain progression in patients with newly diagnosed, painful, inoperable pancreatic cancer. J Clin Oncol. 2011;29:3541-6.
    Pubmed CrossRef
  11. LeBlanc JK, DeWitt J, Johnson C, Okumu W, McGreevy K, Symms M, et al. A prospective randomized trial of 1 versus 2 injections during EUS-guided celiac plexus block for chronic pancreatitis pain. Gastrointest Endosc. 2009;69:835-42. Erratum in: Gastrointest Endosc. 2009;70:406.
    Pubmed CrossRef
  12. Téllez-Ávila FI, Romano-Munive AF, Herrera-Esquivel Jde J, Ramírez-Luna MA. Central is as effective as bilateral endoscopic ultrasound-guided celiac plexus neurolysis in patients with unresectable pancreatic cancer. Endosc Ultrasound. 2013;2: 153-6.
    Pubmed KoreaMed CrossRef
  13. Iwata K, Yasuda I, Enya M, Mukai T, Nakashima M, Doi S, et al. Predictive factors for pain relief after endoscopic ultrasound-guided celiac plexus neurolysis. Dig Endosc. 2011;23:140-5.
    Pubmed CrossRef
  14. Ishiwatari H, Hayashi T, Yoshida M, Ono M, Masuko H, Sato T, et al. Phenol-based endoscopic ultrasound-guided celiac plexus neurolysis for East Asian alcohol-intolerant upper gastrointestinal cancer patients: a pilot study. World J Gastroenterol. 2014;20:10512-7.
    Pubmed KoreaMed CrossRef
  15. Das A, Sivak MV Jr. Endoscopic palliation for inoperable pancreatic cancer. Cancer Control. 2000;7:452-7.
    Pubmed CrossRef
  16. Zhao Y, Guo X, Wang K, Chen Q, Wang Y, Chen L, et al. A retrospective multicenter study comparing bupivacaine and ropivacaine in endoscopic ultrasound guided celiac plexus neurolysis. Ann Palliat Med. 2021;10:1755-62.
    Pubmed CrossRef
  17. Doi S, Yasuda I, Kawakami H, Hayashi T, Hisai H, Irisawa A, et al. Endoscopic ultrasound-guided celiac ganglia neurolysis vs. celiac plexus neurolysis: a randomized multicenter trial. Endoscopy. 2013;45:362-9.
    Pubmed CrossRef
  18. Han CQ, Tang XL, Zhang Q, Nie C, Liu J, Ding Z. Predictors of pain response after endoscopic ultrasound-guided celiac plexus neurolysis for abdominal pain caused by pancreatic malignancy. World J Gastroenterol. 2021;27:69-79.
    Pubmed KoreaMed CrossRef
  19. Koulouris AI, Alexandre L, Hart AR, Clark A. Endoscopic ultrasound-guided celiac plexus neurolysis (EUS-CPN) technique and analgesic efficacy in patients with pancreatic cancer: a systematic review and meta-analysis. Pancreatology. 2021;21: 434-42.
    Pubmed CrossRef
  20. Puli SR, Reddy JB, Bechtold ML, Antillon MR, Brugge WR. EUS-guided celiac plexus neurolysis for pain due to chronic pancreatitis or pancreatic cancer pain: a meta-analysis and systematic review. Dig Dis Sci. 2009;54:2330-7.
    Pubmed CrossRef
  21. Asif AA, Walayat SK, Bechtold ML, Revanur V, Puli SR. EUS-guided celiac plexus neurolysis for pain in pancreatic cancer patients - a meta-analysis and systematic review. J Community Hosp Intern Med Perspect. 2021;11:536-42.
    Pubmed KoreaMed CrossRef
  22. Seicean A, Cainap C, Gulei I, Tantau M, Seicean R. Pain palliation by endoscopic ultrasound-guided celiac plexus neurolysis in patients with unresectable pancreatic cancer. J Gastrointestin Liver Dis. 2013;22:59-64.
  23. Wiechowska-Kozłowska A, Boer K, Wójcicki M, Milkiewicz P. The efficacy and safety of endoscopic ultrasound-guided celiac plexus neurolysis for treatment of pain in patients with pancreatic cancer. Gastroenterol Res Pract. 2012;2012: 503098.
    Pubmed KoreaMed CrossRef
  24. Levy MJ, Gleeson FC, Topazian MD, Fujii-Lau LL, Enders FT, Larson JJ, et al. Combined celiac ganglia and plexus neurolysis shortens survival, without benefit, vs plexus neurolysis alone. Clin Gastroenterol Hepatol. 2019;17:728-38.e9.
    Pubmed CrossRef
  25. Mekaroonkamol P, Willingham FF, Chawla S. Endoscopic management of pain in pancreatic cancer. JOP. 2015;16:33-40.
  26. Gunaratnam NT, Sarma AV, Norton ID, Wiersema MJ. A prospective study of EUS-guided celiac plexus neurolysis for pancreatic cancer pain. Gastrointest Endosc. 2001;54:316-24.
    Pubmed CrossRef
  27. Kanno Y, Koshita S, Masu K, Ogawa T, Kusunose H, Murabayashi T, et al. Efficacy of EUS-guided celiac plexus neurolysis compared with medication alone for unresectable pancreatic cancer in the oxycodone/fentanyl era: a prospective randomized control study. Gastrointest Endosc. 2020;92:120-30.
    Pubmed CrossRef
  28. Gress F, Schmitt C, Sherman S, Ikenberry S, Lehman G. A prospective randomized comparison of endoscopic ultrasound- and computed tomography-guided celiac plexus block for managing chronic pancreatitis pain. Am J Gastroenterol. 1999; 94:900-5.
    Pubmed CrossRef
  29. Gress F, Schmitt C, Sherman S, Ciaccia D, Ikenberry S, Lehman G. Endoscopic ultrasound-guided celiac plexus block for managing abdominal pain associated with chronic pancreatitis: a prospective single center experience. Am J Gastroenterol. 2001;96:409-16.
    Pubmed CrossRef
  30. Moura RN, De Moura EG, Bernardo WM, Otoch JP, Bustamante FA, Albers DV, et al. Endoscopic-ultrasound versus percutaneous-guided celiac plexus block for chronic pancreatitis pain. A systematic review and meta-analysis. Rev Gastroenterol Peru. 2015;35:333-41.
  31. Kaufman M, Singh G, Das S, Concha-Parra R, Erber J, Micames C, et al. Efficacy of endoscopic ultrasound-guided celiac plexus block and celiac plexus neurolysis for managing abdominal pain associated with chronic pancreatitis and pancreatic cancer. J Clin Gastroenterol. 2010;44:127-34.
    Pubmed CrossRef
  32. Teoh AYB, Dhir V, Kida M, Yasuda I, Jin ZD, Seo DW, et al. Consensus guidelines on the optimal management in interventional EUS procedures: results from the Asian EUS group RAND/UCLA expert panel. Gut. 2018;67:1209-28.
    Pubmed CrossRef
  33. Bhatnagar S, Joshi S, Rana SP, Mishra S, Garg R, Ahmed SM. Bedside ultrasound-guided celiac plexus neurolysis in upper abdominal cancer patients: a randomized, prospective study for comparison of percutaneous bilateral paramedian vs. unilateral paramedian needle-insertion technique. Pain Pract. 2014;14:E63-8.
    Pubmed CrossRef
  34. LeBlanc JK, Al-Haddad M, McHenry L, Sherman S, Juan M, McGreevy K, et al. A prospective, randomized study of EUS-guided celiac plexus neurolysis for pancreatic cancer: one injection or two? Gastrointest Endosc. 2011;74:1300-7.
    Pubmed CrossRef
  35. Sahai AV, Lemelin V, Lam E, Paquin SC. Central vs. bilateral endoscopic ultrasound-guided celiac plexus block or neurolysis: a comparative study of short-term effectiveness. Am J Gastroenterol. 2009;104:326-9.
    Pubmed CrossRef
  36. Levy M, Rajan E, Keeney G, Fletcher JG, Topazian M. Neural ganglia visualized by endoscopic ultrasound. Am J Gastroenterol. 2006;101:1787-91.
    Pubmed CrossRef
  37. Gerke H, Silva RG Jr, Shamoun D, Johnson CJ, Jensen CS. EUS characteristics of celiac ganglia with cytologic and histologic confirmation. Gastrointest Endosc. 2006;64:35-9.
    Pubmed CrossRef
  38. Levy MJ, Topazian MD, Wiersema MJ, Clain JE, Rajan E, Wang KK, et al. Initial evaluation of the efficacy and safety of endoscopic ultrasound-guided direct Ganglia neurolysis and block. Am J Gastroenterol. 2008;103:98-103.
    Pubmed CrossRef
  39. Si-Jie H, Wei-Jia X, Yang D, Lie Y, Feng Y, Yong-Jian J, et al. How to improve the efficacy of endoscopic ultrasound-guided celiac plexus neurolysis in pain management in patients with pancreatic cancer: analysis in a single center. Surg Laparosc Endosc Percutan Tech. 2014;24:31-5.
    Pubmed KoreaMed CrossRef
  40. Li M, Wang Z, Chen Y, Wu Z, Huang X, Wu C, et al. EUS-CGN versus EUS-CPN in pancreatic cancer: a qualitative systematic review. Medicine (Baltimore). 2021; 100:e27103.
    Pubmed KoreaMed CrossRef
  41. Sakamoto H, Kitano M, Kamata K, Komaki T, Imai H, Chikugo T, et al. EUS-guided broad plexus neurolysis over the superior mesenteric artery using a 25-gauge needle. Am J Gastroenterol. 2010;105:2599-606.
    Pubmed CrossRef
  42. Minaga K, Kitano M, Sakamoto H, Miyata T, Imai H, Yamao K, et al. Predictors of pain response in patients undergoing endoscopic ultrasound-guided neurolysis for abdominal pain caused by pancreatic cancer. Therap Adv Gastroenterol. 2016;9: 483-94.
    Pubmed KoreaMed CrossRef
  43. Saleh AAG, Sultan A, Hammouda MA, Shawki A, El Ghaffar MA. Value of adding dexmedetomidine in endoscopic ultrasound-guided celiac plexus neurolysis for treatment of pancreatic cancer-associated pain. J Gastrointest Cancer. 2021;52: 682-9.
    Pubmed CrossRef
  44. Bang JY, Sutton B, Hawes RH, Varadarajulu S. EUS-guided celiac ganglion radiofrequency ablation versus celiac plexus neurolysis for palliation of pain in pancreatic cancer: a randomized controlled trial (with videos). Gastrointest Endosc. 2019; 89:58-66.e3.
    Pubmed CrossRef
  45. Facciorusso A, Antonino M, Muscatiello N. Sarcopenia represents a negative prognostic factor in pancreatic cancer patients undergoing EUS celiac plexus neurolysis. Endosc Ultrasound. 2020;9:238-44.
    Pubmed KoreaMed CrossRef
  46. O’Toole TM, Schmulewitz N. Complication rates of EUS-guided celiac plexus blockade and neurolysis: results of a large case series. Endoscopy. 2009;41:593-7.
    Pubmed CrossRef
  47. Mittal MK, Rabinstein AA, Wijdicks EF. Pearls & oy-sters: acute spinal cord infarction following endoscopic ultrasound-guided celiac plexus neurolysis. Neurology. 2012;78:e57-9.
    Pubmed CrossRef
  48. Gimeno-García AZ, Elwassief A, Paquin SC, Sahai AV. Fatal complication after endoscopic ultrasound-guided celiac plexus neurolysis. Endoscopy. 2012;44(Suppl 2):E267.
    Pubmed CrossRef
  49. Fujii L, Clain JE, Morris JM, Levy MJ. Anterior spinal cord infarction with permanent paralysis following endoscopic ultrasound celiac plexus neurolysis. Endoscopy. 2012;44(Suppl 2):E265-6.
    CrossRef
  50. Jang HY, Cha SW, Lee BH, Jung HE, Choo JW, Cho YJ, et al. Hepatic and splenic infarction and bowel ischemia following endoscopic ultrasound-guided celiac plexus neurolysis. Clin Endosc. 2013;46:306-9.
    Pubmed KoreaMed CrossRef