v
Search
Advanced Search

Publications > Journals > Journal of Clinical and Translational Pathology > Article Full Text

  • OPEN ACCESS

Molecular Profiling Using Next-generation Sequencing of Sufficient Endobronchial Ultrasound-guided Transbronchial Needle Aspiration and Liquid Biopsy Samples in Patients with Advanced Lung Cancer

  • Kayla Brice1,
  • Candice Arline1,
  • Luis E. Raez2,
  • Katerine Dumais1 and
  • Mark Block2,* 
 Author information
Journal of Clinical and Translational Pathology   2024;4(3):110-114

doi: 10.14218/JCTP.2024.00021

Abstract

Background and objectives

Tumor molecular analysis using next-generation sequencing (NGS) is the standard of care for guiding lung cancer treatment. Endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) is a minimally invasive technique used to sample mediastinal lymph nodes for diagnosing and staging lung cancer. This study aimed to determine if EBUS-TBNA provided adequate tissue samples for NGS.

Methods

We evaluated EBUS-TBNA samples from adult advanced non-small cell lung cancer patients who had both EBUS-TBNA and liquid biopsy samples analyzed by NGS between July 1, 2015 and June 30, 2021. Additionally, we compared the results with those from liquid biopsies performed on these patients.

Results

Among the 44 evaluated patients, 43% were male, with a median age of 66 years at diagnosis. Seventy-five percent were smokers, 79.5% were White, 6.8% were Black, and 9.1% were Asian. EBUS-TBNA samples were sufficient for NGS in 95.5% of cases. The median turnaround time for EBUS-TBNA NGS was 38.5 days compared with eight days for NGS in liquid biopsies. Actionable genetic aberrations were detected in 71% of patients.

Conclusions

Our findings demonstrated that EBUS-TBNA provided sufficient tissue for identifying actionable genetic aberrations in patients with advanced non-small cell lung cancer.

Keywords

Lung neoplasms, Non-small cell lung cancer, Endobronchial ultrasound-guided transbronchial needle aspiration, Next-generation sequencing, Molecular profiling, Biopsy

Introduction

Lung cancer is the third most common cancer and the leading cause of cancer death in both men and women worldwide, with non-small cell lung cancer (NSCLC) accounting for 85% of all lung cancer diagnoses. Survival rates for patients with advanced NSCLC are low because most patients present with advanced diseases that are minimally responsive to standard therapy. Novel treatments, such as targeted agents and immunotherapy, improve survival rates when initiated early.1–3 Epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), C-ros oncogene 1 (ROS1), and v-raf murine sarcoma viral oncogene homolog B (BRAF) mutations account for approximately 30% to 60% of adenocarcinomas, with overall response rates ranging from 50% to 80% for patients who receive targeted therapy. Additionally, targeted therapy is associated with an increase in overall survival from 18 to 38.6 months. Therefore, evidence-based practice guidelines recommend adequate tissue acquisition for molecular studies.4–6

For patients with advanced-stage NSCLC, less invasive techniques such as needle biopsy are preferred to minimize the morbidity of the diagnostic procedure. However, needle aspiration often obtains a limited amount of tissue, which may be insufficient for molecular testing.7,8 Studies in the United States have shown that approximately one in three lung cancer patients undergo successful next-generation sequencing (NGS) testing before initial therapy.9,10 Unfortunately, nearly half of the patients still enter treatment with inadequate biomarker testing. NGS allows parallel testing of hundreds of genetic mutations, requires smaller samples with lower concentrations of malignant cells, and can be completed more rapidly compared to multiple sequential single-gene assays.11–13 Endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) is a minimally invasive technique for sampling mediastinal and hilar lesions and has proven to be a safe and effective method for the diagnosis and staging of lung cancer.14–16 Core needle biopsy is preferred for obtaining larger and more adequate samples for NGS; however, EBUS-TBNA samples can be processed to produce cell blocks that replicate high-quality histologic cores. Studies have estimated that as few as two passes are sufficient to obtain adequate tissue for molecular profiling.17–19 However, studies evaluating the adequacy of EBUS-TBNA tissue sampling for NGS to identify targetable mutations and initiate appropriate therapy are limited. In one study, adequate tissue for cytological analysis was obtained via EBUS-TBNA in 380 out of 391 patients (97.2%), and EGFR mutation testing on EBUS-TBNA samples had a success rate of 88.8% (95% confidence interval 74.7–95.6%).20–22

The purpose of this study was to review our experience with EBUS-TBNA in patients with advanced lung cancer to determine if this technique provided sufficient tissue for molecular profiling with outcomes similar to core biopsies. A secondary objective was to assess whether EBUS-TBNA samples were sufficient to initiate appropriate treatment and to evaluate the turnaround time (TAT) for NGS assay via tissue and liquid biopsy (LB).

Materials and methods

Record review

Records were reviewed for adult patients (≥18 years) diagnosed with stage III or IV NSCLC who had NGS performed on EBUS-TBNA samples between July 1, 2015, and June 30, 2021. The study included patients who underwent EBUS-TBNA at Memorial Cancer Institute and who began NSCLC therapy at Memorial Cancer Institute. The Epic Beacon Processing System was used to review the electronic medical records, and data were collected into a REDCap database.23,24 Data collected included patient demographics (gender, age at diagnosis, race, ethnicity), smoking history, clinical stage, histology, detection of genomic biomarkers (e.g., ALK, BRAF, MET, EGFR mutation, EGFR type, KRAS mutation, KRAS type, PDL1, PIK3CA, RET, ROS1, NTRK, STK11 type, TP53 mutation, TP53 type, TMB), occurrence of insufficient tissue samples, biopsy TAT, biopsy type used for final treatment decision (tissue vs. liquid), treatment regimen initiated, number of stations sampled, NGS success using EBUS-TBNA tissue samples, and date of disease progression or death. The TAT was defined as the time between the date the NGS was ordered in the electronic medical record and the date the results were reported by the laboratory. EBUS-TBNA tissue samples were formed from multiple aspirates pooled into Roswell Park Memorial Institute cell culture solution to create a cell block, which was then sent for NGS. All collected data were stored on a secure shared drive accessible only to the research team. Records were de-identified and coded to ensure patient confidentiality. Descriptive statistics were performed for all demographic and baseline levels of clinical outcome endpoints.

EBUS-TBNA procedure

EBUS-TBNA procedures were performed using a standardized technique with moderate sedation. After administering topical lidocaine anesthesia throughout the airway and conducting an inspection bronchoscopy, the Olympus EBUS bronchoscope was advanced through the oropharynx and into the trachea. Ultrasound examination of all mediastinal stations was routinely performed. Needle aspiration was conducted with a 22 Ga needle, targeting first the mediastinal node station farthest from the lung cancer (i.e., contralateral stations first, then ipsilateral). Aspiration samples were prepared for cytologic examination by dispensing the aspirate onto a glass slide, which was immediately placed in a fixative. Depending on the clinical situation, these slides were either sent for immediate cytologic interpretation (equivalent to rapid on-site evaluation) or submitted for routine examination. At least three additional aspirates were collected and prepared for histologic and molecular analysis by dispensing them into a single vial of Roswell Park Memorial Institute. This pooled sample was then centrifuged to produce a cell pellet, which was used to create a cell block. EBUS-TBNA samples were sent to CARIS Life Sciences (https://www.carislifesciences.com/ ), headquartered in Irving, Texas, for NGS testing. CARIS Life Sciences is a well-known provider of DNA and RNA NGS solutions and is a national leader in molecular testing in the United States.

Liquid biopsy procedure

Liquid biopsies (defined here as blood collection of free circulating tumor DNA (ctDNA)) were collected from peripheral blood and sent to Guardant Health (https://guardanthealth.com/ ) for ctDNA isolation and NGS of 77 genes. The usual turnaround time for most samples was eight days, and the report was emailed to the team for inclusion in the patients medical records.

Results

Patient characteristics

Of the 44 patients reviewed, approximately half were female, and a large majority were smokers (Table 1). The median age at diagnosis was 66 years (range 4383 years), and most patients were White/Caucasian (35, 79.5%).

Table 1

Patient demographics

CharacteristicNumber (%)
Median age at diagnosis (range) – yr.66 (43–83)
Gender
  Male19 (43.2)
  Female25 (56.8)
Race
  White/Caucasian35 (79.5)
  Black/African American3 (6.8)
  Asian4 (9.1)
  Unknown2 (4.5)
Ethnicity
  Hispanic16 (36.4)
  Non-Hispanic26 (59.1)
Smoking history
  Current smoker1 (2.3)
  Former smoker33 (75.0)
  Never smoker10 (22.7)
Histology
  Squamous cell carcinoma11 (25.0)
  Adenocarcinoma30 (68.2)
  Carcinoid1 (2.3)
  Poorly differentiated carcinoma2 (4.5)

Occurrence of sufficient tissue samples

EBUS-TBNA samples were sufficient for NGS in 42 patients (95.5%). In the remaining two patients, tissue biopsies were inconclusive due to insufficient sampling, but an EGFR mutation was detected by LB. LB was completed in 35 patients (79.5%) at diagnosis.

TAT

The median TAT for EBUS-TBNA was 38.5 days, compared to eight days for LB NGS. Most delays in obtaining NGS results were due to insurance authorization issues.

Actionable genetic aberrations and treatment decision

Thirty patients (71%) had an actionable genetic aberration (AGA) detected by NGS from EBUS-TBNA, while twenty-two patients (63%) had an AGA detected by NGS from liquid biopsies. AGAs identified by NGS from EBUS-TBNA and liquid biopsy samples are shown in Table 2. Final treatment decisions were based on EBUS-TBNA results for 37 patients (84.1%) and on LB results for seven patients (15.9%). Of the five patients with both EBUS-TBNA and LB results, but whose treatment decision was based on LB, four had LB results available earlier, and one had an EGFR mutation detected only by LB. A list of the treatment regimens is provided in Supplementary Table 1.

Table 2

Actionable genetic aberrations (AGA) detected by next-generation sequencing (NGS)

NEGFR n (%)
KRAS n (%)MET n (%)BRAF n (%)ALK n (%)NTRK1 n (%)PDL1+ >1% n (%)
exon 19exon 21
EBUS-TBNA309 (30)9 (30)5 (16)1 (3)01 (3)24 (80)
6 (67)1 (11)
Liquid biopsy2211 (50)9 (41)5 (23)2 (9)1 (5)0-
5 (45)2 (18)

Among seven studies evaluating NGS with EBUS-TBNA, all studies analyzed the feasibility of detecting EGFR mutations in samples obtained by EBUS-TBNA, while three also detected KRAS mutations (Table 3).7,8,25–29 The baseline characteristics of our study, in terms of age, were similar to those in previous literature, with a median age ranging between 60 and 70 years. Our patient population also had a gender distribution similar to that in other studies, although some reported a higher percentage of men (greater than 70%). Detection rates for EGFR and KRAS mutations ranged from 526% and 537%, respectively, in previous studies. Our study detected a higher percentage of EGFR mutations (30%) and similar KRAS mutations (30%). Additionally, our study highlighted a significant proportion of populations with EGFR mutations, including Asians and Hispanics.

Table 3

Previous literature

StudyMethodsEligible for analysis (N)EBUS EGFR positive, Number (%)EBUS KRAS Positive, Number (%)EBUS ALK Positive, Number (%)
Nakajima et al, 20077Analyzed the feasibility of detecting EGFR mutations in samples obtained by EBUS-TBNA in NSCLC patients4311 (25.6)
Garcia-Olivé et al, 20108Assessed the usefulness of EBUS-TBNA for the detection of EGFR mutations in NSCLC patients402 (5)
Navani et al, 201225Determined whether cytology specimens obtained from EBUS-TBNA in routine practice are suitable for phenotyping and genotyping in NSCLC patients647 (10.9)
Billah et al, 201126Analyzed the clinical utilization of routinely prepared cytology specimens for molecular testing to detect EGFR or KRAS mutations in NSCLC patients175 (EGFR)/174 (KRAS)34 (19.4)41 (23.6)
Schuurbiers et al, 201027Investigated the yield and applicability of molecular testing for KRAS and EGFR mutations in cytologic specimens obtained by Transesophageal Ultrasound (EUS) or EBUS-FNA in NSCLC patients272 (7.4)10 (37)
Kang et al, 201228Evaluated the usefulness of bronchoscopic biopsy and EBUS-TBNA biopsy for detecting EGFR and KRAS mutations in routine practice201 (EGFR)/196 (KRAS)30 (14.9)11 (5.6)
Jeyabalan et al, 201629Evaluated the adequacy of EBUS-TBNA samples for EGFR and ALK genetic mutation analysis in confirmed primary lung adenocarcinomas80 (EGFR)/21 (ALK)5 (6.3)0

Discussion

The primary aim of this study was to determine if EBUS-TBNA lung cancer samples were adequate for molecular analysis by NGS. We found that EBUS-TBNA samples were sufficient in 95% of patients and that therapeutic decisions were based on AGA results from EBUS samples in 84% of patients. In the remaining 16%, therapeutic decisions were based on LB results. Furthermore, because our study cohort was more ethnically diverse than those in similar reports, we believe our findings provide important evidence supporting the use of EBUS specimens for NGS in a broader population. This diversity was a characteristic of our patient population rather than an intentional aspect of the study design. Although there was a small number of Black patients in our cohort, this study included a higher proportion of Black patients compared to previous studies. For example, Navani and colleagues reported on 774 patients with known or suspected lung cancer, of whom 88% were Caucasian and less than 1% were African, Caribbean, or other non-Asian.25 Folch and colleagues reported on 42 patients, 81% of whom were white, 4.8% were Asian, and only 4.8% were Black (9.5% were listed as other).6 While the proportion of White/Caucasian patients in our study was slightly lower (79.5%), a higher percentage were Asian (9.1%) or African American (6.8%). Additionally, 36% of our patients were Hispanic, highlighting an underrepresented population in previous trials. Our study is notable for including a broad range of genetic aberrations. Xie et al.5 reported 41 driver mutations in 77 patients, including 32 EGFR mutations, eight ALK rearrangements, and one ROS1 rearrangement. In contrast, our study identified mutations in five genes (BRAF, EGFR, KRAS, MET, and NTRK1) and expression of PDL1. Furthermore, our study is unique in that it also included LB, allowing us to compare the diagnostic value of EBUS-TBNA with LB.

The EGFR mutation rate in our study was higher than the US average, where EGFR positivity is approximately 10% in White patients with NSCLC and up to 50% in Asian patients.30 Of the four Asian patients in our study, one had an EGFR exon 19 deletion detected by both EBUS and LB, one had an EGFR exon 21 mutation detected by LB, and one had an EGFR exon mutation detected by both EBUS and LB. Among the three African American patients, one had an EGFR exon 19 deletion detected by both EBUS and LB, and one had an EGFR exon 19 deletion detected by EBUS only.

The main limitation of this study is its relatively small sample size. Contributing factors include some liquid biopsies being performed outside the specified timeframe and some patients being diagnosed with early-stage lung cancer.

Overall, the patient population, the variety of mutations included, and validation with LB make our results relevant to current practice.

Conclusions

EBUS-TBNA can obtain sufficient tissue samples for identifying AGA by NGS in patients with advanced NSCLC. Based on our study results, additional biopsies are necessary in only a small minority of patients when EBUS-TBNA is performed properly. This includes creating cell blocks and obtaining material sufficient for molecular diagnostics, not just cytologic diagnosis.

Supporting information

Supplementary material for this article is available at https://doi.org/10.14218/JCTP.2024.00021 .

Supplementary Table 1

Treatment regimens based on EBUS-TBNA and LB results.

(DOCX)

Declarations

Acknowledgement

None.

Ethical statement

Ethical review and approval were waived for this study as outlined in 45 CFR 46.101 or 21 CFR 56.104. The research involved only the collection and analysis of information using identifiable health data regulated under 45 CFR parts 160 and 164, subparts A and E [HIPAA], for “health care operations” or “research” as defined in 45 CFR 164.501, or for “public health activities and purposes” as described under 45 CFR 164.512(b). The protocols adhered to the ethical guidelines of the most recent version of the Declaration of Helsinki. Patient consent was waived under 45 CFR 164.512 (i) (2) (ii).

Data sharing statement

The dataset used to support the findings of this study have not been made available because of privacy and/or ethical restrictions.

Funding

This research received no external funding.

Conflict of interest

Luis Raez: Astrazeneca, Lilly Oncology, LOXO, Genentech, Novocure, Merck. Mark Block: Southern Thoracic Surgical Association. Additional authors declare no conflict of interest.

Authors’ contributions

Conceptualization (MB, LR), methodology (MB, LR, CA, KB, KD), software (CA), validation (MB, LR, CA, KB, KD), formal analysis (MB, LR, CA, KB, KD), investigation (MB, LR, CA, KB, KD), resources (MB, LR, CA, KB, KD), data curation (CA), original draft preparation (MB, LR, CA, KB, KD), review and editing (MB, LR, CA, KB, KD), visualization (MB, LR, CA, KB, KD), supervision (MB, LR, KB, KD), and project administration (CA). All authors have read and agreed to the published version of the manuscript.

References

  1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin 2022;72(1):7-33 View Article PubMed/NCBI
  2. Chen R, Manochakian R, James L, Azzouqa AG, Shi H, Zhang Y, et al. Emerging therapeutic agents for advanced non-small cell lung cancer. J Hematol Oncol 2020;13(1):58 View Article PubMed/NCBI
  3. Kohno T, Nakaoku T, Tsuta K, Tsuchihara K, Matsumoto S, Yoh K, et al. Beyond ALK-RET, ROS1 and other oncogene fusions in lung cancer. Transl Lung Cancer Res 2015;4(2):156-164 View Article PubMed/NCBI
  4. Lindeman NI, Cagle PT, Aisner DL, Arcila ME, Beasley MB, Bernicker EH, et al. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. J Thorac Oncol 2018;13(3):323-358 View Article PubMed/NCBI
  5. Xie F, Zheng X, Mao X, Zhao R, Ye J, Zhang Y, et al. Next-Generation Sequencing for Genotyping of Endobronchial Ultrasound-Guided Transbronchial Needle Aspiration Samples in Lung Cancer. Ann Thorac Surg 2019;108(1):219-226 View Article PubMed/NCBI
  6. Folch E, Yamaguchi N, VanderLaan PA, Kocher ON, Boucher DH, Goldstein MA, et al. Adequacy of lymph node transbronchial needle aspirates using convex probe endobronchial ultrasound for multiple tumor genotyping techniques in non-small-cell lung cancer. J Thorac Oncol 2013;8(11):1438-1444 View Article PubMed/NCBI
  7. Nakajima T, Yasufuku K, Suzuki M, Hiroshima K, Kubo R, Mohammed S, et al. Assessment of epidermal growth factor receptor mutation by endobronchial ultrasound-guided transbronchial needle aspiration. Chest 2007;132(2):597-602 View Article PubMed/NCBI
  8. Garcia-Olivé I, Monsó E, Andreo F, Sanz-Santos J, Taron M, Molina-Vila MA, et al. Endobronchial ultrasound-guided transbronchial needle aspiration for identifying EGFR mutations. Eur Respir J 2010;35(2):391-395 View Article PubMed/NCBI
  9. Yarmus L, Akulian J, Gilbert C, Feller-Kopman D, Lee HJ, Zarogoulidis P, et al. Optimizing endobronchial ultrasound for molecular analysis. How many passes are needed?. Ann Am Thorac Soc 2013;10(6):636-643 View Article PubMed/NCBI
  10. Lindeman NI, Cagle PT, Beasley MB, Chitale DA, Dacic S, Giaccone G, et al. Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. J Thorac Oncol 2013;8(7):823-859 View Article PubMed/NCBI
  11. Robert NJ, Espirito JL, Chen L, Nwokeji E, Karhade M, Evangelist M, et al. Biomarker testing and tissue journey among patients with metastatic non-small cell lung cancer receiving first-line therapy in The US Oncology Network. Lung Cancer 2022;166:197-204 View Article PubMed/NCBI
  12. Bruno D, Hess L, Li X, Su E, Zhu Y, Patel M. Racial disparities in biomarker testing and clinical trial enrollment in non-small cell lung cancer (NSCLC). J Clin Oncol 2021;39(Suppl 15):9005-9005 View Article
  13. Diep R, MacDonald M, Cooper R, Grzegorczyk A, Rakocevic R, Chang CF, et al. Biopsy Method and Needle Size on Success of Next-Generation Sequencing in NSCLC: A Brief Report. JTO Clin Res Rep 2023;4(4):100497 View Article PubMed/NCBI
  14. Kage H, Kohsaka S, Shinozaki-Ushiku A, Hiraishi Y, Sato J, Nagayama K, et al. Small lung tumor biopsy samples are feasible for high quality targeted next generation sequencing. Cancer Sci 2019;110(8):2652-2657 View Article PubMed/NCBI
  15. Endris V, Penzel R, Warth A, Muckenhuber A, Schirmacher P, Stenzinger A, et al. Molecular diagnostic profiling of lung cancer specimens with a semiconductor-based massive parallel sequencing approach: feasibility, costs, and performance compared with conventional sequencing. J Mol Diagn 2013;15(6):765-775 View Article PubMed/NCBI
  16. Head SR, Komori HK, LaMere SA, Whisenant T, Van Nieuwerburgh F, Salomon DR, et al. Library construction for next-generation sequencing: overviews and challenges. Biotechniques 2014;56(2):61-77 View Article PubMed/NCBI
  17. Zhang C, Kim RY, McGrath CM, Andronov M, Haas AR, Ma KC, et al. The Performance of an Extended Next Generation Sequencing Panel Using Endobronchial Ultrasound-Guided Fine Needle Aspiration Samples in Non-Squamous Non-Small Cell Lung Cancer: A Pragmatic Study. Clin Lung Cancer 2023;24(2):e105-e112 View Article PubMed/NCBI
  18. Kinsey CM, Arenberg DA. Endobronchial ultrasound-guided transbronchial needle aspiration for non-small cell lung cancer staging. Am J Respir Crit Care Med 2014;189(6):640-649 View Article PubMed/NCBI
  19. Kunimasa K, Matsumoto S, Nishino K, Honma K, Maeda N, Kuhara H, et al. Comparison of sampling methods for next generation sequencing for patients with lung cancer. Cancer Med 2022;11(14):2744-2754 View Article PubMed/NCBI
  20. Uchimura K, Yanase K, Imabayashi T, Takeyasu Y, Furuse H, Tanaka M, et al. The Impact of Core Tissues on Successful Next-Generation Sequencing Analysis of Specimens Obtained through Endobronchial Ultrasound-Guided Transbronchial Needle Aspiration. Cancers (Basel) 2021;13(23):5879 View Article PubMed/NCBI
  21. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: Non-Small Cell Lung Cancer. Version 1.2023. Plymouth Meeting (PA): National Comprehensive Cancer Network; 2022
  22. Esterbrook G, Anathhanam S, Plant PK. Adequacy of endobronchial ultrasound transbronchial needle aspiration samples in the subtyping of non-small cell lung cancer. Lung Cancer 2013;80(1):30-34 View Article PubMed/NCBI
  23. Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)—a metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inform 2009;42(2):377-381 View Article PubMed/NCBI
  24. Harris PA, Taylor R, Minor BL, Elliott V, Fernandez M, O’Neal L, et al. The REDCap consortium: Building an international community of software platform partners. J Biomed Inform 2019;95:103208 View Article PubMed/NCBI
  25. Navani N, Brown JM, Nankivell M, Woolhouse I, Harrison RN, Jeebun V, et al. Suitability of endobronchial ultrasound-guided transbronchial needle aspiration specimens for subtyping and genotyping of non-small cell lung cancer: a multicenter study of 774 patients. Am J Respir Crit Care Med 2012;185(12):1316-1322 View Article PubMed/NCBI
  26. Billah S, Stewart J, Staerkel G, Chen S, Gong Y, Guo M. EGFR and KRAS mutations in lung carcinoma: molecular testing by using cytology specimens. Cancer Cytopathol 2011;119(2):111-117 View Article PubMed/NCBI
  27. Schuurbiers OC, Looijen-Salamon MG, Ligtenberg MJ, van der Heijden HF. A brief retrospective report on the feasibility of epidermal growth factor receptor and KRAS mutation analysis in transesophageal ultrasound- and endobronchial ultrasound-guided fine needle cytological aspirates. J Thorac Oncol 2010;5(10):1664-1667 View Article PubMed/NCBI
  28. Kang YR, Park HY, Jeon K, Koh WJ, Suh GY, Chung MP, et al. EGFR and KRAS mutation analyses from specimens obtained by bronchoscopy and EBUS-TBNA. Thorac Cancer 2013;4(3):264-272 View Article PubMed/NCBI
  29. Jeyabalan A, Bhatt N, Plummeridge MJ, Medford AR. Adequacy of endobronchial ultrasound-guided transbronchial needle aspiration samples processed as histopathological samples for genetic mutation analysis in lung adenocarcinoma. Mol Clin Oncol 2016;4(1):119-125 View Article PubMed/NCBI
  30. Hirsch FR, Bunn PA. EGFR testing in lung cancer is ready for prime time. Lancet Oncol 2009;10(5):432-433 View Article PubMed/NCBI
  • Journal of Clinical and Translational Pathology
  • pISSN 2993-5202
  • eISSN 2771-165X
Back to Top

Molecular Profiling Using Next-generation Sequencing of Sufficient Endobronchial Ultrasound-guided Transbronchial Needle Aspiration and Liquid Biopsy Samples in Patients with Advanced Lung Cancer

Kayla Brice, Candice Arline, Luis E. Raez, Katerine Dumais, Mark Block
  • Reset Zoom
  • Download TIFF