Skip to main content
Erschienen in: Journal of Gastroenterology 5/2019

Open Access 26.02.2019 | Review

Recent advancement in EUS-guided fine needle sampling

verfasst von: Pujan Kandel, Michael B. Wallace

Erschienen in: Journal of Gastroenterology | Ausgabe 5/2019

Abstract

EUS-guided tissue acquisition technique plays an essential role for evaluation of gastrointestinal tumors. Several components affect the yield of EUS-guided tissue acquisition outcomes such as sampling techniques, use of ROSE (rapid onsite evaluation), training and experience, and needle designs. In this review we discuss advancement in EUS-guided fine needle sampling.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
EUS
Endoscopic ultrasound
FNA
Fine needle aspiration
FNB
Fine needle biopsy
EUS–FNA
Endoscopic ultrasound fine needle aspiration
EUS–FNB
Endoscopic ultrasound fine needle biopsy
RCT
Randomized controlled trial
FFPE
Formalin fixed, paraffin-embedded
GIST
Gastrointestinal Stromal tumor
PDXs
Patient-derived xenografts
EUS–TTNFB
EUS-guided through-the-needle forceps biopsy
ROSE
Rapid onsite evaluation

Introduction

Endoscopic ultrasound is one of the important tools for evaluation of gastrointestinal tumors and areas around the gastrointestinal tract. Tissue procurement techniques and tools have evolved significantly over a period of 25 years. Endoscopic ultrasound-guided fine needle aspiration (EUS–FNA) and fine needle biopsy (EUS–FNB) are useful for screening, pathological diagnosis, and staging such as pancreaticobiliary, esophageal, gastric, rectal, and lung diseases [1]. The main end results of EUS-guided fine needle samplings include adverse events, accuracy, histology, and diagnostic yield [2]. EUS-guided tissue sampling is relatively safe and accurate [3]. There are several factors that affect the outcome of this technique such as lesion location and characteristics, experience of endoscopist, EUS fine needle aspiration (FNA) versus fine needle biopsy (FNB), needle gauge, sampling technique, and the presence of onsite cytotechnician [4]. One of the main challenges associated with this technique is low diagnostic yield (false-negative diagnosis) which ranges up to 4%–45% in solid pancreatic mass, 21%–53% in pancreatic cystic neoplasms, and 6%–14% in lymph nodes [5]. The main objective of this review is to focus on the recent advancement in EUS-guided tissue acquisition techniques, needle technologies, and its clinical applications.

Indication of EUS–FNA

EUS–FNA is useful technique for diagnosis and staging of lesions in and around the proximity of gastrointestinal tract [611]. It is superior and effective compared to CT-guided or ultrasound-guided biopsy of lesions [12]. For diagnosis of solid pancreatic lesions, sensitivity and specificity of EUS–FNA is 85%–89% and 96%–99% [13, 14], whereas for pancreatic cystic lesions it is 54% and 93% [15]. The lower sensitivity for diagnosis of cystic lesions is often due to sampling errors, insufficient aspirates, or may be due to different approaches (transgastric or transduodenal) [16, 17]. EUS–FNA is useful technique for pathological diagnosis of abdominal and mediastinal lymph nodes [18]. Tissue procurement from lymph nodes is quite challenging especially in cases of hematologic malignancies such as lymphoproliferative disorders which require adequate tissue to perform immunophenotyping and describe histological architecture. Some studies demonstrated that accuracy of FNA for abnormal lymphocytes is around 70%–90% [1925]. For mediastinal lymph nodes sensitivity and specificity of EUS–FNA are 88% and 96% vs 84% vs 88% with EUS imaging alone [26]. Thus, EUS–FNA is safer and less invasive compared to mediastinoscopy and is the first choice for small cell cancer staging when combined with bronchoscopy [27, 28]. In addition EUS–FNA is useful for evaluation of sub epithelial lesions, nodal staging of esophageal cancer, liver lesion, and malignant biliary structures [2931]. Summary of diagnostic values EUS–FNA for different lesions are illustrated below (Table 1). Optimal tissue acquisition from lesions depends on various factors such as needle FNA needle sizes and gauges, fine needle biopsy (FNB) needles, presence of cytotechnologists for rapid onsite evaluation (ROSE), expertise of endoscopist, and tissue handling techniques [32]. American Society for Gastrointestinal Endoscopy (ASGE) guidelines recommend 150 supervised EUS procedures of which 75 should involve pancreaticobiliary system with 50 of them include EUS–FNA [33]. However, recent metaanalysis emphasizes on no clear number of EUS procedures [34] but the success of endoscopist depends on performing EUS at high volume center as procedure is greatly operator dependent [35].
Table 1
Diagnostic values for EUS–FNA for different lesions
Lesions
Sensitivity
Specificity
Accuracy
Malignant biliary strictures
 Navaneethan et al. (2015) [117]
66%
88%
 Sadeghi et al. (2016) [116]
100%
80%
Subepithelial lesions (upper and lower GI tract)
 Turhan et al. (2011) [115]
82.9%
73.3%
80%
 Larghi et al. (2014) [114]
93%
Liver lesions
   
 Tenberge et al. (2002) [31]
89%
Solid pancreatic lesions
 Hebert-Magee et al. (2013) [113]
88.6%
99.3%
 Hewitt MJ et al. (2012) [13]
85%
98%
 Puli SR et al. (2013) [14]
86.8%
95.8%
Cystic Pancreatic lesions
 Thronton et al. (2013) [15]
54%
93%
 Suzuki et al. (2014) [112]
64.8%
90.6%
 
Mediastinal lymph nodes
 Puli et al. (2008) [26]
88%
96%

Advancement in EUS-guided FNA and FNB

Needle gauge and type

For last 2 decades EUS–FNA has been a mainstay for sampling of tissue from pancreas, lymph nodes, liver, and subepithelial lesions. Currently 3 types of EUS-FNA needles are available in market for clinical use: 19G, 22G, and 25G. Several studies have been published comparing 22G vs 25G needles on lymph nodes and solid lesions [3645]. Most studies showed similar diagnostic yield of malignancy between groups. However, result of the two meta-analyses comparing 25G vs 22G showed that 25G is more sensitive than 22G for diagnosing pancreatic malignancy with overall adequate specimen obtained with 25-gauge needle [46, 47]. Therefore, “the use of 25G needle is associated with a higher diagnostic yield compared with a 22G needle in patients undergoing EUS-FNA of pancreatic masses” [4]. In addition RCT (randomized controlled trials) have shown no significant difference in diagnostic yield malignancy using 19G needle compared with the 22G/25G needle [4850]. EUS–FNA technique is relatively safe with few adverse events such as pain, pancreatitis, bleeding, and infections [51] with an morbidity of 0.98%. Pancreatitis rate is about 0.44% and overall mortality is 0.02% [52].
It is often difficult to diagnose with FNA cytology alone in conditions such as autoimmune pancreatitis, lymphoma, and well differentiated adenocarcinoma where histology with preserved tissue architecture is important. Therefore, fine needle biopsy (FNB) is valuable as it provides well-preserved quality tissue samples. Quick-Core (Cook Medical, Limerick, Ireland) was the first FNB needle (also called Tru-Cut core biopsy needle) introduced in market. This 19G needle had an ability to procure large core tissue with a diagnostic yield ranging from 52 to 95% [49, 5364]. Despite acquisition of large core tissue, it was later removed from marketplace due to lack of its flexibility, availability in only 19-gauge platform, and difficult access through transduodenal route. In addition Quick-Core did not improve diagnostic yield significantly over FNA. This was later replaced by new needle called ProCore (Echo Tip ProCore [ETP], Cook Medical, Limerick, Ireland) with reverse bevel design, Fig. 1. This needle is available in all seizes 19 g, 22 g, and 25 g. The flexibility of this needle allows access through stomach and duodenum without difficulties. Two RCTs have demonstrated similar diagnostic yield of malignancy for FNB and FNA [65, 66]. One recent meta-analysis included nine studies including retrospective, RCTs, and prospective studies [67]. There was no significant difference between ProCore and standard FNA needles in terms of diagnostic adequacy (78% vs 77%), accuracy (86% vs 86%), and core specimen procurement (78% vs 77%) but mean number of passes was significantly lower in ProCore (mean difference 1.2; p < 0.01).
In over past 2–3 years, there have been significant paradigm changes in tip of EUS–FNB needle designs. Two FNB needles: one with fork-tip design with two leading sharp tips on the opposite side of the lumen to improve the tissue capture (SharkCore, Medtronic, Minneapolis, Minn.), Fig. 2 and another with three symmetric cutting edges (Acquire, Boston Scientific Corp, Natick, Mass.), Fig. 3 have been introduced in marketplace. Several studies have been published since then and results have demonstrated significant performance in terms of diagnostic yield and histology yield [6873] for both the needles. In one randomized trial 22-gauge Franseen and fork-tip needles in sampling of solid pancreatic masses were reported. The main outcomes of the study were tumor morphology and histologic adequacy. A total of 50 patients were included in whom sampling was performed using both the needles with the order randomized. Results from this trial showed comparable diagnostic yield (96% vs 92%, p = 0.32) and diagnostic adequacy with ROSE (94% vs 96%, p = .32) between Franseen and fork-tip needles. In addition there was no statistically significant difference in terms of tissue quality or quantity, total tissue obtained, total tumor tissue, and the desmoplastic fibrosis yield by the two needles [74]. However, in another retrospective study, diagnostic yield was significantly lower in Franseen needle compared to the fork-tip needle (63% vs 77%, p = 0.2). In subgroup analysis taking account of only solid pancreatic masses, lower yield was reported with Franseen needle compared to fork-tip needle (64% vs 85%, p = 0.01). Therefore, it is more important to have larger multicenter randomized trials to address the fine discrepancies in new FNB needles [75].
Published literatures have demonstrated that FNB is superior to provide adequate histological tissue compared to FNA and may save cost associated with ROSE but the questions still remain: how many dedicated passes are required for different lesions such as gastrointestinal stromal tumor (GIST), solid pancreatic tumor, etc., if to replace ROSE? How safe is FNB? What is the ideal technique? Thus, there is still need of multicenter studies to address these subtle issues.

EUS-guided tissue acquisition technique

ROSE (rapid onsite evaluation)

The main objectives of the ROSE are to provide real time feedback during endoscopy regarding the content and adequacy of specimen, to minimize the number of passes, to decrease inadequate samples, and to increase efficiency of procedure, Fig. 4. In one retrospective study EUS-guided FNA with or without ROSE was compared for sampling of pancreatic mass. Results showed that EUS–FNA with ROSE yielded greater sensitivity (96% vs 78%) and fewer insufficient samples (1% vs 12.6%) with less number of passes [76]. However, results from recent multicenter randomized controlled trial (RCT) showed similar diagnostic yield of malignancy and proportion of inadequate samples when sampling of EUS–FNA of pancreatic masses with and without ROSE. Although the EUS–FNA with ROSE arm required fewer passes, the diagnostic and cytologic yield, adverse events, total procedure time, accuracy, and number of repeat procedures were similar between two groups [77].
With the arrival of new FNB needles, ROSE has less significant impact on diagnostic yield [3, 7880]. Use of FNB may eliminate ROSE as dedicated core tissue could be obtained with less number of passes. However, in centers with low adequacy rate (< 90%) and less experienced endoscopist, ROSE may have some substantial role [81].

Stylet and suction use, fanning technique, capillary/slow pull, and wet suction technique

The main aim of using stylet is to prevent blockade lumen of needle as it passes though the gastrointestinal wall. Studies have reported similar diagnostic yield and specimen adequacy with and without stylet [8284]. In one study flushing of air slowly was superior than insertion of stylet second time to aspirate the samples form solid pancreatic mass [85]. Use of suction yielded higher cellularity, accuracy, and diagnostic yield.
Fanning technique is usually utilized for acquisition of more tissue. One of the studies demonstrated that fanning technique yielded more tissue with fewer number of passes compared to standard technique [86].
Another technique called capillary/slow pull technique is usually utilized to increase aspiration of tissue as it creates negative pressure as stylet is slowly withdrawn during FNA passes. Result from one study demonstrated that adequate and good samples were obtained with capillary technique. However, malignancy yield was similar between suction and capillary technique (90% vs 90%, p = 1.00) [86].
In wet suction technique, air is replaced with saline. Whole idea of this technique to acquire more tissue during EUS–FNA passes. In one RCT wet suction yielded more tissue with higher cellularity compared to standard technique [87], Fig. 5a, b.

Confocal laser endomicroscopy (CLE) during EUS–FNA

Confocal laser endomicroscopy is one of the novel imaging technologies that allows microscopic visualization of the mucosal surface epithelium. Optical biopsy at realtime may further improve the diagnostic yield by reducing the sampling error. In addition this may provide the realtime feedback at the time of procedure when onsite cytologist is not available. Needle-based confocal laser endomicroscopy (nCLE) was initially studied in the rat liver and porcine model [88, 89] but the use of this prototype in human solid organs such as pancreas and lymph nodes has been started recently, Fig. 6. This technology involves passage of mini-CLE probe through 19G during EUS–FNA and tissue level can be pictured at realtime. Some studies demonstrated feasibility and safety of nCLE in solid organs [9092]. In one prospective study, reproducibility and diagnostic value of nCLE for solid lesions were evaluated. Results from this study demonstrated that diagnostic values and interobserver agreement between experts for nCLE parameter were poor [93]. The main limitation of this technology is heterogenicity in histology, interobserver variability, reproducibility, need of pathologists and endoscopists for better interpretation, image quality, and sampling error [94]. Further training and research are needed for applicability in realtime practice.

EUS-guided needle forceps biopsy

EUS-guided fine needle aspiration is important for cytological diagnosis. Advancement in needle technologies has improved yield of EUS sampling benefiting for both cytological and histological analysis. EUS-guided fine needle forceps biopsy technique was initially started in porcine model [95] which involved the use of small caliber biopsy forceps through 19-gauge FNA needle. In one retrospective study safety and efficiency of EUS-guided through-the-needle forceps biopsy (EUS–TTNFB) were studied. Results from this study demonstrated that EUS–TTNFB was feasible and safe, and provided additional tissue for histological analysis, Fig. 7. No adverse events were reported. Macroscopic core tissue was obtained at a rate of 71% per pass [96].
In addition to solid organs, this novel tissue procurement technique has also been studied in pancreas cystic lesions. Distinguishing the cyst lesions appropriately may help in its management. Cystic lesions that represent diagnostic challenge can be better evaluated histologically with the help of EUS-guided biopsy of the pancreatic cystic walls. Results from one case series demonstrated that needle biopsy forceps was useful in distinguishing the nature of the cyst and stratify their management [97]. Another multicenter retrospective study also evaluated the utility of this technique in evaluation of cystic lesions. The technical and clinical success rate of this technique was found to be 85.7% and 71.4%. Adverse events were observed only in 10.7% of cases [98].
Results from early studies have demonstrated that the use of EUS-guided fine needle biopsy forceps is feasible with reasonable success rates. Multicenter prospective studies are needed to determine its wide spread use in clinical practice compared to other EUS-guided sampling techniques.

EUS-guided sampling in precision therapy

Precision therapy means providing an individualized management to patient with the use of genomic information [99]. EUS-guided sampling plays a significant role in targeting gastrointestinal cancers in which tissue can be utilized for genomic analysis. This approach increases the therapeutic goal of chemotherapy and reduces side effects associated with chemotherapeutic drug [100]. Studies have reported that cytologic samples obtained by EUS–FNA are an excellent source for genetic analysis [101105]. However, for years; formalin fixed, paraffin-embedded (FFPE) tissue blocks obtained from surgical samples have been used for genomic analysis.
Recent advancement in needle designs has improved the ability to obtain core biopsies that could be utilized for whole exome sequencing and additional genomic analysis. Development of tumor models such as organoid and xenograft has been possible with core biopsies sample which can be used for in vitro drug testing. Result from one study demonstrated that successful organoid creation was possible in 85% of patients with pancreas cancer [106].
Patient-derived xenografts (PDXs) is created by transmitting the resected tumor tissue from surgery to an immunocompromised host to stimulate the human biology in vivo [100]. This model provides important information on tumor biology that could be utilized for evaluation of different cancers and evaluation of chemotherapeutic drugs [107]. PDXs models can be developed with the use of EUS–FNB technique but delay in engraftment may limit its value in real time management [108].
Newer FNB needle designs have demonstrated adequate core tissue procurement with fewer numbers of passes [109, 110]. More DNA can be extracted from the tumor tissue which could be utilized for full genomic analysis. In one ongoing study by Kandel et al. demonstrated that 74% of EUS–FNB samples of pancreatic cancer were adequate for whole exome sequencing compared to 54% of EUS–FNA samples [111]. Therefore, newer generation needles may be helpful in the era of precision medicine especially in patients with pancreas cancer.

Conclusion

Advancement in EUS-guided tissue sampling techniques and development of new needle designs that have improved the diagnostic yield of solid lesions. This innovation in EUS has also opened the door for early diagnosis and precision therapy in the management of cancer patients.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Publisher's Note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Kandel P, Wallace MB. Optimizing endoscopic ultrasound guided tissue sampling of the pancreas. JOP J Pancreas. 2016;17:160–5. Kandel P, Wallace MB. Optimizing endoscopic ultrasound guided tissue sampling of the pancreas. JOP J Pancreas. 2016;17:160–5.
2.
Zurück zum Zitat Early DS, Ben-Menachem T, Decker GA, et al. Aproppiate use of gastrointestinal endoscopy. Gastrointest Endosc. 2012;75:1127–31.CrossRef Early DS, Ben-Menachem T, Decker GA, et al. Aproppiate use of gastrointestinal endoscopy. Gastrointest Endosc. 2012;75:1127–31.CrossRef
3.
Zurück zum Zitat Keswani RN, Krishnan K, Wani S, et al. Addition of endoscopic ultrasound (EUS)-guided fine needle aspiration and on-site cytology to EUS-guided fine needle biopsy increases procedure time but not diagnostic accuracy. Clin Endosc. 2014;47:242–7.CrossRefPubMedPubMedCentral Keswani RN, Krishnan K, Wani S, et al. Addition of endoscopic ultrasound (EUS)-guided fine needle aspiration and on-site cytology to EUS-guided fine needle biopsy increases procedure time but not diagnostic accuracy. Clin Endosc. 2014;47:242–7.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Wani S, Muthusamy VR, Komanduri S. EUS-guided tissue acquisition: an evidence-based approach (with videos). Gastrointest Endosc. 2014;80(939–959):e937. Wani S, Muthusamy VR, Komanduri S. EUS-guided tissue acquisition: an evidence-based approach (with videos). Gastrointest Endosc. 2014;80(939–959):e937.
5.
Zurück zum Zitat Fujii LL, Levy MJ. Pitfalls in EUS FNA. Gastrointest Endosc Clin N Am. 2014;24:125–42.CrossRef Fujii LL, Levy MJ. Pitfalls in EUS FNA. Gastrointest Endosc Clin N Am. 2014;24:125–42.CrossRef
6.
Zurück zum Zitat Alomari AK, Ustun B, Aslanian HR, et al. Endoscopic ultrasound-guided fine-needle aspiration diagnosis of secondary tumors involving the pancreas: an institution’s experience. Cyto J. 2016;13:1. Alomari AK, Ustun B, Aslanian HR, et al. Endoscopic ultrasound-guided fine-needle aspiration diagnosis of secondary tumors involving the pancreas: an institution’s experience. Cyto J. 2016;13:1.
7.
Zurück zum Zitat Philipper M, Hollerbach S, Gabbert HE, et al. Prospective comparison of endoscopic ultrasound-guided fine-needle aspiration and surgical histology in upper gastrointestinal submucosal tumors. Endoscopy. 2010;42:300–5.CrossRefPubMed Philipper M, Hollerbach S, Gabbert HE, et al. Prospective comparison of endoscopic ultrasound-guided fine-needle aspiration and surgical histology in upper gastrointestinal submucosal tumors. Endoscopy. 2010;42:300–5.CrossRefPubMed
8.
Zurück zum Zitat Patil R, Ona MA, Papafragkakis C, et al. Endoscopic ultrasound-guided fine-needle aspiration in the diagnosis of adrenal lesions. Ann Gastroenterol. 2016;29:307–11.PubMedPubMedCentral Patil R, Ona MA, Papafragkakis C, et al. Endoscopic ultrasound-guided fine-needle aspiration in the diagnosis of adrenal lesions. Ann Gastroenterol. 2016;29:307–11.PubMedPubMedCentral
9.
Zurück zum Zitat Levy MJ, Abu Dayyeh BK, Fujii LL, et al. Detection of peritoneal carcinomatosis by EUS fine-needle aspiration: impact on staging and resectability (with videos). Gastrointest Endosc. 2015;81:1215–24.CrossRefPubMed Levy MJ, Abu Dayyeh BK, Fujii LL, et al. Detection of peritoneal carcinomatosis by EUS fine-needle aspiration: impact on staging and resectability (with videos). Gastrointest Endosc. 2015;81:1215–24.CrossRefPubMed
10.
Zurück zum Zitat Levy MJ, Gleeson FC, Zhang L. Endoscopic ultrasound fine-needle aspiration detection of extravascular migratory metastasis from a remotely located pancreatic cancer. Clin Gastroenterol Hepatol. 2009;7:246–8.CrossRefPubMed Levy MJ, Gleeson FC, Zhang L. Endoscopic ultrasound fine-needle aspiration detection of extravascular migratory metastasis from a remotely located pancreatic cancer. Clin Gastroenterol Hepatol. 2009;7:246–8.CrossRefPubMed
11.
Zurück zum Zitat Dumonceau JM, Deprez PH, Jenssen C, et al. Indications, results, and clinical impact of endoscopic ultrasound (EUS)-guided sampling in gastroenterology: european society of gastrointestinal endoscopy (ESGE) clinical guideline—updated January 2017. Endoscopy. 2017;49:695–714.CrossRefPubMed Dumonceau JM, Deprez PH, Jenssen C, et al. Indications, results, and clinical impact of endoscopic ultrasound (EUS)-guided sampling in gastroenterology: european society of gastrointestinal endoscopy (ESGE) clinical guideline—updated January 2017. Endoscopy. 2017;49:695–714.CrossRefPubMed
12.
Zurück zum Zitat Horwhat JD, Paulson EK, McGrath K, et al. A randomized comparison of EUS-guided FNA versus CT or US-guided FNA for the evaluation of pancreatic mass lesions. Gastrointest Endosc. 2006;63:966–75.CrossRefPubMed Horwhat JD, Paulson EK, McGrath K, et al. A randomized comparison of EUS-guided FNA versus CT or US-guided FNA for the evaluation of pancreatic mass lesions. Gastrointest Endosc. 2006;63:966–75.CrossRefPubMed
13.
Zurück zum Zitat Hewitt MJ, McPhail MJ, Possamai L, et al. EUS-guided FNA for diagnosis of solid pancreatic neoplasms: a meta-analysis. Gastrointest Endosc. 2012;75:319–31.CrossRefPubMed Hewitt MJ, McPhail MJ, Possamai L, et al. EUS-guided FNA for diagnosis of solid pancreatic neoplasms: a meta-analysis. Gastrointest Endosc. 2012;75:319–31.CrossRefPubMed
14.
Zurück zum Zitat Puli SR, Bechtold ML, Buxbaum JL, et al. How good is endoscopic ultrasound-guided fine-needle aspiration in diagnosing the correct etiology for a solid pancreatic mass?: a meta-analysis and systematic review. Pancreas. 2013;42:20–6.CrossRefPubMed Puli SR, Bechtold ML, Buxbaum JL, et al. How good is endoscopic ultrasound-guided fine-needle aspiration in diagnosing the correct etiology for a solid pancreatic mass?: a meta-analysis and systematic review. Pancreas. 2013;42:20–6.CrossRefPubMed
15.
Zurück zum Zitat Thornton GD, McPhail MJ, Nayagam S, et al. Endoscopic ultrasound guided fine needle aspiration for the diagnosis of pancreatic cystic neoplasms: a meta-analysis. Pancreatology. 2013;13:48–57.CrossRefPubMed Thornton GD, McPhail MJ, Nayagam S, et al. Endoscopic ultrasound guided fine needle aspiration for the diagnosis of pancreatic cystic neoplasms: a meta-analysis. Pancreatology. 2013;13:48–57.CrossRefPubMed
16.
17.
Zurück zum Zitat Woolf KMW, Liang H, Sletten ZJ, et al. False-negative rate of endoscopic ultrasound-guided fine-needle aspiration for pancreatic solid and cystic lesions with matched surgical resections as the gold standard. Cancer Cytopathology. 2013;121:449–58.CrossRefPubMed Woolf KMW, Liang H, Sletten ZJ, et al. False-negative rate of endoscopic ultrasound-guided fine-needle aspiration for pancreatic solid and cystic lesions with matched surgical resections as the gold standard. Cancer Cytopathology. 2013;121:449–58.CrossRefPubMed
18.
Zurück zum Zitat Dietrich CF, Annema JT, Clementsen P, et al. Ultrasound techniques in the evaluation of the mediastinum, part I: endoscopic ultrasound (EUS), endobronchial ultrasound (EBUS) and transcutaneous mediastinal ultrasound (TMUS), introduction into ultrasound techniques. J Thorac Dis. 2015;7:E311–25.PubMedPubMedCentral Dietrich CF, Annema JT, Clementsen P, et al. Ultrasound techniques in the evaluation of the mediastinum, part I: endoscopic ultrasound (EUS), endobronchial ultrasound (EBUS) and transcutaneous mediastinal ultrasound (TMUS), introduction into ultrasound techniques. J Thorac Dis. 2015;7:E311–25.PubMedPubMedCentral
19.
Zurück zum Zitat Tournoy KG, Praet MM, Van Maele G, et al. Esophageal endoscopic ultrasound with fine-needle aspiration with an on-site cytopathologist: high accuracy for the diagnosis of mediastinal lymphadenopathy. Chest. 2005;128:3004–9.CrossRefPubMed Tournoy KG, Praet MM, Van Maele G, et al. Esophageal endoscopic ultrasound with fine-needle aspiration with an on-site cytopathologist: high accuracy for the diagnosis of mediastinal lymphadenopathy. Chest. 2005;128:3004–9.CrossRefPubMed
20.
Zurück zum Zitat Sǎftoiu A, Vilmann P, Guldhammer Skov B, et al. Endoscopic ultrasound (EUS)-guided trucut biopsy adds significant information to EUS-guided fine-needle aspiration in selected patients: a prospective study. Scand J Gastroenterol. 2007;42:117–25.CrossRefPubMed Sǎftoiu A, Vilmann P, Guldhammer Skov B, et al. Endoscopic ultrasound (EUS)-guided trucut biopsy adds significant information to EUS-guided fine-needle aspiration in selected patients: a prospective study. Scand J Gastroenterol. 2007;42:117–25.CrossRefPubMed
21.
Zurück zum Zitat Jue TL, Sharaf RN, Appalaneni V, et al. Role of EUS for the evaluation of mediastinal adenopathy. Gastrointest Endosc. 2011;74:239–45.CrossRefPubMed Jue TL, Sharaf RN, Appalaneni V, et al. Role of EUS for the evaluation of mediastinal adenopathy. Gastrointest Endosc. 2011;74:239–45.CrossRefPubMed
22.
Zurück zum Zitat Yasuda I, Goto N, Tsurumi H, et al. Endoscopic ultrasound-guided fine needle aspiration biopsy for diagnosis of lymphoproliferative disorders: feasibility of immunohistological, flow cytometric, and cytogenetic assessments. Am J Gastroenterol. 2012;107:397–404.CrossRefPubMed Yasuda I, Goto N, Tsurumi H, et al. Endoscopic ultrasound-guided fine needle aspiration biopsy for diagnosis of lymphoproliferative disorders: feasibility of immunohistological, flow cytometric, and cytogenetic assessments. Am J Gastroenterol. 2012;107:397–404.CrossRefPubMed
23.
Zurück zum Zitat Al-Haddad M, Savabi MS, Sherman S, et al. Role of endoscopic ultrasound-guided fine-needle aspiration with flow cytometry to diagnose lymphoma: a single center experience. J Gastroenterol Hepatol (Aust). 2009;24:1826–33.CrossRef Al-Haddad M, Savabi MS, Sherman S, et al. Role of endoscopic ultrasound-guided fine-needle aspiration with flow cytometry to diagnose lymphoma: a single center experience. J Gastroenterol Hepatol (Aust). 2009;24:1826–33.CrossRef
24.
Zurück zum Zitat Mehra M, Tamhane A, Eloubeidi MA. EUS-guided FNA combined with flow cytometry in the diagnoses of suspected or recurrent intrathoracic or retroperitoneal lymphoma. Gastrointest Endosc. 2005;62:508–13.CrossRefPubMed Mehra M, Tamhane A, Eloubeidi MA. EUS-guided FNA combined with flow cytometry in the diagnoses of suspected or recurrent intrathoracic or retroperitoneal lymphoma. Gastrointest Endosc. 2005;62:508–13.CrossRefPubMed
25.
Zurück zum Zitat Nakahara O, Yamao K, Bhatia V, et al. Usefulness of endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) for undiagnosed intra-abdominal lymphadenopathy. J Gastroenterol. 2009;44:562–7.CrossRefPubMed Nakahara O, Yamao K, Bhatia V, et al. Usefulness of endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) for undiagnosed intra-abdominal lymphadenopathy. J Gastroenterol. 2009;44:562–7.CrossRefPubMed
26.
Zurück zum Zitat Puli SR, Batapati Krishna Reddy J, Bechtold ML, et al. Endoscopic ultrasound: it’s accuracy in evaluating mediastinal lymphadenopathy? A meta-analysis and systematic review. World J Gastroenterol. 2008;14:3028–37.CrossRefPubMedPubMedCentral Puli SR, Batapati Krishna Reddy J, Bechtold ML, et al. Endoscopic ultrasound: it’s accuracy in evaluating mediastinal lymphadenopathy? A meta-analysis and systematic review. World J Gastroenterol. 2008;14:3028–37.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Bhutani MS. Transesophageal endoscopic ultrasound-guided mediastinal lymph node aspiration: does the end justify the means? Chest. 2000;117:298–301.CrossRefPubMed Bhutani MS. Transesophageal endoscopic ultrasound-guided mediastinal lymph node aspiration: does the end justify the means? Chest. 2000;117:298–301.CrossRefPubMed
28.
Zurück zum Zitat Vilmann P, Clementsen PF, Colella S, et al. Combined endobronchial and oesophageal endosonography for the diagnosis and staging of lung cancer. European society of gastrointestinal endoscopy (ESGE) guideline, in cooperation with the european respiratory society (ERS) and the European society of thoracic surgeons (ESTS). Euro Respir J. 2015;46:40–60.CrossRef Vilmann P, Clementsen PF, Colella S, et al. Combined endobronchial and oesophageal endosonography for the diagnosis and staging of lung cancer. European society of gastrointestinal endoscopy (ESGE) guideline, in cooperation with the european respiratory society (ERS) and the European society of thoracic surgeons (ESTS). Euro Respir J. 2015;46:40–60.CrossRef
29.
Zurück zum Zitat Yamabe A, Irisawa A, Bhutani MS, et al. Usefulness of endoscopic ultrasound-guided fine-needle aspiration with a forward-viewing and curved linear-array echoendoscope for small gastrointestinal subepithelial lesions. Endosc Int Open. 2015;3:E161–4.CrossRefPubMedPubMedCentral Yamabe A, Irisawa A, Bhutani MS, et al. Usefulness of endoscopic ultrasound-guided fine-needle aspiration with a forward-viewing and curved linear-array echoendoscope for small gastrointestinal subepithelial lesions. Endosc Int Open. 2015;3:E161–4.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Puli SR, Reddy JB, Bechtold ML, et al. Staging accuracy of esophageal cancer by endoscopic ultrasound: a meta-analysis and systematic review. World J Gastroenterol. 2008;14:1479–90.CrossRefPubMedPubMedCentral Puli SR, Reddy JB, Bechtold ML, et al. Staging accuracy of esophageal cancer by endoscopic ultrasound: a meta-analysis and systematic review. World J Gastroenterol. 2008;14:1479–90.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat TenBerge J, Hoffman BJ, Hawes RH, et al. EUS-guided fine needle aspiration of the liver: indications, yield, and safety based on an international survey of 167 cases. Gastrointestinal Endosc. 2002;55:859–62.CrossRef TenBerge J, Hoffman BJ, Hawes RH, et al. EUS-guided fine needle aspiration of the liver: indications, yield, and safety based on an international survey of 167 cases. Gastrointestinal Endosc. 2002;55:859–62.CrossRef
32.
Zurück zum Zitat Jani BS, Rzouq F, Saligram S, et al. Endoscopic Ultrasound-guided fine-needle aspiration of pancreatic lesions: a systematic review of technical and procedural variables. North Am J Med Sci. 2016;8:1–11.CrossRef Jani BS, Rzouq F, Saligram S, et al. Endoscopic Ultrasound-guided fine-needle aspiration of pancreatic lesions: a systematic review of technical and procedural variables. North Am J Med Sci. 2016;8:1–11.CrossRef
33.
Zurück zum Zitat Eisen GM, Dominitz JA, Faigel DO, et al. Guidelines for credentialing and granting privileges for endoscopic ultrasound. Gastrointest Endosc. 2001;54:811–4.CrossRefPubMed Eisen GM, Dominitz JA, Faigel DO, et al. Guidelines for credentialing and granting privileges for endoscopic ultrasound. Gastrointest Endosc. 2001;54:811–4.CrossRefPubMed
34.
Zurück zum Zitat Shahidi N, Ou G, Lam E, et al. When trainees reach competency in performing endoscopic ultrasound: a systematic review. Endosc Int Open. 2017;5:E239-e243.PubMedPubMedCentral Shahidi N, Ou G, Lam E, et al. When trainees reach competency in performing endoscopic ultrasound: a systematic review. Endosc Int Open. 2017;5:E239-e243.PubMedPubMedCentral
35.
Zurück zum Zitat Weston BR, Bhutani MS. Optimizing diagnostic yield for EUS-guided sampling of solid pancreatic lesions: a technical review. Gastroenterol Hepatol. 2013;9:352–63. Weston BR, Bhutani MS. Optimizing diagnostic yield for EUS-guided sampling of solid pancreatic lesions: a technical review. Gastroenterol Hepatol. 2013;9:352–63.
36.
Zurück zum Zitat Gimeno-Garcia AZ, Elwassief A, Paquin SC, et al. Endoscopic ultrasound-guided fine needle aspiration cytology and biopsy in the evaluation of lymphoma. Endosc Ultrasound. 2012;1:17–22.CrossRefPubMedPubMedCentral Gimeno-Garcia AZ, Elwassief A, Paquin SC, et al. Endoscopic ultrasound-guided fine needle aspiration cytology and biopsy in the evaluation of lymphoma. Endosc Ultrasound. 2012;1:17–22.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Carrara S, Anderloni A, Jovani M, et al. A prospective randomized study comparing 25-G and 22-G needles of a new platform for endoscopic ultrasound-guided fine needle aspiration of solid masses. Dig Liver Dis. 2016;48:49–54.CrossRefPubMed Carrara S, Anderloni A, Jovani M, et al. A prospective randomized study comparing 25-G and 22-G needles of a new platform for endoscopic ultrasound-guided fine needle aspiration of solid masses. Dig Liver Dis. 2016;48:49–54.CrossRefPubMed
38.
Zurück zum Zitat Camellini L, Carlinfante G, Azzolini F, et al. A randomized clinical trial comparing 22G and 25G needles in endoscopic ultrasound-guided fine-needle aspiration of solid lesions. Endoscopy. 2011;43:709–15.CrossRefPubMed Camellini L, Carlinfante G, Azzolini F, et al. A randomized clinical trial comparing 22G and 25G needles in endoscopic ultrasound-guided fine-needle aspiration of solid lesions. Endoscopy. 2011;43:709–15.CrossRefPubMed
39.
Zurück zum Zitat Vilmann P, Saftoiu A, Hollerbach S, et al. Multicenter randomized controlled trial comparing the performance of 22 gauge versus 25 gauge EUS-FNA needles in solid masses. Scand J Gastroenterol. 2013;48:877–83.CrossRefPubMed Vilmann P, Saftoiu A, Hollerbach S, et al. Multicenter randomized controlled trial comparing the performance of 22 gauge versus 25 gauge EUS-FNA needles in solid masses. Scand J Gastroenterol. 2013;48:877–83.CrossRefPubMed
40.
Zurück zum Zitat Lee JH, Stewart J, Ross WA, et al. Blinded prospective comparison of the performance of 22-gauge and 25-gauge needles in endoscopic ultrasound-guided fine needle aspiration of the pancreas and peri-pancreatic lesions. Dig Dis Sci. 2009;54:2274–81.CrossRefPubMed Lee JH, Stewart J, Ross WA, et al. Blinded prospective comparison of the performance of 22-gauge and 25-gauge needles in endoscopic ultrasound-guided fine needle aspiration of the pancreas and peri-pancreatic lesions. Dig Dis Sci. 2009;54:2274–81.CrossRefPubMed
41.
Zurück zum Zitat Siddiqui UD, Rossi F, Rosenthal LS, et al. EUS-guided FNA of solid pancreatic masses: a prospective, randomized trial comparing 22-gauge and 25-gauge needles. Gastrointest Endosc. 2009;70:1093–7.CrossRefPubMed Siddiqui UD, Rossi F, Rosenthal LS, et al. EUS-guided FNA of solid pancreatic masses: a prospective, randomized trial comparing 22-gauge and 25-gauge needles. Gastrointest Endosc. 2009;70:1093–7.CrossRefPubMed
42.
Zurück zum Zitat Yusuf TE, Ho S, Pavey DA, et al. Retrospective analysis of the utility of endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA) in pancreatic masses, using a 22-gauge or 25-gauge needle system: a multicenter experience. Endoscopy. 2009;41:445–8.CrossRefPubMed Yusuf TE, Ho S, Pavey DA, et al. Retrospective analysis of the utility of endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA) in pancreatic masses, using a 22-gauge or 25-gauge needle system: a multicenter experience. Endoscopy. 2009;41:445–8.CrossRefPubMed
43.
Zurück zum Zitat Siddiqui AA, Lyles T, Avula H, et al. Endoscopic ultrasound-guided fine needle aspiration of pancreatic masses in a veteran population: comparison of results with 22- and 25-gauge needles. Pancreas. 2010;39:685–6.CrossRefPubMed Siddiqui AA, Lyles T, Avula H, et al. Endoscopic ultrasound-guided fine needle aspiration of pancreatic masses in a veteran population: comparison of results with 22- and 25-gauge needles. Pancreas. 2010;39:685–6.CrossRefPubMed
44.
Zurück zum Zitat Camellini L, Carlinfante G, Azzolini F, et al. A randomized clinical trial comparing 22G and 25G needles in endoscopic ultrasound-guided fine-needle aspiration of solid lesions. Endoscopy. 2011;43:709–15.CrossRefPubMed Camellini L, Carlinfante G, Azzolini F, et al. A randomized clinical trial comparing 22G and 25G needles in endoscopic ultrasound-guided fine-needle aspiration of solid lesions. Endoscopy. 2011;43:709–15.CrossRefPubMed
45.
Zurück zum Zitat Uehara H, Ikezawa K, Kawada N, et al. Diagnostic accuracy of endoscopic ultrasound-guided fine needle aspiration for suspected pancreatic malignancy in relation to the size of lesions. J Gastroenterol Hepatol (Aust). 2011;26:1256–61.CrossRef Uehara H, Ikezawa K, Kawada N, et al. Diagnostic accuracy of endoscopic ultrasound-guided fine needle aspiration for suspected pancreatic malignancy in relation to the size of lesions. J Gastroenterol Hepatol (Aust). 2011;26:1256–61.CrossRef
46.
Zurück zum Zitat Madhoun MF, Wani SB, Rastogi A, et al. The diagnostic accuracy of 22-gauge and 25-gauge needles in endoscopic ultrasound-guided fine needle aspiration of solid pancreatic lesions: a meta-analysis. Endoscopy. 2013;45:86–92.CrossRefPubMed Madhoun MF, Wani SB, Rastogi A, et al. The diagnostic accuracy of 22-gauge and 25-gauge needles in endoscopic ultrasound-guided fine needle aspiration of solid pancreatic lesions: a meta-analysis. Endoscopy. 2013;45:86–92.CrossRefPubMed
47.
Zurück zum Zitat Affolter KE, Schmidt RL, Matynia AP, et al. Needle size has only a limited effect on outcomes in EUS-guided fine needle aspiration: a systematic review and meta-analysis. Dig Dis Sci. 2013;58:1026–34.CrossRefPubMed Affolter KE, Schmidt RL, Matynia AP, et al. Needle size has only a limited effect on outcomes in EUS-guided fine needle aspiration: a systematic review and meta-analysis. Dig Dis Sci. 2013;58:1026–34.CrossRefPubMed
48.
Zurück zum Zitat Song TJ, Kim JH, Lee SS, et al. The prospective randomized, controlled trial of endoscopic ultrasound-guided fine-needle aspiration using 22G and 19G aspiration needles for solid pancreatic or peripancreatic masses. Am J Gastroenterol. 2010;105:1739–45.CrossRefPubMed Song TJ, Kim JH, Lee SS, et al. The prospective randomized, controlled trial of endoscopic ultrasound-guided fine-needle aspiration using 22G and 19G aspiration needles for solid pancreatic or peripancreatic masses. Am J Gastroenterol. 2010;105:1739–45.CrossRefPubMed
49.
Zurück zum Zitat Itoi T, Itokawa F, Sofuni A, et al. Puncture of solid pancreatic tumors guided by endoscopic ultrasonography: a pilot study series comparing trucut and 19-gauge and 22-gauge aspiration needles. Endoscopy. 2005;37:362–6.CrossRefPubMed Itoi T, Itokawa F, Sofuni A, et al. Puncture of solid pancreatic tumors guided by endoscopic ultrasonography: a pilot study series comparing trucut and 19-gauge and 22-gauge aspiration needles. Endoscopy. 2005;37:362–6.CrossRefPubMed
50.
Zurück zum Zitat Hasan M, Ramesh J, Bang JY, et al. 100 multi-center randomized trial comparing the 19G and 25G needles for EUS-guided FNA of large solid pancreatic mass lesions. Gastrointest Endosc. 2014;9(5, Supplement):AB112.CrossRef Hasan M, Ramesh J, Bang JY, et al. 100 multi-center randomized trial comparing the 19G and 25G needles for EUS-guided FNA of large solid pancreatic mass lesions. Gastrointest Endosc. 2014;9(5, Supplement):AB112.CrossRef
51.
Zurück zum Zitat Wang K-X, Ben Q-W, Jin Z-D, et al. Assessment of morbidity and mortality associated with EUS-guided FNA: a systematic review. Gastrointest Endosc. 2011;73:283–90.CrossRefPubMed Wang K-X, Ben Q-W, Jin Z-D, et al. Assessment of morbidity and mortality associated with EUS-guided FNA: a systematic review. Gastrointest Endosc. 2011;73:283–90.CrossRefPubMed
52.
Zurück zum Zitat Gress F, Michael H, Gelrud D, et al. EUS-guided fine-needle aspiration of the pancreas: evaluation of pancreatitis as a complication. Gastrointest Endosc. 2002;56:864–7.CrossRefPubMed Gress F, Michael H, Gelrud D, et al. EUS-guided fine-needle aspiration of the pancreas: evaluation of pancreatitis as a complication. Gastrointest Endosc. 2002;56:864–7.CrossRefPubMed
53.
Zurück zum Zitat Sakamoto H, Kitano M, Komaki T, et al. Prospective comparative study of the EUS guided 25-gauge FNA needle with the 19-gauge Trucut needle and 22-gauge FNA needle in patients with solid pancreatic masses. J Gastroenterol Hepatol (Aust). 2009;24:384–90.CrossRef Sakamoto H, Kitano M, Komaki T, et al. Prospective comparative study of the EUS guided 25-gauge FNA needle with the 19-gauge Trucut needle and 22-gauge FNA needle in patients with solid pancreatic masses. J Gastroenterol Hepatol (Aust). 2009;24:384–90.CrossRef
54.
Zurück zum Zitat Gerke H, Rizk MK, Vanderheyden AD, et al. Randomized study comparing endoscopic ultrasound-guided Trucut biopsy and fine needle aspiration with high suction. Cytopathology. 2010;21:44–51.CrossRefPubMed Gerke H, Rizk MK, Vanderheyden AD, et al. Randomized study comparing endoscopic ultrasound-guided Trucut biopsy and fine needle aspiration with high suction. Cytopathology. 2010;21:44–51.CrossRefPubMed
55.
Zurück zum Zitat Varadarajulu S, Fraig M, Schmulewitz N, et al. Comparison of EUS-guided 19-gauge trucut needle biopsy with EUS-guided fine-needle aspiration. Endoscopy. 2004;36:397–401.CrossRefPubMed Varadarajulu S, Fraig M, Schmulewitz N, et al. Comparison of EUS-guided 19-gauge trucut needle biopsy with EUS-guided fine-needle aspiration. Endoscopy. 2004;36:397–401.CrossRefPubMed
56.
Zurück zum Zitat Wittmann J, Kocjan G, Sgouros SN, et al. Endoscopic ultrasound-guided tissue sampling by combined fine needle aspiration and trucut needle biopsy: a prospective study. Cytopathology. 2006;17:27–33.CrossRefPubMed Wittmann J, Kocjan G, Sgouros SN, et al. Endoscopic ultrasound-guided tissue sampling by combined fine needle aspiration and trucut needle biopsy: a prospective study. Cytopathology. 2006;17:27–33.CrossRefPubMed
57.
Zurück zum Zitat Larghi A, Verna EC, Stavropoulos SN, et al. EUS-guided trucut needle biopsies in patients with solid pancreatic masses: a prospective study. Gastrointest Endosc. 2004;59:185–90.CrossRefPubMed Larghi A, Verna EC, Stavropoulos SN, et al. EUS-guided trucut needle biopsies in patients with solid pancreatic masses: a prospective study. Gastrointest Endosc. 2004;59:185–90.CrossRefPubMed
58.
Zurück zum Zitat Aithal GP, Anagnostropoulos GK, Tam W, et al. EUS-guided tissue sampling: comparison of “dual sampling” (trucut biopsy plus FNA) with “sequential sampling” (trucut biopsy and then FNA as required). Endoscopy. 2007;39:725–30.CrossRefPubMed Aithal GP, Anagnostropoulos GK, Tam W, et al. EUS-guided tissue sampling: comparison of “dual sampling” (trucut biopsy plus FNA) with “sequential sampling” (trucut biopsy and then FNA as required). Endoscopy. 2007;39:725–30.CrossRefPubMed
59.
Zurück zum Zitat Berger LPV, Scheffer RCH, Weusten BLAM, et al. The additional value of EUS-guided Tru-cut biopsy to EUS-guided FNA in patients with mediastinal lesions. Gastrointest Endosc. 2009;69:1045–51.CrossRefPubMed Berger LPV, Scheffer RCH, Weusten BLAM, et al. The additional value of EUS-guided Tru-cut biopsy to EUS-guided FNA in patients with mediastinal lesions. Gastrointest Endosc. 2009;69:1045–51.CrossRefPubMed
60.
Zurück zum Zitat Cho CM, Al-Haddad M, LeBlanc JK, et al. Rescue Endoscopic Ultrasound (EUS)-guided trucut biopsy following suboptimal EUS-guided fine needle aspiration for mediastinal lesions. Gut and Liver. 2013;7:150–6.CrossRefPubMedPubMedCentral Cho CM, Al-Haddad M, LeBlanc JK, et al. Rescue Endoscopic Ultrasound (EUS)-guided trucut biopsy following suboptimal EUS-guided fine needle aspiration for mediastinal lesions. Gut and Liver. 2013;7:150–6.CrossRefPubMedPubMedCentral
61.
Zurück zum Zitat Dewitt J, Emerson RE, Sherman S, et al. Endoscopic ultrasound-guided trucut biopsy of gastrointestinal mesenchymal tumor. Surg Endosc Other Interv Tech. 2011;25:2192–202.CrossRef Dewitt J, Emerson RE, Sherman S, et al. Endoscopic ultrasound-guided trucut biopsy of gastrointestinal mesenchymal tumor. Surg Endosc Other Interv Tech. 2011;25:2192–202.CrossRef
62.
Zurück zum Zitat Ginès A, Wiersema MJ, Clain JE, et al. Prospective study of a trucut needle for performing EUS-guided biopsy with EUS-guided FNA rescue. Gastrointest Endosc. 2005;62:597–601.CrossRefPubMed Ginès A, Wiersema MJ, Clain JE, et al. Prospective study of a trucut needle for performing EUS-guided biopsy with EUS-guided FNA rescue. Gastrointest Endosc. 2005;62:597–601.CrossRefPubMed
63.
Zurück zum Zitat Ribeiro A, Pereira D, Escalón MP, et al. EUS-guided biopsy for the diagnosis and classification of lymphoma. Gastrointest Endosc. 2010;71:851–5.CrossRefPubMed Ribeiro A, Pereira D, Escalón MP, et al. EUS-guided biopsy for the diagnosis and classification of lymphoma. Gastrointest Endosc. 2010;71:851–5.CrossRefPubMed
64.
Zurück zum Zitat Yun SS, Remotti H, Vazquez MF, et al. Endoscopic ultrasound-guided biopsies of pancreatic masses: comparison between fine needle aspirations and needle core biopsies. Diagn Cytopathol. 2007;35:276–82.CrossRefPubMed Yun SS, Remotti H, Vazquez MF, et al. Endoscopic ultrasound-guided biopsies of pancreatic masses: comparison between fine needle aspirations and needle core biopsies. Diagn Cytopathol. 2007;35:276–82.CrossRefPubMed
65.
Zurück zum Zitat Alatawi A, Beuvon F, Grabar S, et al. Comparison of 22G reverse-beveled versus standard needle for endoscopic ultrasound-guided sampling of solid pancreatic lesions. United Euro Gastroenterol J. 2015;3:343–52.CrossRef Alatawi A, Beuvon F, Grabar S, et al. Comparison of 22G reverse-beveled versus standard needle for endoscopic ultrasound-guided sampling of solid pancreatic lesions. United Euro Gastroenterol J. 2015;3:343–52.CrossRef
66.
Zurück zum Zitat Kim GH, Cho YK, Kim EY, et al. Comparison of 22-gauge aspiration needle with 22-gauge biopsy needle in endoscopic ultrasonography-guided subepithelial tumor sampling. Scand J Gastroenterol. 2014;49:347–54.CrossRefPubMed Kim GH, Cho YK, Kim EY, et al. Comparison of 22-gauge aspiration needle with 22-gauge biopsy needle in endoscopic ultrasonography-guided subepithelial tumor sampling. Scand J Gastroenterol. 2014;49:347–54.CrossRefPubMed
67.
Zurück zum Zitat Bang JY, Hawes R, Varadarajulu S. A meta-analysis comparing ProCore and standard fine-needle aspiration needles for endoscopic ultrasound-guided tissue acquisition. Endoscopy. 2016;48:339–49.PubMed Bang JY, Hawes R, Varadarajulu S. A meta-analysis comparing ProCore and standard fine-needle aspiration needles for endoscopic ultrasound-guided tissue acquisition. Endoscopy. 2016;48:339–49.PubMed
68.
Zurück zum Zitat Rodrigues-Pinto E, Jalaj S, Grimm IS, et al. Impact of EUS-guided fine-needle biopsy sampling with a new core needle on the need for onsite cytopathologic assessment: a preliminary study. Gastrointest Endosc. 2016;84:1040–6.CrossRefPubMed Rodrigues-Pinto E, Jalaj S, Grimm IS, et al. Impact of EUS-guided fine-needle biopsy sampling with a new core needle on the need for onsite cytopathologic assessment: a preliminary study. Gastrointest Endosc. 2016;84:1040–6.CrossRefPubMed
69.
Zurück zum Zitat Kandel P, Tranesh G, Nassar A, et al. EUS-guided fine needle biopsy sampling using a novel fork-tip needle: a case-control study. Gastrointest Endosc. 2016;84:1034–9.CrossRefPubMed Kandel P, Tranesh G, Nassar A, et al. EUS-guided fine needle biopsy sampling using a novel fork-tip needle: a case-control study. Gastrointest Endosc. 2016;84:1034–9.CrossRefPubMed
70.
Zurück zum Zitat Nayar MK, Paranandi B, Dawwas MF, et al. Comparison of the diagnostic performance of 2 core biopsy needles for EUS-guided tissue acquisition from solid pancreatic lesions. Gastrointest Endosc. 2017;85:1017–24.CrossRefPubMed Nayar MK, Paranandi B, Dawwas MF, et al. Comparison of the diagnostic performance of 2 core biopsy needles for EUS-guided tissue acquisition from solid pancreatic lesions. Gastrointest Endosc. 2017;85:1017–24.CrossRefPubMed
71.
Zurück zum Zitat Bang JY, Hebert-Magee S, Hasan MK, et al. Endoscopic ultrasonography-guided biopsy using a Franseen needle design: initial assessment. Dig Endosc. 2017;29:338–46.CrossRefPubMed Bang JY, Hebert-Magee S, Hasan MK, et al. Endoscopic ultrasonography-guided biopsy using a Franseen needle design: initial assessment. Dig Endosc. 2017;29:338–46.CrossRefPubMed
72.
Zurück zum Zitat El H II, Wu H, Reuss S, et al. Prospective assessment of the performance of a new fine needle biopsy device for EUS-guided sampling of solid lesions. Clinical Endosc. 2018;51:576.CrossRef El H II, Wu H, Reuss S, et al. Prospective assessment of the performance of a new fine needle biopsy device for EUS-guided sampling of solid lesions. Clinical Endosc. 2018;51:576.CrossRef
73.
Zurück zum Zitat Bang JY, Hebert-Magee S, Navaneethan U, et al. Randomized trial comparing the Franseen and Fork-tip needles for EUS-guided fine-needle biopsy sampling of solid pancreatic mass lesions. Gastrointest Endosc. 2018;87:1432–8.CrossRefPubMed Bang JY, Hebert-Magee S, Navaneethan U, et al. Randomized trial comparing the Franseen and Fork-tip needles for EUS-guided fine-needle biopsy sampling of solid pancreatic mass lesions. Gastrointest Endosc. 2018;87:1432–8.CrossRefPubMed
74.
Zurück zum Zitat Bang JY, Hebert-Magee S, Navaneethan U, et al. Randomized trial comparing the franseen and fork-tip needles for EUS-guided fine-needle biopsy sampling of solid pancreatic mass lesions. Gastrointest Endosc. 2018;87:1432–8.CrossRefPubMed Bang JY, Hebert-Magee S, Navaneethan U, et al. Randomized trial comparing the franseen and fork-tip needles for EUS-guided fine-needle biopsy sampling of solid pancreatic mass lesions. Gastrointest Endosc. 2018;87:1432–8.CrossRefPubMed
75.
Zurück zum Zitat Al-Haddad M. Fine-needle biopsy sampling under EUS guidance: is one needle tip really better than the other? Gastrointest Endosc. 2018;87:1163.CrossRefPubMed Al-Haddad M. Fine-needle biopsy sampling under EUS guidance: is one needle tip really better than the other? Gastrointest Endosc. 2018;87:1163.CrossRefPubMed
76.
Zurück zum Zitat Iglesias-Garcia J, Dominguez-Munoz JE, Abdulkader I, et al. Influence of on-site cytopathology evaluation on the diagnostic accuracy of endoscopic ultrasound-guided fine needle aspiration (EUS–FNA) of solid pancreatic masses. Am J Gastroenterol. 2011;106:1705–10.CrossRefPubMed Iglesias-Garcia J, Dominguez-Munoz JE, Abdulkader I, et al. Influence of on-site cytopathology evaluation on the diagnostic accuracy of endoscopic ultrasound-guided fine needle aspiration (EUS–FNA) of solid pancreatic masses. Am J Gastroenterol. 2011;106:1705–10.CrossRefPubMed
77.
Zurück zum Zitat Wani S, Mullady D, Early DS, et al. The clinical impact of immediate on-site cytopathology evaluation during endoscopic ultrasound-guided fine needle aspiration of pancreatic masses: a prospective multicenter randomized controlled trial. Am J Gastroenterol. 2015;110:1429.CrossRefPubMed Wani S, Mullady D, Early DS, et al. The clinical impact of immediate on-site cytopathology evaluation during endoscopic ultrasound-guided fine needle aspiration of pancreatic masses: a prospective multicenter randomized controlled trial. Am J Gastroenterol. 2015;110:1429.CrossRefPubMed
78.
Zurück zum Zitat Aadam AA, Wani S, Amick A, et al. A randomized controlled cross-over trial and cost analysis comparing endoscopic ultrasound fine needle aspiration and fine needle biopsy. Endosc Int Open. 2016;4:E497–505.CrossRefPubMedPubMedCentral Aadam AA, Wani S, Amick A, et al. A randomized controlled cross-over trial and cost analysis comparing endoscopic ultrasound fine needle aspiration and fine needle biopsy. Endosc Int Open. 2016;4:E497–505.CrossRefPubMedPubMedCentral
79.
Zurück zum Zitat Rodrigues-Pinto E, Jalaj S, Grimm IS, et al. Impact of EUS-guided fine-needle biopsy sampling with a new core needle on the need for onsite cytopathologic assessment: a preliminary study. Gastrointest Endosc. 2016;84:1040–6.CrossRefPubMed Rodrigues-Pinto E, Jalaj S, Grimm IS, et al. Impact of EUS-guided fine-needle biopsy sampling with a new core needle on the need for onsite cytopathologic assessment: a preliminary study. Gastrointest Endosc. 2016;84:1040–6.CrossRefPubMed
80.
Zurück zum Zitat Fabbri C, Fuccio L, Fornelli A, et al. The presence of rapid on-site evaluation did not increase the adequacy and diagnostic accuracy of endoscopic ultrasound-guided tissue acquisition of solid pancreatic lesions with core needle. Surg Endosc. 2017;31:225–30.CrossRefPubMed Fabbri C, Fuccio L, Fornelli A, et al. The presence of rapid on-site evaluation did not increase the adequacy and diagnostic accuracy of endoscopic ultrasound-guided tissue acquisition of solid pancreatic lesions with core needle. Surg Endosc. 2017;31:225–30.CrossRefPubMed
81.
Zurück zum Zitat Schmidt RL, Witt BL, Matynia AP, et al. Rapid on-site evaluation increases endoscopic ultrasound-guided fine-needle aspiration adequacy for pancreatic lesions. Dig Dis Sci. 2013;58:872–82.CrossRefPubMed Schmidt RL, Witt BL, Matynia AP, et al. Rapid on-site evaluation increases endoscopic ultrasound-guided fine-needle aspiration adequacy for pancreatic lesions. Dig Dis Sci. 2013;58:872–82.CrossRefPubMed
82.
Zurück zum Zitat Wani S, Early D, Kunkel J, et al. Diagnostic yield of malignancy during EUS-guided FNA of solid lesions with and without a stylet: a prospective, single blind, randomized, controlled trial. Gastrointest Endosc. 2012;76:328–35.CrossRefPubMed Wani S, Early D, Kunkel J, et al. Diagnostic yield of malignancy during EUS-guided FNA of solid lesions with and without a stylet: a prospective, single blind, randomized, controlled trial. Gastrointest Endosc. 2012;76:328–35.CrossRefPubMed
83.
Zurück zum Zitat Kim JH, Park SW, Kim MK, et al. Meta-analysis for cyto-pathological outcomes in endoscopic ultrasonography-guided fine-needle aspiration with and without the stylet. Dig Dis Sci. 2016;61:2175–84.CrossRefPubMed Kim JH, Park SW, Kim MK, et al. Meta-analysis for cyto-pathological outcomes in endoscopic ultrasonography-guided fine-needle aspiration with and without the stylet. Dig Dis Sci. 2016;61:2175–84.CrossRefPubMed
84.
Zurück zum Zitat Abe Y, Kawakami H, Oba K, et al. Effect of a stylet on a histological specimen in EUS-guided fine-needle tissue acquisition by using 22-gauge needles: a multicenter, prospective, randomized, controlled trial. Gastrointest Endosc. 2015;82(837–844):e831. Abe Y, Kawakami H, Oba K, et al. Effect of a stylet on a histological specimen in EUS-guided fine-needle tissue acquisition by using 22-gauge needles: a multicenter, prospective, randomized, controlled trial. Gastrointest Endosc. 2015;82(837–844):e831.
85.
Zurück zum Zitat Lee JK, Choi JH, Lee KH, et al. A prospective, comparative trial to optimize sampling techniques in EUS-guided FNA of solid pancreatic masses. Gastrointest Endosc. 2013;77:745–51.CrossRefPubMed Lee JK, Choi JH, Lee KH, et al. A prospective, comparative trial to optimize sampling techniques in EUS-guided FNA of solid pancreatic masses. Gastrointest Endosc. 2013;77:745–51.CrossRefPubMed
86.
Zurück zum Zitat Kin T, Katanuma A, Yane K, et al. Diagnostic ability of EUS-FNA for pancreatic solid lesions with conventional 22-gauge needle using the slow pull technique: a prospective study. Scand J Gastroenterol. 2015;50:900–7.CrossRefPubMed Kin T, Katanuma A, Yane K, et al. Diagnostic ability of EUS-FNA for pancreatic solid lesions with conventional 22-gauge needle using the slow pull technique: a prospective study. Scand J Gastroenterol. 2015;50:900–7.CrossRefPubMed
87.
Zurück zum Zitat Attam R, Arain MA, Bloechl SJ, et al. “Wet suction technique (WEST)”: a novel way to enhance the quality of EUS-FNA aspirate. Results of a prospective, single-blind, randomized, controlled trial using a 22-gauge needle for EUS-FNA of solid lesions. Gastrointest Endosc. 2015;81:1401–7.CrossRefPubMed Attam R, Arain MA, Bloechl SJ, et al. “Wet suction technique (WEST)”: a novel way to enhance the quality of EUS-FNA aspirate. Results of a prospective, single-blind, randomized, controlled trial using a 22-gauge needle for EUS-FNA of solid lesions. Gastrointest Endosc. 2015;81:1401–7.CrossRefPubMed
88.
Zurück zum Zitat Mennone A, Nathanson MH. Needle-based confocal laser endomicroscopy to assess liver histology in vivo. Gastrointest Endosc. 2011;73:338–44.CrossRefPubMed Mennone A, Nathanson MH. Needle-based confocal laser endomicroscopy to assess liver histology in vivo. Gastrointest Endosc. 2011;73:338–44.CrossRefPubMed
89.
Zurück zum Zitat Becker V, Wallace MB, Fockens P, et al. Needle-based confocal endomicroscopy for in vivo histology of intra-abdominal organs: first results in a porcine model (with videos). Gastrointest Endosc. 2010;71:1260–6.CrossRefPubMed Becker V, Wallace MB, Fockens P, et al. Needle-based confocal endomicroscopy for in vivo histology of intra-abdominal organs: first results in a porcine model (with videos). Gastrointest Endosc. 2010;71:1260–6.CrossRefPubMed
90.
Zurück zum Zitat Giovannini M, Caillol F, Monges G, et al. Endoscopic ultrasound-guided needle-based confocal laser endomicroscopy in solid pancreatic masses. Endoscopy. 2016;48:892–8.CrossRefPubMed Giovannini M, Caillol F, Monges G, et al. Endoscopic ultrasound-guided needle-based confocal laser endomicroscopy in solid pancreatic masses. Endoscopy. 2016;48:892–8.CrossRefPubMed
91.
Zurück zum Zitat Kongkam P, Pittayanon R, Sampatanukul P, et al. Endoscopic ultrasound-guided needle-based confocal laser endomicroscopy for diagnosis of solid pancreatic lesions (ENES): a pilot study. Endosc Int Open. 2016;4:E17–23.PubMed Kongkam P, Pittayanon R, Sampatanukul P, et al. Endoscopic ultrasound-guided needle-based confocal laser endomicroscopy for diagnosis of solid pancreatic lesions (ENES): a pilot study. Endosc Int Open. 2016;4:E17–23.PubMed
92.
Zurück zum Zitat Konda VJA, Aslanian HR, Wallace MB, et al. First assessment of needle-based confocal laser endomicroscopy during EUS–FNA procedures of the pancreas (with videos). Gastrointest Endosc. 2011;74:1049–60.CrossRefPubMed Konda VJA, Aslanian HR, Wallace MB, et al. First assessment of needle-based confocal laser endomicroscopy during EUS–FNA procedures of the pancreas (with videos). Gastrointest Endosc. 2011;74:1049–60.CrossRefPubMed
93.
Zurück zum Zitat Karstensen JG, Cartana T, Constantinescu C, et al. Endoscopic ultrasound guided needle-based confocal laser endomicroscopy in solid pancreatic masses—a prospective validation study. Endosc Int Open. 2018;6:E78–85.CrossRefPubMedPubMedCentral Karstensen JG, Cartana T, Constantinescu C, et al. Endoscopic ultrasound guided needle-based confocal laser endomicroscopy in solid pancreatic masses—a prospective validation study. Endosc Int Open. 2018;6:E78–85.CrossRefPubMedPubMedCentral
94.
Zurück zum Zitat Bhutani MS, Koduru P, Joshi V, et al. EUS-guided needle-based confocal laser endomicroscopy: a novel technique with emerging applications. Gastroenterol Hepatol. 2015;11:235. Bhutani MS, Koduru P, Joshi V, et al. EUS-guided needle-based confocal laser endomicroscopy: a novel technique with emerging applications. Gastroenterol Hepatol. 2015;11:235.
95.
Zurück zum Zitat Chang KJ, Samarasena JB, Nakai Y, et al. Mo1502 endoscopic ultrasound (EUS) guided biopsy of the pancreas surface using a novel “through-the-needle” technique: can we biopsy the pancreas without trauma to the ducts? Gastrointest Endosc. 2013;77:AB405–6.CrossRef Chang KJ, Samarasena JB, Nakai Y, et al. Mo1502 endoscopic ultrasound (EUS) guided biopsy of the pancreas surface using a novel “through-the-needle” technique: can we biopsy the pancreas without trauma to the ducts? Gastrointest Endosc. 2013;77:AB405–6.CrossRef
96.
Zurück zum Zitat Nakai Y, Isayama H, Chang KJ, et al. A pilot study of EUS-guided through-the-needle forceps biopsy (with video). Gastrointest Endosc. 2016;84:158–62.CrossRefPubMed Nakai Y, Isayama H, Chang KJ, et al. A pilot study of EUS-guided through-the-needle forceps biopsy (with video). Gastrointest Endosc. 2016;84:158–62.CrossRefPubMed
97.
Zurück zum Zitat Shakhatreh MH, Naini SR, Brijbassie AA, et al. Use of a novel through-the-needle biopsy forceps in endoscopic ultrasound. Endosc Int Open. 2016;4:E439–42.CrossRefPubMedPubMedCentral Shakhatreh MH, Naini SR, Brijbassie AA, et al. Use of a novel through-the-needle biopsy forceps in endoscopic ultrasound. Endosc Int Open. 2016;4:E439–42.CrossRefPubMedPubMedCentral
98.
Zurück zum Zitat Kovacevic B, Karstensen JG, Havre RF, et al. Initial experience with EUS-guided microbiopsy forceps in diagnosing pancreatic cystic lesions: a multicenter feasibility study (with video). Endosc Ultrasound. 2018;7(6):383.CrossRefPubMedPubMedCentral Kovacevic B, Karstensen JG, Havre RF, et al. Initial experience with EUS-guided microbiopsy forceps in diagnosing pancreatic cystic lesions: a multicenter feasibility study (with video). Endosc Ultrasound. 2018;7(6):383.CrossRefPubMedPubMedCentral
99.
Zurück zum Zitat Wani S, Muthusamy VR, McGrath CM, et al. AGA white paper: optimizing endoscopic ultrasound-guided tissue acquisition and future directions. Clin Gastroenterol Hepatol. 2018;16:318–27.CrossRefPubMed Wani S, Muthusamy VR, McGrath CM, et al. AGA white paper: optimizing endoscopic ultrasound-guided tissue acquisition and future directions. Clin Gastroenterol Hepatol. 2018;16:318–27.CrossRefPubMed
100.
101.
Zurück zum Zitat Wei S, Lieberman D, Morrissette JJ, et al. Using “residual” FNA rinse and body fluid specimens for next-generation sequencing: an institutional experience. Cancer Cytopathol. 2016;124:324–9.CrossRefPubMed Wei S, Lieberman D, Morrissette JJ, et al. Using “residual” FNA rinse and body fluid specimens for next-generation sequencing: an institutional experience. Cancer Cytopathol. 2016;124:324–9.CrossRefPubMed
102.
Zurück zum Zitat Aisner DL, Rumery MD, Merrick DT, et al. Do more with less: tips and techniques for maximizing small biopsy and cytology specimens for molecular and ancillary testing: the university of Colorado experience. Arch Pathol lab Med. 2016;140(11):1206–20.CrossRef Aisner DL, Rumery MD, Merrick DT, et al. Do more with less: tips and techniques for maximizing small biopsy and cytology specimens for molecular and ancillary testing: the university of Colorado experience. Arch Pathol lab Med. 2016;140(11):1206–20.CrossRef
103.
Zurück zum Zitat Roy-Chowdhuri S, Chen H, Singh RR, et al. Concurrent fine needle aspirations and core needle biopsies: a comparative study of substrates for next-generation sequencing in solid organ malignancies. Mod Pathol. 2017;30:499–508.CrossRef Roy-Chowdhuri S, Chen H, Singh RR, et al. Concurrent fine needle aspirations and core needle biopsies: a comparative study of substrates for next-generation sequencing in solid organ malignancies. Mod Pathol. 2017;30:499–508.CrossRef
104.
Zurück zum Zitat Rodriguez SA, Impey SD, Pelz C, et al. RNA sequencing distinguishes benign from malignant pancreatic lesions sampled by EUS-guided FNA. Gastrointest Endosc. 2016;84:252–8.CrossRefPubMed Rodriguez SA, Impey SD, Pelz C, et al. RNA sequencing distinguishes benign from malignant pancreatic lesions sampled by EUS-guided FNA. Gastrointest Endosc. 2016;84:252–8.CrossRefPubMed
105.
Zurück zum Zitat Roy-Chowdhuri S, Stewart J. Preanalytic variables in cytology: lessons learned from next-generation sequencing-the MD anderson experience. Arch Pathol Lab Med. 2016;140(11):1191–9.CrossRef Roy-Chowdhuri S, Stewart J. Preanalytic variables in cytology: lessons learned from next-generation sequencing-the MD anderson experience. Arch Pathol Lab Med. 2016;140(11):1191–9.CrossRef
106.
Zurück zum Zitat Tiriac H, Bucobo JC, Tzimas D, et al. Successful creation of pancreatic cancer organoids by means of EUS-guided fine-needle biopsy sampling for personalized cancer treatment. Gastrointest Endosc. 2018;87:1474–80.CrossRefPubMedPubMedCentral Tiriac H, Bucobo JC, Tzimas D, et al. Successful creation of pancreatic cancer organoids by means of EUS-guided fine-needle biopsy sampling for personalized cancer treatment. Gastrointest Endosc. 2018;87:1474–80.CrossRefPubMedPubMedCentral
107.
Zurück zum Zitat Lai Y, Wei X, Lin S, et al. Current status and perspectives of patient-derived xenograft models in cancer research. J Hematol Oncol. 2017;10:106.CrossRefPubMedPubMedCentral Lai Y, Wei X, Lin S, et al. Current status and perspectives of patient-derived xenograft models in cancer research. J Hematol Oncol. 2017;10:106.CrossRefPubMedPubMedCentral
108.
Zurück zum Zitat Kim MP, Evans DB, Wang H, et al. Generation of orthotopic and heterotopic human pancreatic cancer xenografts in immunodeficient mice. Nat Protoc. 2009;4:1670–80.CrossRefPubMedPubMedCentral Kim MP, Evans DB, Wang H, et al. Generation of orthotopic and heterotopic human pancreatic cancer xenografts in immunodeficient mice. Nat Protoc. 2009;4:1670–80.CrossRefPubMedPubMedCentral
109.
Zurück zum Zitat Kandel P, Tranesh G, Nassar A, et al. EUS-guided fine needle biopsy sampling using a novel fork-tip needle: a case-control study. Gastrointest Endosc. 2016;84:1034–9.CrossRefPubMed Kandel P, Tranesh G, Nassar A, et al. EUS-guided fine needle biopsy sampling using a novel fork-tip needle: a case-control study. Gastrointest Endosc. 2016;84:1034–9.CrossRefPubMed
110.
Zurück zum Zitat Nayar MK, Paranandi B, Dawwas MF, et al. Comparison of the diagnostic performance of 2 core biopsy needles for EUS-guided tissue acquisition from solid pancreatic lesions. Gastrointest Endosc. 2017;85:1017–24.CrossRefPubMed Nayar MK, Paranandi B, Dawwas MF, et al. Comparison of the diagnostic performance of 2 core biopsy needles for EUS-guided tissue acquisition from solid pancreatic lesions. Gastrointest Endosc. 2017;85:1017–24.CrossRefPubMed
111.
Zurück zum Zitat Kandel P, Nassar A, Graddy C, et al. Mo1345 whole exome sequencing and genomic profiling of pancreas tumor tissue obtained with a novelfork-tipped eus guided fine needle core biopsy: a randomized controlled trial. Gastrointest Endosc. 2018;87:455–6.CrossRef Kandel P, Nassar A, Graddy C, et al. Mo1345 whole exome sequencing and genomic profiling of pancreas tumor tissue obtained with a novelfork-tipped eus guided fine needle core biopsy: a randomized controlled trial. Gastrointest Endosc. 2018;87:455–6.CrossRef
112.
Zurück zum Zitat Suzuki R, Thosani N, Annangi S, Guha S, Bhutani MS. Diagnostic yield of EUS-FNA-based cytology distinguishing malignant and benign IPMNs: a systematic review and meta-analysis. Pancreatol off J Intern Assoc Pancreatol. (IAP) 2014;14(5):380–84.CrossRef Suzuki R, Thosani N, Annangi S, Guha S, Bhutani MS. Diagnostic yield of EUS-FNA-based cytology distinguishing malignant and benign IPMNs: a systematic review and meta-analysis. Pancreatol off J Intern Assoc Pancreatol. (IAP) 2014;14(5):380–84.CrossRef
113.
Zurück zum Zitat Hebert-Magee S, Bae S, Varadarajulu S, et al. The presence of a cytopathologist increases the diagnostic accuracy of endoscopic ultrasound-guided fine needle aspiration cytology for pancreatic adenocarcinoma: a meta-analysis. Cytopathol off J Br Soc Clin Cytol. 2013;24(3):159–71.CrossRef Hebert-Magee S, Bae S, Varadarajulu S, et al. The presence of a cytopathologist increases the diagnostic accuracy of endoscopic ultrasound-guided fine needle aspiration cytology for pancreatic adenocarcinoma: a meta-analysis. Cytopathol off J Br Soc Clin Cytol. 2013;24(3):159–71.CrossRef
114.
Zurück zum Zitat Larghi A, Fuccio L, Chiarello G, et al. Fine-needle tissue acquisition from subepithelial lesions using a forward-viewing linear echoendoscope. Endoscopy. 2014;46(1):39–45.PubMed Larghi A, Fuccio L, Chiarello G, et al. Fine-needle tissue acquisition from subepithelial lesions using a forward-viewing linear echoendoscope. Endoscopy. 2014;46(1):39–45.PubMed
115.
Zurück zum Zitat Turhan N, Aydog G, Ozin Y, Cicek B, Kurt M, Oguz D. Endoscopic ultrasonography-guided fine-needle aspiration for diagnosing upper gastrointestinal submucosal lesions: a prospective study of 50 cases. Diagn Cytopathol. 2011;39(11):808–17.CrossRefPubMed Turhan N, Aydog G, Ozin Y, Cicek B, Kurt M, Oguz D. Endoscopic ultrasonography-guided fine-needle aspiration for diagnosing upper gastrointestinal submucosal lesions: a prospective study of 50 cases. Diagn Cytopathol. 2011;39(11):808–17.CrossRefPubMed
116.
Zurück zum Zitat Sadeghi A, Mohamadnejad M, Islami F, et al. Diagnostic yield of EUS-guided FNA for malignant biliary stricture: a systematic review and meta-analysis. Gastrointest Endosc. 2016;83(2):290–98.e291. Sadeghi A, Mohamadnejad M, Islami F, et al. Diagnostic yield of EUS-guided FNA for malignant biliary stricture: a systematic review and meta-analysis. Gastrointest Endosc. 2016;83(2):290–98.e291.
117.
Zurück zum Zitat Navaneethan U, Njei B, Lourdusamy V, Konjeti R, Vargo JJ, Parsi MA. Comparative effectiveness of biliary brush cytology and intraductal biopsy for detection of malignant biliary strictures: a systematic review and meta-analysis. Gastrointest Endosc. 2015;81(1):168–76.CrossRefPubMed Navaneethan U, Njei B, Lourdusamy V, Konjeti R, Vargo JJ, Parsi MA. Comparative effectiveness of biliary brush cytology and intraductal biopsy for detection of malignant biliary strictures: a systematic review and meta-analysis. Gastrointest Endosc. 2015;81(1):168–76.CrossRefPubMed
Metadaten
Titel
Recent advancement in EUS-guided fine needle sampling
verfasst von
Pujan Kandel
Michael B. Wallace
Publikationsdatum
26.02.2019
Verlag
Springer Japan
Erschienen in
Journal of Gastroenterology / Ausgabe 5/2019
Print ISSN: 0944-1174
Elektronische ISSN: 1435-5922
DOI
https://doi.org/10.1007/s00535-019-01552-2

Weitere Artikel der Ausgabe 5/2019

Journal of Gastroenterology 5/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.