Retrospective Cohort Study Open Access
Copyright ©The Author(s) 2020. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Endosc. Aug 16, 2020; 12(8): 212-219
Published online Aug 16, 2020. doi: 10.4253/wjge.v12.i8.212
Improved diagnostic yield of endoscopic ultrasound-fine needle biopsy with histology specimen processing
Lawrence Ku, Mohammad A Shahshahan, Linda A Hou, Viktor E Eysselein, Sofiya Reicher, Department of Medicine, Division of Gastroenterology and Hepatology, Harbor-UCLA Medical Center, Torrance, CA 90509, United States
ORCID number: Lawrence Ku (0000-0001-6201-7092); Mohammad A Shahshahan (0000-0003-2752-6670); Linda A Hou (0000-0001-8289-3903); Viktor E Eysselein (0000-0002-1400-8367); Sofiya Reicher (0000-0002-2983-4370).
Author contributions: Ku L and Reicher S contributed to study design, data collection, data analysis, manuscript drafting, manuscript revision, and final approval of the manuscript; Shahshahan MA contributed to data collection, data analysis, manuscript drafting, and manuscript revision; Hou LA and Eysselein VE contributed to data collection, manuscript drafting, and manuscript revision.
Supported by the National Center for Advancing Translational Sciences through University of California, Los Angeles Clinical and Translational Science Institute Grant, No. UL1TR001881-01.
Institutional review board statement: The study was approved by the Institutional Review Board, No. 31297-01.
Informed consent statement: Not applicable to this retrospective cohort study.
Conflict-of-interest statement: Sofiya Reicher is a consultant for Boston Scientific; all other authors have no conflicts of interest.
Data sharing statement: Technical appendix, statistical code, and dataset available from the corresponding author at sreicher@dhs.lacounty.gov. Consent was not obtained, but the presented data are anonymized and risk of identification is low.
STROBE statement: The authors have read the STROBE Statement checklist of items, and the manuscript was prepared and revised according to the STROBE Statement checklist of items.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Sofiya Reicher, MD, Associate Professor, Attending Doctor, Director, Department of Medicine, Division of Gastroenterology and Hepatology, Harbor-UCLA Medical Center, 21840 South Normandie Ave, Ste 850, Torrance, CA 90509, United States. sreicher@dhs.lacounty.gov
Received: March 26, 2020
Peer-review started: March 26, 2020
First decision: April 22, 2020
Revised: June 4, 2020
Accepted: July 18, 2020
Article in press: July 18, 2020
Published online: August 16, 2020
Processing time: 139 Days and 9.3 Hours

Abstract
BACKGROUND

Endoscopic ultrasound-guided fine needle biopsy (EUS-FNB) has emerged as a safe, efficacious alternative to fine needle aspiration (FNA) for tissue acquisition. EUS-FNB is reported to have higher diagnostic yield while preserving specimen tissue architecture. However, data on the optimal method of EUS-FNB specimen processing is limited.

AIM

To evaluate EUS-FNB with specimen processing as histology vs EUS-FNA cytology with regards to diagnostic yield and specimen adequacy.

METHODS

All EUS-FNA and EUS-FNB performed at our institution from July 1, 2016, to January 31, 2018, were retrospectively analyzed. We collected data on demographics, EUS findings, pathology, clinical outcomes, and procedural complications in two periods, July 2016 through March 2017, and April 2017 through January 2018, with predominant use of FNB in the second data collection time period. FNA specimens were processed as cytology with cell block technique and reviewed by a cytopathologist; FNB specimens were fixed in formalin, processed for histopathologic analysis and immunohistochemical staining, and reviewed by an anatomic pathologist. Final diagnosis was based on surgical pathology when available, repeat biopsy or imaging, and length of clinical follow up.

RESULTS

One hundred six EUS-FNA and EUS-FNB procedures were performed. FNA alone was performed in 17 patients; in 56 patients, FNB alone was done; and in 33 patients, both FNA and FNB were performed. For all indications, diagnostic yield was 47.1% (8/17) in FNA alone cases, 85.7% (48/56) in FNB alone cases, and 84.8% (28/33) in cases where both FNA and FNB were performed (P = 0.0039). Specimens were adequate for pathologic evaluation in 52.9% (9/17) of FNA alone cases, in 89.3% (50/56) of FNB alone cases, and 84.8% (28/33) in cases where FNA with FNB were performed (P = 0.0049). Tissue could not be aspirated for cytology in 10.0% (5/50) of cases where FNA was done, while in 3.4% (3/89) of FNB cases, tissue could not be obtained for histology. In patients who underwent FNA with FNB, there was a statistically significant difference in both specimen adequacy (P = 0.0455) and diagnostic yield (P = 0.0455) between the FNA and FNB specimens (processed correspondingly as cytology or histology).

CONCLUSION

EUS-FNB has a higher diagnostic yield and specimen adequacy than EUS-FNA. In our experience, specimen processing as histology may have contributed to the overall increased diagnostic yield of EUS-FNB.

Key Words: Fine needle biopsy, Endoscopic ultrasound, Fine needle aspiration, Pancreatic cancer, Histology, Cytopathology

Core tip: Endoscopic ultrasound-guided fine needle biopsy (EUS-FNB) is rapidly gaining in popularity. However, the optimal method for EUS-FNB specimen processing is not well defined, with recent studies on fine needle biopsy (FNB) varying widely in the use of histology vs cytology for FNB sample evaluation. Our data suggest that processing FNB specimens in formalin for histology, followed by evaluation by an anatomic pathologist, could contribute to overall improved diagnostic yield of EUS-FNB. An additional benefit is the decreased need for on-site cytopathology.



INTRODUCTION

Endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) is a well-established modality for tissue acquisition of a variety of lesions in the gastrointestinal tract and surrounding structures. It has low complication rates and high diagnostic yield[1,2].

However, several factors can limit the sensitivity of EUS-FNA. EUS-FNA samples are typically processed as cytology, which does not allow for preservation of tissue architecture necessary for diagnosis of diseases such as gastrointestinal stromal tumor (GIST)[3], lymphoma[4], autoimmune pancreatitis[5], and pancreatic lesions with non-hypovascular contrast-enhancement pattern on EUS[6]. The diagnostic yield of EUS-FNA may be further compromised by the limited availability of on-site cytopathologists[7-11].

EUS-guided fine needle biopsy (EUS-FNB) has emerged as an alternative to EUS-FNA for tissue acquisition, with a reported similar rate of complications[12,13]. Initial studies have demonstrated its non-inferiority and possible superiority, depending on the indication[14-19]. FNB needle tip design enables the procurement of an intact core tissue, and the preserved architecture allows for histological and immunohistochemical evaluation. Studies differ in their approach to FNB sample processing as histology vs cytology. There is limited data on which approach is preferable.

We evaluate the performance of EUS-FNB with specimen processing as histology vs EUS-FNA cytology with regards to diagnostic yield and specimen adequacy.

MATERIALS AND METHODS

Data was retrospectively collected on all patients who underwent EUS-FNA or EUS-FNB from July 1, 2016, to January 31, 2018, at our institution, a large tertiary safety-net hospital. Data was collected in two periods: July 2016 through March 2017, and April 2017 through January 2018.

Procedures were performed by three experienced endosonographers who each have performed over 1000 EUS procedures. FNA specimens were collected for cytology in Plasma-Lyte A injection solution pH 7.4 (Baxter Healthcare Corporation, Deerfield, IL, United States), processed as a cell block with the Collodion bag technique[20,21], and subsequently evaluated by a cytopathologist. FNB specimens were collected, immediately fixed in formalin, and sent to pathology, where they were processed for histopathologic analysis and immunohistochemical staining in accordance with a previously reported standardized protocol[22], and subsequently evaluated by an anatomic pathologist. Rapid on-site evaluation is not available at our institution.

Echoendoscopes used were GF-UE160-AL5, GF-UC140-AL5, GF-UC140P-AL5, and GF-UCT180 (Olympus America, Center Valley, PA, United States). EUS-FNA and FNB needles were from a variety of manufacturers: Expect FNA and Acquire FNB needles (Boston Scientific, Marlborough, MA, United States), SharkCore FNB needles (Medtronic, Sunnyvale, CA, United States), and EchoTip ProCore FNB needles (Cook Medical, Bloomington, IN, United States).

Data collected from hospital Electronic Health Records and EUS databases included patient demographics, clinical outcomes, and pathology. Procedure-related data included indications, technical aspects, and complications.

Standardized definitions of specimen adequacy and diagnostic yield were used[23]. Specimen adequacy was defined as the percentage of lesions sampled in which the specimens were from the intended site and sufficient for diagnosis by a pathologist. Acellular or hypocellular samples were considered inadequate. Diagnostic yield was defined as the percentage of lesions sampled in which a tissue diagnosis was obtained. Final diagnosis was based on surgical pathology when available, repeat biopsy or imaging, and length of clinical follow up.

The study was approved by the Institutional Review Board (#31297-01).

Statistical analysis

Statistical analysis was performed using Fisher’s exact test, Kruskal-Wallis test, and McNemar’s test, with P value < 0.05 as statistically significant. The Bonferroni correction was applied to all sub-group analyses. All analyses were performed with R, version 3.6.0 (R Core Team, Vienna, Austria), and reviewed by a biostatistician, Youngju Pak, Ph.D., from the UCLA Clinical and Translational Science Institute at Harbor-UCLA Medical Center.

RESULTS
Demographics

From July 2016 through January 2018, EUS-FNA or EUS-FNB was performed in 106 procedures in 97 patients. The mean patient age was 55.5 years (23-84), and 41.5% were males (Table 1).

Table 1 Baseline patient and procedural characteristics.
FNA alone (n = 17)FNB alone (n = 56)FNA with FNB (n = 33)P value
Age, mean (range)55.2 (30-75)55.0 (23-84)56.6 (37-76)0.7502
Male, n (%)5 (29.4)24 (42.9)15 (25.5)0.5533
Needle type
FNA needle (Expect)16031-
Franseen needle (Acquire)05032-
Fork-tip needle (SharkCore)030-
Reverse bevel needle (ProCore)011-
Other1013-
Not documented132-
Needle passes, mean (range)3.4 (1-5)3.4 (1-8)2.8 (1-8)-

The most common indications were pancreatic mass 31.1% (33/106), gastric mass 17.9% (19/106), liver biopsy 14.2% (15/106), and pancreatic cyst 13.2% (14/106). Other conditions evaluated included lymph nodes, biliary abnormalities, extraluminal lesions, pancreatitis, rectal masses, small bowel lesions, mediastinal lesions, and esophageal lesions (Table 2).

Table 2 Indications for endoscopic ultrasound-guided fine needle aspiration and endoscopic ultrasound-guided fine needle biopsy.
Indication (n = 106)n (%)
Pancreatic mass33 (31.1)
Gastric mass19 (17.9)
Liver biopsy15 (14.2)
Pancreatic cyst14 (13.2)
Lymph node6 (5.7)
Biliary5 (4.7)
Extraluminal4 (3.8)
Pancreatitis4 (3.8)
Rectal2 (1.9)
Small bowel2 (1.9)
Mediastinal1 (0.9)
Esophageal1 (0.9)

FNA alone was performed in 17 cases (16.0%); in 56 cases (52.8%), FNB alone was done; and in 33 cases (31.2%), FNA with FNB was performed. There was an overall mean of 3.3 (1-8) passes per needle; the mean was 3.4 (1-5) passes per needle in FNA alone cases, 3.4 (1-8) in FNB alone cases, and 2.8 (1-8) in cases where both FNA and FNB were performed (Table 1). The most commonly used needle size was 22-Gauge; a 22-Gauge FNA needle was used in 60.0% (30/50) of FNA needle cases, and a 22-Gauge FNB needle was used in 82.0% (73/89) of FNB cases.

Diagnostic yield

For all indications, diagnostic yield was 47.1% (8/17) in FNA alone cases, and 85.7% (48/56) in FNB alone cases (Table 3).

Table 3 Diagnostic yield and specimen adequacy, n (%).
Diagnostic yieldSpecimen adequacy
FNA alone (n = 17)8 (47.1)9 (52.9)
FNB alone (n = 56)48 (85.7)50 (89.3)
FNA with FNB (n = 33)28 (84.8)28 (84.8)

There was a significant difference in diagnostic yield between the three groups (Fisher’s exact test, P = 0.0039). In sub-group analysis, there was a significant difference between the FNA alone and FNB alone groups, (Fisher’s exact test, Bonferroni adjusted, P = 0.0067) and between the FNA alone and FNA with FNB groups (Fisher’s exact test, Bonferroni adjusted, P = 0.0238), but not between the FNB alone and FNA with FNB groups (Fisher’s exact test, Bonferroni adjusted, P = 1).

In cases where both FNA and FNB were performed in the same procedure, the overall diagnostic yield was 84.8% (28/33); 60.6% (20/33) in samples from FNA needles and 81.8% (27/33) in samples from FNB needles. There was a statistically significant difference in diagnostic yield (McNemar’s test, P = 0.0455) between the FNA and FNB specimen subgroups.

Specimen adequacy

Specimens were adequate in 52.9% (9/17) of FNA alone cases and adequate in 89.3% (50/56) of FNB alone cases (Table 3).

There was a significant difference in sample adequacy between the three groups (Fisher’s exact test, P = 0.0049). In sub-group analysis, there was a significant difference between the FNA alone and FNB alone groups, (Fisher’s exact test, Bonferroni adjusted, P = 0.0072), but not between the FNA alone and FNA with FNB groups (Fisher’s exact test, Bonferroni adjusted, P = 0.063), or between the FNB alone and FNA with FNB groups (Fisher’s exact test, Bonferroni adjusted, P = 1).

In cases where both FNA and FNB were performed, overall specimen adequacy was 84.8% (28/33); samples from FNA needles were adequate in 60.6% (20/33), while samples from FNB needles were adequate in 81.8% (27/33).There was a statistically significant difference in specimen adequacy (McNemar’s test, P = 0.0455) between the FNA and FNB specimen subgroups.

In 10.0% (5/50) of FNA cases, tissue could not be aspirated for cytology, while in 3.4% (3/89) of FNB cases, core tissue could not be obtained for histology. In 2 cases of pancreatic cystic lesions, when samples from FNB needles were grossly inadequate for histology, material was sent for cytology instead.

EUS-FNA/FNB of pancreatic masses

EUS-FNA or EUS-FNB performed for pancreatic masses produced adequate samples for pathologic analysis in 30/33 (90.9%). There was a trend towards improved sample adequacy from the first to second data collection time period with the predominant use of FNB (Fisher’s exact test, P = 0.0524). 26 patients had pancreatic malignancy on final diagnosis. Sensitivity for pancreatic malignancy was 96.2% (25/26); one case of benign EUS-FNB was confirmed malignant operatively. There were no cases of false positive EUS-FNA or EUS-FNB. Yield for malignancy for all pancreatic masses sampled via FNA or FNB was 75.8% (25/33). Importantly, there was a significant increase in the diagnostic yield from 46.2% (6/13) in the first collection period to 95.0% (19/20) in the second data collection time period (Fisher’s exact test, P = 0.0026). Mean follow up was 29.1 months (21.7-32.4).

Complications

Two patients (1.9%) had minor post-procedural bleeding after EUS-FNB; one was self-limiting, and one required the use of a hemoclip. There were no infectious complications due to FNA or FNB in our cohort; all patients (14/14) undergoing EUS-FNA of cystic lesions received prophylactic antibiotics.

DISCUSSION

The preferred approach to specimen preparation and processing of EUS-FNB samples is not well defined. In a recent trial evaluating the clinical performance of a fork-tip FNB needle (SharkCore, Medtronic, Sunnyvale, CA, United States), all FNB specimens were processed for histology[12]. However, in a trial examining the clinical performance of a Franseen needle (Acquire, Boston Scientific, Marlborough, MA, United States), 42.5% of FNB specimens were only sent for cytology, even though 90.3% of specimens had an adequate tissue core[13]. Diagnostic concordance in cytology specimen analysis varies significantly[24,25]. Inter-study heterogeneity has prevented identification of independent factors that contribute to the higher diagnostic yield of EUS-FNB noted in many studies. Recent studies have suggested alternate methods to increase diagnostic yield. In particular, contrast-enhanced EUS could be of significant benefit in characterizing pancreatic lesions[6], and touch-imprint cytology allows for processing of a single specimen for both cytology and histology[26].

Our institution has transitioned from the predominate use of EUS-FNA to EUS-FNB, and thus, to processing tissue core for histology rather than cytology. In our experience, utilization of EUS-FNB led to significant improvements in both diagnostic yield and specimen adequacy, as suggested by statistically significant differences in both parameters between FNA and FNB subgroups in patients who underwent FNA and FNB for the same lesion. Our results are comparable to recently published studies demonstrating specimen adequacies of 90.3% for a Franseen needle, 67% to 84.6% for a fork-tip needle, and 92.6% for a reverse bevel FNB needle (ProCore, Cook Medical, Bloomington, IN, United States)[12,13,27,28].

Limitations of our study include its retrospective nature, being a single-center experience, the use of multiple FNB needle types, and the heterogeneity of lesion types sampled.

In conclusion, EUS-FNB with subsequent processing of tissue core for histology improves diagnostic yield and specimen adequacy compared to EUS-FNA cytology. Specimen processing as histology may have contributed to the overall increased diagnostic yield of EUS-FNB.

ARTICLE HIGHLIGHTS
Research background

Endoscopic ultrasound-guided fine needle biopsy (EUS-FNB) has emerged as a safe, efficacious alternative to EUS-guided fine needle aspiration (EUS-FNA) for tissue acquisition. EUS-FNB is reported to have higher diagnostic yield while preserving specimen tissue architecture.

Research motivation

Data on the optimal method of EUS-FNB specimen processing is limited.

Research objectives

We evaluate EUS-FNB with specimen processing as histology vs EUS-FNA cytology with regards to diagnostic yield and specimen adequacy.

Research methods

A retrospective observational study of all EUS-FNA and EUS-FNB procedures performed at our institution from July 1, 2016, to January 31, 2018, was performed. The primary outcomes were diagnostic yield and specimen adequacy.

Research results

106 EUS-FNA and EUS-FNB procedures were analyzed. For all indications, diagnostic yield was 47.1% (8/17) in FNA alone cases, 85.7% (48/56) in FNB alone cases, and 84.8% (28/33) in cases where both FNA and FNB were performed (P = 0.0039). Specimens were adequate for pathologic evaluation in 52.9% (9/17) of FNA alone cases, in 89.3% (50/56) of FNB alone cases, and 84.8% (28/33) in cases where FNA with FNB were performed (P = 0.0049). In patients who underwent FNA with FNB, there was a statistically significant difference in both specimen adequacy (P = 0.0455) and diagnostic yield (P = 0.0455) between the FNA and FNB specimens.

Research conclusions

The study suggests that EUS-FNB with processing of tissue core for histology improves diagnostic yield and specimen adequacy compared to EUS-FNA cytology. Specimen processing as histology may have contributed to the overall increased diagnostic yield of EUS-FNB.

Research perspectives

Prospective research is needed to clarify optimal specimen processing of EUS-FNB in clinical settings with varied resources.

Footnotes

Manuscript source: Invited manuscript

Corresponding Author's Membership(s) in Professional Societies: American College of Gastroenterology; and American Society for Gastrointestinal Endoscopy.

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: United States

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Crinò SF S-Editor: Gong ZM L-Editor: A P-Editor: Wang LL

References
1.  Hewitt MJ, McPhail MJ, Possamai L, Dhar A, Vlavianos P, Monahan KJ. EUS-guided FNA for diagnosis of solid pancreatic neoplasms: a meta-analysis. Gastrointest Endosc. 2012;75:319-331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 477]  [Cited by in F6Publishing: 487]  [Article Influence: 40.6]  [Reference Citation Analysis (0)]
2.  Turner BG, Cizginer S, Agarwal D, Yang J, Pitman MB, Brugge WR. Diagnosis of pancreatic neoplasia with EUS and FNA: a report of accuracy. Gastrointest Endosc. 2010;71:91-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 150]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
3.  Ribeiro A, Vazquez-Sequeiros E, Wiersema LM, Wang KK, Clain JE, Wiersema MJ. EUS-guided fine-needle aspiration combined with flow cytometry and immunocytochemistry in the diagnosis of lymphoma. Gastrointest Endosc. 2001;53:485-491.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 155]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
4.  Mizuno N, Bhatia V, Hosoda W, Sawaki A, Hoki N, Hara K, Takagi T, Ko SB, Yatabe Y, Goto H, Yamao K. Histological diagnosis of autoimmune pancreatitis using EUS-guided trucut biopsy: a comparison study with EUS-FNA. J Gastroenterol. 2009;44:742-750.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in F6Publishing: 130]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
5.  Obuch J, Wani S. EUS-guided tissue acquisition in GI stromal tumors. Gastrointest Endosc. 2017;86:516-518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
6.  Crinó SF, Brandolese A, Vieceli F, Paiella S, Conti Bellocchi MC, Manfrin E, Bernardoni L, Sina S, D'Onofrio M, Marchegiani G, Larghi A, Frulloni L, Landoni L, Gabbrielli A. Endoscopic Ultrasound Features Associated with Malignancy and Aggressiveness of Nonhypovascular Solid Pancreatic Lesions: Results from a Prospective Observational Study. Ultraschall Med. 2019;Online ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
7.  Iglesias-Garcia J, Dominguez-Munoz JE, Abdulkader I, Larino-Noia J, Eugenyeva E, Lozano-Leon A, Forteza-Vila J. 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;106:1705-1710.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 235]  [Cited by in F6Publishing: 256]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
8.  Hébert-Magee S, Bae S, Varadarajulu S, Ramesh J, Frost AR, Eloubeidi MA, Eltoum IA. The presence of a cytopathologist increases the diagnostic accuracy of endoscopic ultrasound-guided fine needle aspiration cytology for pancreatic adenocarcinoma: a meta-analysis. Cytopathology. 2013;24:159-171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 236]  [Cited by in F6Publishing: 232]  [Article Influence: 21.1]  [Reference Citation Analysis (0)]
9.  Matynia AP, Schmidt RL, Barraza G, Layfield LJ, Siddiqui AA, Adler DG. Impact of rapid on-site evaluation on the adequacy of endoscopic-ultrasound guided fine-needle aspiration of solid pancreatic lesions: a systematic review and meta-analysis. J Gastroenterol Hepatol. 2014;29:697-705.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 86]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
10.  Kong F, Zhu J, Kong X, Sun T, Deng X, Du Y, Li Z. Rapid On-Site Evaluation Does Not Improve Endoscopic Ultrasound-Guided Fine Needle Aspiration Adequacy in Pancreatic Masses: A Meta-Analysis and Systematic Review. PLoS One. 2016;11:e0163056.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 56]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
11.  Mohamadnejad M, Mullady D, Early DS, Collins B, Marshall C, Sams S, Yen R, Rizeq M, Romanas M, Nawaz S, Ulusarac O, Hollander T, Wilson RH, Simon VC, Kushnir V, Amateau SK, Brauer BC, Gaddam S, Azar RR, Komanduri S, Shah R, Das A, Edmundowicz S, Muthusamy VR, Rastogi A, Wani S. Increasing Number of Passes Beyond 4 Does Not Increase Sensitivity of Detection of Pancreatic Malignancy by Endoscopic Ultrasound-Guided Fine-Needle Aspiration. Clin Gastroenterol Hepatol. 2017;15:1071-1078.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
12.  DiMaio CJ, Kolb JM, Benias PC, Shah H, Shah S, Haluszka O, Maranki J, Sharzehi K, Lam E, Gordon SR, Hyder SM, Kaimakliotis PZ, Allaparthi SB, Gress FG, Sethi A, Shah AR, Nieto J, Kaul V, Kothari S, Kothari TH, Ho S, Izzy MJ, Sharma NR, Watson RR, Muthusamy VR, Pleskow DK, Berzin TM, Sawhney M, Aljahdi E, Ryou M, Wong CK, Gupta P, Yang D, Gonzalez S, Adler DG. Initial experience with a novel EUS-guided core biopsy needle (SharkCore): results of a large North American multicenter study. Endosc Int Open. 2016;4:E974-E979.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 67]  [Article Influence: 8.4]  [Reference Citation Analysis (1)]
13.  Adler DG, Muthusamy VR, Ehrlich DS, Parasher G, Thosani NC, Chen A, Buscaglia JM, Appannagari A, Quintero E, Aslanian H, Taylor LJ, Siddiqui A. A multicenter evaluation of a new EUS core biopsy needle: Experience in 200 patients. Endosc Ultrasound. 2019;8:99-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 47]  [Article Influence: 9.4]  [Reference Citation Analysis (1)]
14.  Bang JY, Hebert-Magee S, Trevino J, Ramesh J, Varadarajulu S. Randomized trial comparing the 22-gauge aspiration and 22-gauge biopsy needles for EUS-guided sampling of solid pancreatic mass lesions. Gastrointest Endosc. 2012;76:321-327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 185]  [Cited by in F6Publishing: 202]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
15.  Kim GH, Cho YK, Kim EY, Kim HK, Cho JW, Lee TH, Moon JS; Korean EUS Study Group. Comparison of 22-gauge aspiration needle with 22-gauge biopsy needle in endoscopic ultrasonography-guided subepithelial tumor sampling. Scand J Gastroenterol. 2014;49:347-354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 102]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
16.  Cheng B, Zhang Y, Chen Q, Sun B, Deng Z, Shan H, Dou L, Wang J, Li Y, Yang X, Jiang T, Xu G, Wang G. Analysis of Fine-Needle Biopsy vs Fine-Needle Aspiration in Diagnosis of Pancreatic and Abdominal Masses: A Prospective, Multicenter, Randomized Controlled Trial. Clin Gastroenterol Hepatol. 2018;16:1314-1321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 97]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
17.  de Moura DTH, McCarty TR, Jirapinyo P, Ribeiro IB, Farias GFA, Ryou M, Lee LS, Thompson CC. Endoscopic Ultrasound Fine-Needle Aspiration versus Fine-Needle Biopsy for Lymph Node Diagnosis: A Large Multicenter Comparative Analysis. Clin Endosc. 2019;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 22]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
18.  Lee BS, Cho CM, Jung MK, Jang JS, Bae HI. Comparison of Histologic Core Portions Acquired from a Core Biopsy Needle and a Conventional Needle in Solid Mass Lesions: A Prospective Randomized Trial. Gut Liver. 2017;11:559-566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
19.  Ang TL, Li JW, Kwek ABE, Thurairajah PH, Wang LM. The difference in histological yield between 19G EUS-FNA and EUS-fine-needle biopsy needles. Endosc Ultrasound. 2019;8:255-260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 19]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
20.  Ieni A, Barresi V, Todaro P, Caruso RA, Tuccari G. Cell-block procedure in endoscopic ultrasound-guided-fine-needle-aspiration of gastrointestinal solid neoplastic lesions. World J Gastrointest Endosc. 2015;7:1014-1022.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 23]  [Cited by in F6Publishing: 21]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
21.  Jain D, Mathur SR, Iyer VK. Cell blocks in cytopathology: a review of preparative methods, utility in diagnosis and role in ancillary studies. Cytopathology. 2014;25:356-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 61]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
22.  Hébert-Magee S. Basic technique for solid lesions: Cytology, core, or both? Endosc Ultrasound. 2014;3:28-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 14]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
23.  Wani S, Muthusamy VR, McGrath CM, Sepulveda AR, Das A, Messersmith W, Kochman ML, Shah J. AGA White Paper: Optimizing Endoscopic Ultrasound-Guided Tissue Acquisition and Future Directions. Clin Gastroenterol Hepatol. 2018;16:318-327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 58]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
24.  Mounzer R, Yen R, Marshall C, Sams S, Mehrotra S, Said MS, Obuch JC, Brauer B, Attwell A, Fukami N, Shah R, Amateau S, Hall M, Hosford L, Wilson R, Rastogi A, Wani S. Interobserver agreement among cytopathologists in the evaluation of pancreatic endoscopic ultrasound-guided fine needle aspiration cytology specimens. Endosc Int Open. 2016;4:E812-E819.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
25.  Marshall C, Mounzer R, Hall M, Simon V, Centeno B, Dennis K, Dhillon J, Fan F, Khazai L, Klapman J, Komanduri S, Lin X, Lu D, Mehrotra S, Muthusamy VR, Nayar R, Paintal A, Rao J, Sams S, Shah J, Watson R, Rastogi A, Wani S. Suboptimal Agreement Among Cytopathologists in Diagnosis of Malignancy Based on Endoscopic Ultrasound Needle Aspirates of Solid Pancreatic Lesions: A Validation Study. Clin Gastroenterol Hepatol. 2018;16:1114-1122.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 8]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
26.  Crinò SF, Larghi A, Bernardoni L, Parisi A, Frulloni L, Gabbrielli A, Parcesepe P, Scarpa A, Manfrin E. Touch imprint cytology on endoscopic ultrasound fine-needle biopsy provides comparable sample quality and diagnostic yield to standard endoscopic ultrasound fine-needle aspiration specimens in the evaluation of solid pancreatic lesions. Cytopathology. 2019;30:179-186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
27.  Armellini E, Manfrin E, Trisolini E, Andorno S, Ballarè M, Bernardoni L, Boldorini RL, Gabbrielli A, Frulloni L, Larghi A, Occhipinti P, Scarpa A, Crinò SF. Histologic retrieval rate of a newly designed side-bevelled 20G needle for EUS-guided tissue acquisition of solid pancreatic lesions. United European Gastroenterol J. 2019;7:96-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 35]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
28.  Di Leo M, Crinò SF, Bernardoni L, Rahal D, Auriemma F, Correale L, Donato G, Massidda M, Anderloni A, Manfrin E, Armellini E, Poliani L, Fugazza A, Semeraro R, Occhipinti P, Repici A, Carrara S. EUS-guided core biopsies of pancreatic solid masses using a new fork-tip needle: A multicenter prospective study. Dig Liver Dis. 2019;51:1275-1280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]