Context.—

Endobronchial ultrasound–guided transbronchial needle aspiration (EBUS-TBNA) has emerged as a very useful tool in the field of diagnostic respiratory cytology. Rapid on-site evaluation (ROSE) of EBUS-TBNA not only has the potential to improve diagnostic yield of the procedure but also to triage samples for predictive molecular testing to guide personalized treatments for lung cancer.

Objective.—

To provide an overview of the current status of the literature regarding ROSE of EBUS-TBNA in the diagnosis of lung cancer.

Data Sources.—

An electronic literature search in PubMed and Google databases was performed using the following key words: cytology, lung cancer, on-site evaluation, rapid on-site evaluation, and ROSE EBUS-TBNA. Only articles published in English were included in this review.

Conclusions.—

Rapid on-site evaluation can ensure that the targeted lesion is being sampled and can enable appropriate specimen triage. If available, it should be used with EBUS-TBNA in the diagnosis of lung cancer because it can minimize repeat procedures for additional desired testing (ie, molecular studies). Some studies have shown that ROSE does not adversely affect the number of aspirations, total procedure time of EBUS-TBNA, or the rate of postprocedure complications; it is also helpful in providing a preliminary diagnosis that can reduce the number of additional invasive procedures, such as mediastinoscopy. As EBUS technology continues to evolve, our knowledge of the role of ROSE in EBUS-TBNA for the diagnosis of lung cancer will also continue to grow and evolve.

Endobronchial ultrasound (EBUS) has emerged as an alternative minimally invasive method for surgical mediastinal staging of lung cancer.1,2  It allows real-time EBUS-guided transbronchial needle aspiration (EBUS-TBNA) of mediastinal and hilar lymph nodes (LNs) by providing access to the paratracheal LN stations (levels 2R, 2L, 4R, and 4L), the subcarinal LN (level 7), and the hilar, interlobar, and lobar LNs (levels 10, 11, and 12; Figure 1).3  The main indications of EBUS-TBNA include primary staging and restaging of lung cancer, staging of extrathoracic malignancies, and the diagnosis of granulomatous disease (eg, sarcoidosis and mycobacterial infection), in addition to the obtaining of tissue for diagnosis and/or ancillary testing for almost any centrally located mediastinal mass of undetermined etiology.4,5 

Figure 1. 

Lymph node stations accessible by endobronchial ultrasound–guided transbronchial needle aspirate, which include paratracheal lymph node stations (levels 2R, 2L, 4R, and 4L), the subcarinal lymph node (level 7), and the hilar, interlobar, and lobar lymph nodes (levels 10, 11, and 12).

Figure 1. 

Lymph node stations accessible by endobronchial ultrasound–guided transbronchial needle aspirate, which include paratracheal lymph node stations (levels 2R, 2L, 4R, and 4L), the subcarinal lymph node (level 7), and the hilar, interlobar, and lobar lymph nodes (levels 10, 11, and 12).

Close modal

Rapid on-site evaluation (ROSE) of EBUS-TBNA specimens aims to check sample adequacy and establish a preliminary diagnosis by performing a rapid stain in the bronchoscopy suite or operating room, with evaluation by a cytopathologist or a trained cytotechnologist.6,7  The basic purpose of ROSE is to increase the adequacy rate, diagnostic yield, and accuracy of the procedure. In addition, when performed in the operating room, ROSE of EBUS-TBNA is akin to frozen section evaluation by providing a preliminary diagnosis that directly impacts the management of the patient in deciding whether to proceed to mediastinoscopy. More importantly, in an era of targeted therapy, ROSE is performed to ensure collection of adequate and sufficient material for ancillary studies, including immunohistochemistry (IHC) for subtyping of lung cancer and potential molecular testing in these patients.812 

  1. To evaluate sampling adequacy of mediastinal LNs as evidenced by the presence of representative normal tissue (lymphoid tissue or anthracotic pigment-laden macrophages) and/or other lesional material (eg, granulomatous inflammation, malignancy).

  2. To evaluate the diagnostic yield for neoplastic or nonneoplastic disease.

  3. To ensure sampling of adequate material for appropriate triage of the sample for ancillary studies, including immunohistochemistry, microbiology studies, flow cytometry analysis, and molecular assays.

  4. To provide a preliminary diagnosis to direct immediate patient care, akin to a frozen section evaluation.

Rapid on-site evaluation of EBUS-TBNA guides the interventional pulmonologist or thoracic surgeon in real time and allows termination of sampling after the appropriate acquisition of diagnostic material and sufficient material for ancillary testing. Multiple targeted sites are usually sampled, and the TBNAs from each pass are triaged appropriately by ROSE (Figure 2).

Figure 2. 

Schematic diagram to show purpose and algorithmic flow of rapid on-site evaluation (ROSE) for endobronchial ultrasound–guided transbronchial needle aspirate (EBUS-TBNA) specimens. Abbreviation: Pap, Papanicolaou.

Figure 2. 

Schematic diagram to show purpose and algorithmic flow of rapid on-site evaluation (ROSE) for endobronchial ultrasound–guided transbronchial needle aspirate (EBUS-TBNA) specimens. Abbreviation: Pap, Papanicolaou.

Close modal

Type of Needles

There are 3 types of EBUS needles available, which include different sizes (21, 22, and 25 gauge) and material (stainless steel and nitinol). The most commonly used sizes are 21- and 22-gauge needles, with limited data available for 25-gauge needles.13  Studies indicate that there is no difference in sample adequacy and diagnostic yield between 21- and 22-gauge needles, although 21-gauge needles in combination with ROSE have been reported to be associated with fewer number of passes per LN station, as well as better histology.14  Because of the larger bore size of the 21-gauge needle, on the other hand, the samples tend to be more hemorrhagic than the samples obtained using the 22-gauge needle, and a trend toward more inadequate samples have been noted in some studies.14,15  However, there is no evidence that indicates superiority of one needle size compared with the other.16,17 

A small pilot study using ProCore ultrasound biopsy needles (Cook Medical, Bloomington, Indiana) found that they did not provide additional value in comparison with conventional fine-needle aspiration (FNA) needles.18 

Current literature recommends the use of either a 21- or a 22-gauge needle, and the choice of needle size is usually determined by the operator based on the station of the LNs, vascularity of the node, and the type of specimen processing (cytology versus histology).13  In general, the larger needles have a tendency to be less flexible, and this may impact a proceduralist's choice when trying to access more difficult LN stations, such as the hilar nodes, that require more flexibility for sampling. On the other hand, larger needles might help in obtaining tissue fragments for histologic evaluation of lymphoproliferative disorders.

Number of Passes

A single needle pass encompasses a single insertion of the aspiration needle, from entry to exit, through the airway wall into the target site, and includes 5 to 15 excursions of the needle within the target lesion. Studies indicate that in lung cancer diagnosis and staging, optimal diagnostic yield can be obtained after a minimum of 3 passes with EBUS-TBNA.19  For lung cancer staging, it is recommended to have a minimum of 3 aspiration passes at each LN station.19  Only small improvements in diagnostic yield after 4 or more passes have been seen, because the diagnostic yield tends to plateau after 3 passes. Rapid on-site evaluation of lung cancer specimens may also be needed to determine adequacy for molecular analysis20,21 ; however, in this setting, a single study showed that a median of 4 passes along with ROSE is required to obtain adequate material.20Anesthesia time is usually sufficient for multiple passes to increase sampling, allowing triage and support for the technician for ROSE.

There are many variables that impact the adequacy of material for ancillary testing. An advantage of ROSE is that it allows gross inspection of the material collected in the needle rinse for producing cell blocks and preparation of smears. Those steps can be crucial in optimizing these small samples for molecular testing.

Specimen Type

The EBUS-TBNA is essentially FNA performed through the bronchial wall using a bronchoscope and real-time ultrasound guidance. Although there is some controversy in the literature regarding the use of core needle biopsy versus FNA in other organs, inferior results of FNA are frequently due to a lack of experience and expertise of the operator, and the absence of proper adequacy assessment.22,23A meta-analysis of 20 studies found no significant difference in sensitivity and specificity of lung cancer diagnosis between core needle biopsy and transthoracic FNA samples.24 

The FNA cell blocks and direct FNA smears show comparable results with core needle biopsy samples for molecular testing.25,26  Recently published commentary by van Zante and Ljung22  discuss the importance of correct technique and application of ROSE in FNAs for obtaining an adequate sample that is superior to that of a core needle biopsy sample.

Similarly, in mediastinal lesions, EBUS with ROSE is preferred to more invasive mediastinoscopy and video-assisted thoracic surgery–guided biopsies for the diagnosis and staging of lung cancer, because of the increased diagnostic yield of the technique.2729 

Studies have shown that EBUS with ROSE can diagnose metastatic lung cancers with a sensitivity and positive predictive value of greater than 90%. The negative predictive value, however, varies from 61% to 97%, which may be due to a combination of factors, such as limited time for ROSE, lack of diagnostic material in an otherwise adequate aspirate, type of mediastinal station, and suboptimal staining quality of the smear.3032 

Interestingly it has been seen that the vast majority of LNs sampled during EBUS-FNA with a diagnosis of negative and unsatisfactory for evaluation are likely to be truly negative on follow-up.33 

The EBUS-TBNA material is immediately expressed over labeled glass slides for direct smears. The smears are stained by rapid stains (Romanowsky stains on unfixed air-dried [eg, Diff-Quik or Giemsa stains] and rapid Papanicolaou or hematoxylin-eosin stains on wet alcohol-fixed smears) and evaluated under light microscopy by the cytopathologist/cytotechnologist for sample adequacy and a preliminary diagnosis. The needle rinses from the FNA passes are typically collected in saline, RPMI, Hanks solution, formalin, or a preservative solution, like Cytolyt, and processed as a cell block, liquid-based cytology, or cytospin preparation. If a diagnosis of lung cancer is established on-site, additional passes may be requested to yield additional sample material to assess predictive biomarkers.34  If a sample looks concerning for a lymphoproliferative process, material may also be collected and submitted for flow cytometry.

Staining Methods for ROSE

A wide variety of cytologic stains are employed across laboratories, and the choice depends on the infrastructure of the hospital for on-site services, budget allocation, and the availability of trained cytology staff. There are various commercial staining kits available (Table 1), among which Diff-Quik is the most commonly employed stain because of the rapidity of the procedure, the ability to perform the stain on air-dried slides, and its superiority over the Papanicolaou stain in evaluating lymphoid samples. The staining method employed during ROSE, however, does not influence the final diagnosis.35 

Table 1. 

Comparison of Cytologic Stains Frequently Used for Rapid On-Site Evaluation

Comparison of Cytologic Stains Frequently Used for Rapid On-Site Evaluation
Comparison of Cytologic Stains Frequently Used for Rapid On-Site Evaluation

Adequacy Assessment and Performing ROSE

The availability of an on-site experienced cytopathologist for ROSE is a considerable issue at most institutions and depends largely on the institutional infrastructure, the location of the EBUS-TBNA (eg, operating room with the ability to immediately convert to mediastinoscopy versus bronchoscopy without the ability to convert to mediastinoscopy), and the case volume. To alleviate the costs and time associated with ROSE and encourage its use in clinical practice, several institutions have implemented the use of ROSE by trained interventional pulmonologists and cytotechnologists.36,37  Although a single study has shown satisfactory interobserver agreement (greater than 90%) between cytopathologists and interventional pulmonologists or trained cytotechnologists,38  those professionals cannot render a preliminary diagnosis and can only assess adequacy. Another option is the use of telecytology to provide ROSE services to a remote location without physically having the cytopathologist on-site. The telecytology system can serve as a valid substitute for on-site assessment of EBUS-TBNA using a digital camera attached to the microscope to transmit stained slide images via a secure Ethernet connection to a cytopathologist who can perform ROSE remotely, with the results communicated by a voice communication system to the proceduralist.39,40 

However, resources are still needed for this approach, including personnel trained to generate the immediately stained slides, and information technology infrastructure to ensure a reliable and secure transmission of the slide imaging.

While evaluating EBUS-TBNA specimens, it is important to be aware of airway elements that may be present in the sample, because the EBUS-TBNA needle passes via the upper aerodigestive tract through the wall of the bronchus into the target lesion. These elements can include squamous cells with bacteria from the oral cavity, ciliated respiratory columnar epithelial cells, histiocytes with anthracotic pigment, chondromyxoid fragments of cartilage, mesothelial cells, mucus, and cuboidal glandular cells of submucosal glands.

Reactive bronchial epithelial cells can often be challenging to interpret in ROSE when lung cancer is suspected clinically, particularly in the setting of smoking exposure, prior radiation therapy, and especially on air-dried smears because of the enlarged size of cells and nuclei. Attention should be focused on the periphery of cell clusters, where bronchial epithelial cells are thinly spread, for the presence of cilia, terminal bars, and columnar morphology that indicates the benign nature of these reactive cells. Familiarity with the spectrum of reactive respiratory epithelial changes and identification of ciliated cells are crucial in the setting of florid goblet cell metaplasia, which may mimic low-grade invasive mucinous adenocarcinoma. In addition, attention should be given to cellular dyscohesion and background necrosis, which are usually seen in neoplastic lesions, especially if cells are pleomorphic and show anaplastic features. However, caution should be used because dyscohesive lymphocytes and epithelioid histiocytes should not be misdiagnosed as tumor cells. On the other hand, signet ring cells of adenocarcinoma can sometimes resemble macrophages. The key features for a correct diagnosis are the presence of anthracotic pigment within macrophages and nuclear atypia within tumor cells.41  A detailed clinical history and meticulous evaluation of the morphology can help at the time of ROSE to diagnose less common diagnostic possibilities,42  such as granulomatous disease and lymphoproliferative disorders.

Sometimes lymphohistiocytic aggregates or germinal center fragments can mimic nonnecrotizing granulomas during ROSE. The presence of epithelioid histiocytes in clusters can be indicative of granulomatous lymphadenitis, which can help in triaging the specimen for microbial cultures and special stains. Although the diagnostic yield of EBUS-TBNA in sarcoidosis is good, fibrotic LNs may pose a challenge for yielding adequate tissue for diagnosis.43  A careful inspection for malignant cells is always important at the time of ROSE, even in the presence of granulomas, because granulomas may coexist with malignancy (eg, germ cell tumors, squamous cell carcinoma, lymphoma, etc). Other diagnostic pitfalls include the cytomorphologic overlap between reserve cell hyperplasia and small cell carcinoma, in addition to goblet cell or squamous metaplasia and non–small cell lung carcinoma at the time of ROSE. Given that cell block and immunohistochemistry slides are not available at the time of ROSE, the experience and knowledge of the cytopathologist evaluating the cytomorphology in EBUS-TBNA specimens are crucial to maximize the concordance of preliminary and final diagnoses, minimize indeterminate diagnoses, and appropriately direct patient care.

The evaluation of EBUS-TBNA aspirates for lymphoproliferative disorders can be particularly problematic for small cell lymphomas, especially in the absence of appropriate clinical history or clinical suspicion. However, if a lymphoid sample is obtained, consideration of a possible lymphoproliferative process may prompt allocation of material for flow cytometry and/or clonality or gene rearrangement studies in order to increase the likelihood of a definitive and accurate diagnosis. The typical findings include a monomorphic population of lymphoid cells (eg, monotony in cell size and chromatin pattern) without germinal center fragments or tingible body macrophages.

Adequacy for Peribronchial/Peritracheal LNs

An adequate LN sample should possess lymphocytes and/or lymphohistiocytic aggregates or germinal center fragments. However, if the lymphoid sampling is limited, qualitative criteria must be applied when evaluating sampling adequacy. Although there are no universally accepted criteria for EBUS LN adequacy, structured semiquantitative scoring schemes for ROSE and diagnostic category assignments have been proposed in previous publications (Table 2).11,4446 

Table 2. 

Adequacy Criteria of Rapid On-Site Evaluation Specimens of Endobronchial Ultrasound (EBUS)–Guided Transbronchial Needle Aspirate Samples of Lymph Node

Adequacy Criteria of Rapid On-Site Evaluation Specimens of Endobronchial Ultrasound (EBUS)–Guided Transbronchial Needle Aspirate Samples of Lymph Node
Adequacy Criteria of Rapid On-Site Evaluation Specimens of Endobronchial Ultrasound (EBUS)–Guided Transbronchial Needle Aspirate Samples of Lymph Node

A sample is considered to be adequate if there is sufficient diagnostic lesional material (eg, tumor or granulomatous pathology) even in the absence of lymphoid tissue, or sufficient benign lymphoid tissue to suggest adequate sampling of a benign/reactive LN.

Adequacy for Parenchymal Lung Lesions

Adequacy assessment of parenchymal lesions is more challenging because of the distinction between nondiagnostic and negative categories. Caution should be exercised when assigning the negative category in the presence of a mass lesion by imaging studies, especially if smears are sparsely cellular (containing only benign elements, pneumocytes, alveolar macrophages, and respiratory columnar cells).

Sample Adequacy Assessment

This is likely the most important component of performing ROSE. It reduces sampling error and controls the diagnostic yield of the procedure by directing the interventional pulmonologist or thoracic surgeon with real-time ultrasound guidance regarding sampling adequacy and the exact timing of termination of the procedure.47 

There are few observational studies and comparative randomized trials that assess the performance of ROSE in EBUS-TBNA for lung cancer diagnosis, staging, and molecular profiling (Table 3).4855 

Table 3. 

Studies Analyzing the Role of Rapid On-Site Evaluation (ROSE) in Endobronchial Ultrasound–Guided Transbronchial Needle Aspirate (EBUS-TBNA) in the Diagnosis of Lung Cancer

Studies Analyzing the Role of Rapid On-Site Evaluation (ROSE) in Endobronchial Ultrasound–Guided Transbronchial Needle Aspirate (EBUS-TBNA) in the Diagnosis of Lung Cancer
Studies Analyzing the Role of Rapid On-Site Evaluation (ROSE) in Endobronchial Ultrasound–Guided Transbronchial Needle Aspirate (EBUS-TBNA) in the Diagnosis of Lung Cancer

Morphologic Diagnosis

Subtyping lung cancer by ROSE is feasible when performed by an experienced cytopathologist or cytotechnologist. The cytomorphologic features for subtyping non–small cell carcinoma have been classically described. Morphologic criteria used for a cytologic diagnosis of adenocarcinoma include small to medium-size cell clusters (often 3-dimensional) with enlarged irregular nuclei, nuclear crowding/overlapping, prominent nucleoli, and wispy vacuolated cytoplasm and/or intracytoplasmic mucin. Sheets of cells with dense cytoplasm, distinct cytoplasmic borders, and irregular nuclei with multiple small nucleoli are more characteristic of squamous cell carcinoma. Often, background necrosis and hyperchromatic nuclei are seen. Typical findings of small cell carcinoma include predominantly singly dispersed cells with scant cytoplasm, nuclear molding, readily identifiable mitotic figures, basophilic cytoplasm with paranuclear blue bodies, and moderate necrosis/apoptosis.

Acquisition of Material for Ancillary Studies

After acquisition of a diagnostic sample on ROSE, the EBUS-TBNA could be terminated to minimize complications, or, if safe, additional passes can be obtained for ancillary testing and/or additional sampling of other stations for further staging information.50,53 

For instance, if lymphoma is suspected on ROSE, additional passes can be obtained for immunophenotyping by flow cytometry. If granulomatous inflammation is seen on ROSE, additional passes can be sent for microbiologic culture and special stains. To increase the sensitivity of the diagnostic bronchoscopy, EBUS-TBNA can be combined with bronchoalveolar lavage and bronchial brushing for sending specimens for microbiologic culture studies.

Extra passes (in formalin, saline, RPMI, or Hanks solution) can be requested for ancillary studies after a ROSE diagnosis of carcinoma. Cytology smears provide excellent-quality DNA and consistent genotyping results; however, the current molecular testing guidelines for non–small cell lung carcinoma, which recommend the use of cell blocks instead of conventional cytologic preparations, may reflect the preference of most molecular pathologists used to working with preparations that are similar to histology samples.56  Given that a variety of different cytology preparations can be used for molecular testing, institutional protocols should be used to allocate material in the appropriate manner (eg, additional unstained smears or cell block), because the choice depends on individual laboratory practice and validation of ancillary tests.57 

Non–small cell lung carcinoma accounts for more than 80% of newly diagnosed lung cancer cases, and most patients receive a diagnosis of an advanced stage of the disease in which surgical resection is not possible.58,59A large fraction of the lung cancer patients receive a diagnosis by either cytology or small biopsy. Endobronchial ultrasound–TBNA has been incorporated as a first step in the diagnosis and staging of suspected lung cancer.2  Often, TBNA is the only material available for both the diagnosis and the molecular testing that are required for patient care. Use of ROSE in combination with EBUS ensures adequate material for the diagnosis, staging, and molecular testing of lung cancer.

Molecular testing accuracy depends on multiple factors that include overall cellularity, method of fixation, tumor fraction of the sample (the ratio of tumor cells compared with all nucleated cells), and the analytic sensitivity of the molecular testing platform used for the analysis. Rapid on-site evaluation helps in triaging material for molecular and cytogenetic studies, including EGFR and KRAS mutations and ALK and ROS1 rearrangement.20,52,6063  The material obtained by EBUS-TBNA is suitable for molecular analysis in more than 90% of the samples.64,65  Specimens were satisfactory and adequate for molecular analysis in 95% of all cases in the study by Yarmus et al,20  where they used material for EGFR and KRAS sequencing and ALK fluorescence in situ hybridization analysis.

It is noteworthy that ROSE is useful for the confirmation of the presence of tumor cells and judging tumor burden within the samples for molecular analysis.

A randomized controlled trial by Trisolini et al53  evaluated the role of ROSE in EBUS-TBNA samples for molecular testing. The results, although not statistically significant, were clinically relevant and showed a 10% increase in the success rate of EBUS-TBNA for optimal lung cancer genotyping.

Current data are insufficient to know exactly how many passes are needed to obtain adequate material for molecular analysis, but it is strongly recommended to obtain additional material for molecular analysis after acquiring a diagnostic sample obtained using ROSE.

Of note, cytopathologists must understand the overall cellularity and tumor fraction required by the molecular laboratory to confidently assess adequacy for downstream testing, and conversely, to know when to request additional material. Thus, close correspondence or workshops with the molecular laboratory can assist cytopathologists in recognizing the lower limits of adequacy for molecular testing. Additionally, in situations in which the molecular laboratory accepts smeared slides for testing, ROSE can assist in allocating materials for distribution to the molecular testing facility.

Two task force guidelines on the role of EBUS-TBNA in the diagnosis and molecular testing of lung cancer have critically examined the utility of ROSE based on the literature.13,66  The guidelines found that ROSE is effective in reducing additional procedures. Interestingly, no statistically significant differences were seen in terms of the number of aspirations, diagnostic yield, procedure time, and rate of complications in procedures with or without ROSE. The expert panel recommended tissue sampling with or without ROSE in patients undergoing EBUS-TBNA for diagnostic evaluation of lung cancer (grade 1C).13 

Rapid on-site evaluation was used initially with conventional TBNA to increase the diagnostic yield of the procedure. Some argue that because EBUS is a real-time technique where needle and target can be seen during the procedure to check for the procedure's accuracy, ROSE is theoretically not needed. Liquid-based cytology further militates against ROSE and in fact ultimately eliminates the need for ROSE entirely. In liquid-based cytology, the sample is directly transferred to the preservative fluid for processing in the laboratory without making any direct smear on-site. Other arguments indicate the need for an experienced dedicated cytopathologist as a limitation of ROSE, because it may not be possible to accommodate in all hospitals or institutes. However, the increasing use of telecytology for ROSE may allow both an adequacy assessment and an immediate diagnosis if performed by a cytopathologist from a remote location. Other criticisms of ROSE include the uncertainty of the diagnostic material in a cell block despite the use of ROSE services, the time spent in performing the evaluation, and the cost. Rapid on-site evaluation is a time-consuming process because of repeated staining and examination of the samples, and the cost of ROSE may not be fully recoverable.67 

Because procedures can take a long time, the on-site presence of a pathologist is considered to be prohibitive by some laboratories. However, overall cost analysis shows significant benefit in the global patient care.68  The cost of ROSE depends on local factors, such as the geographic location of a hospital with physical proximity to laboratories and bronchoscopy suites, staff availability, hospital infrastructure, and access to appropriate follow-up health care. ROSE is cost effective; it reduces the need for additional procedures, and it reduces the number of cytology slides examined. The ability to better triage the sample for ancillary studies can save both time and money, and reduce complications in terms of the lower number of repeat procedures for ancillary studies.

In addition, as stated earlier, ROSE quickly guides appropriate sampling, which includes sending a sample to microbiology in cases of granulomatous pathology, and triage for flow cytometry/cytogenetics/molecular biology if a lymphoproliferative lesion or lung cancer is suspected.

It is recommended that patients with a negative diagnosis on EBUS-TBNA should undergo a more invasive procedure, such as mediastinoscopy, for confirmation of diagnosis.69 

In these situations, ROSE has evolved as a very useful tool to reduce the percentage of inadequate specimens, and consequently contributes in a big way to patient care.51 

Table 4 briefly summarizes the advantages and limitations of ROSE.

Table 4. 

Advantages and Limitations of Rapid On-Site Evaluation (ROSE)

Advantages and Limitations of Rapid On-Site Evaluation (ROSE)
Advantages and Limitations of Rapid On-Site Evaluation (ROSE)

The ROSE of EBUS-TBNA is a well-established tool to improve yield of the procedure if performed by a cytopathologist or an experienced cytotechnologist. Rapid on-site evaluation used in combination with TBNA ultimately reduces the workload of the cytopathology laboratory through a substantial reduction in the total number of slides sent by pulmonologists compared with samples without ROSE. Most importantly, improved patient care is achieved with ROSE. Different clinical trials and large series have reported lower complications rates when ROSE is performed.13,51,70,71  The evidence supports inclusion of ROSE in every EBUS-TBNA procedure, whenever feasible, in patients with suspected lung cancer and enlarged mediastinal or hilar LN and/or centrally located tumors.48,50  The decision of whether or not to provide ROSE for EBUS-TBNA procedures is largely institution dependent, and it is impacted by where the procedure is occurring (eg, bronchoscopy versus operating room), if a preliminary diagnosis is needed for immediate patient care, and if special triage is desired. The ability to provide ROSE for EBUS-TBNA and prevent a more invasive procedure like mediastinoscopy in lung cancer patients with advanced disease is an important advantage. Additional studies are needed to compare efficacy of ROSE for optimal performance of EBUS-TBNA in the diagnosis and workup of lung cancer patients.

1
Rivera
MP.
Mehta
AC.
Wahidi
MM.
Establishing the diagnosis of lung cancer: diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines
.
Chest
.
2013
;
143
(
suppl 5
):
e142S
165S
.
2
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
.
3
Ernst
A.
Anantham
D.
Eberhardt
R.
Krasnik
M.
Herth
FJ.
Diagnosis of mediastinaladenopathy-real-time endobronchial ultrasound guided needle aspiration versus mediastinoscopy
.
J Thorac Oncol
.
2008
;
3
(
6
):
577
582
.
4
Groth
SS.
Andrade
RS.
Endobronchial ultrasound-guided transbronchial needle aspiration for mediastinal lymph node staging in non-small cell lung cancer
.
Semin Thorac Cardiovasc Surg
.
2008
;
20
(
4
):
274
278
.
5
Herth
FJ.
Annema
JT.
Eberhardt
R.
et al.
Endobronchial ultrasound with transbronchial needle aspiration for restaging the mediastinum in lung cancer
.
J Clin Oncol
.
2008
;
26
(
20
):
3346
3350
.
6
Nakajima
T.
Yasufuku
K.
Saegusa
F.
et al.
Rapid on-site cytologic evaluation during endobronchial ultrasound-guided transbronchial needle aspiration for nodal staging in patients with lung cancer
.
Ann Thorac Surg
.
2013
;
95
(
5
):
1695
1699
.
7
Guo
H.
Liu
S.
Guo
J.
et al.
Rapid on-site evaluation during endobronchial ultrasound-guided trans bronchial needle aspiration for the diagnosis of hilar and mediastinal lymphadenopathy in patients with lung cancer
.
Cancer Lett
.
2016
;
371
(
2
):
182
186
.
8
Madan
NK.
Madan
K.
Jain
D.
et al.
Utility of conventional transbronchial needle aspiration with rapid on-site evaluation (c-TBNA ROSE) at a tertiary care center with endobronchial ultrasound (EBUS) facility
.
J Cytol
.
2016
;
33
(
1
):
22
26
.
9
Mfokazi
A.
Wright
CA.
Louw
M.
et al.
Direct comparison of liquid-based and smear-based cytology with and without rapid on site evaluation for fine needle aspirates of thoracic tumors
.
Diagn Cytopathol
.
2016
;
44
(
5
):
363
368
.
10
Thiryayi
SA.
Rana
DN.
Narine
N.
Najib
M.
Bailey
S.
Establishment of an endobronchial ultrasound-guided transbronchial fine needle aspiration service with rapid on-site evaluation: 2 years' experience of a single UK centre
.
Cytopathology
.
2016
;
27
(
5
):
335
343
.
11
Jeffus
SK.
Joiner
AK.
Siegel
ER.
et al.
Rapid on-site evaluation of EBUS-TBNA specimens of lymph nodes: comparative analysis and recommendations for standardization
.
Cancer Cytopathol
.
2015
;
123
(
6
):
362
372
.
12
Walia
R.
Madan
K.
Mohan
A.
et al.
Diagnostic utility of conventional transbronchial needle aspiration without rapid on-site evaluation inpatients with lung cancer
.
Lung India
.
2015
;
32
(
2
):
198
199
.
13
Wahidi
MM.
Herth
F.
Yasufuku
K.
et al.
Technical aspects of endobronchial ultrasound-guided transbronchial needle aspiration: CHEST guideline and expert panel report
.
Chest
.
2016
;
149
(
3
):
816
835
.
14
Yarmus
LB.
Akulian
J.
Lechtzin
N.
et al
American College of Chest Physicians Quality Improvement Registry, Education, and Evaluation (AQuIRE) Participants. Comparison of 21-gauge and 22-gauge aspiration needle in endobronchial ultrasound-guided transbronchial needle aspiration: results of the American College of Chest Physicians Quality Improvement Registry, Education, and Evaluation Registry
.
Chest
.
2013
;
143
(
4
):
1036
1043
.
15
Nakajima
T.
Yasufuku
K.
Takahashi
R.
et al.
Comparison of 21-gauge and 22-gauge aspiration needle during endobronchial ultrasound-guided transbronchial needle aspiration
.
Respirology
.
2011
;
16
(
1
):
90
94
.
16
Jeyabalan
A.
Shelley-Fraser
G.
Medford
AR.
Impact of needle gauge on characterization of endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) histology samples
.
Respirology
.
2014
;
19
(
5
):
735
739
.
17
Oki
M.
Saka
H.
Kitagawa
C.
et al.
Randomized study of 21-gauge versus 22-gauge endobronchial ultrasound-guided transbronchial needle aspiration needles for sampling histology specimens
.
J Bronchology Interv Pulmonol
.
2011
;
18
(
4
):
306
310
.
18
Xing
J.
Manos
S.
Monaco
SE.
Wilson
DO.
Pantanowitz
L.
Endobronchial ultrasound-guided transbronchial needle aspiration: a pilot study to evaluate the utility of the procore biopsy needle for lymph node sampling
.
Acta Cytol
.
2016
;
60
(
3
):
254
259
19
Lee
HS.
Lee
GK.
Lee
HS.
et al.
Real-time endobronchial ultrasound-guided transbronchial needle aspiration in mediastinal staging of non-small cell lung cancer: how many aspirations per target lymph node station?
Chest
.
2008
;
134
(
2
):
368
374
.
20
Yarmus
L.
Akulian
J.
Gilbert
C.
et al.
Optimizing endobronchial ultrasound for molecular analysis: how many passes are needed?
Ann Am Thorac Soc
.
2013
;
10
(
6
):
636
643
.
21
Detterbeck
FC.
Jantz
MA.
Wallace
M.
Vansteenkiste
J.
Silvestri
GA.
American College of Chest Physicians. Invasive mediastinal staging of lung cancer: ACCP evidence-based clinical practice guidelines (2nd edition)
.
Chest
.
2007
;
132
(
suppl 3
):
202S
220S
.
22
van Zante
A.
Ljung
BM.
Fine-needle aspiration versus core needle biopsy: reconsidering the evidence of superiority
.
Cancer
.
2016
;
124
(
12
):
853
856
.
23
Van der Laan
PA.
Fine-needle aspiration and core needle biopsy: an update on 2 common minimally invasive tissue sampling modalities
.
Cancer
.
2016
;
124
(
12
):
862
870
.
24
Yao
X.
Gomes
MM.
Tsao
MS.
Allen
CJ.
Geddie
W.
Sekhon
H.
Fine-needle aspiration biopsy versus core-needle biopsy in diagnosing lung cancer: a systematic review
.
Curr Oncol
.
2012
;
19
(
1
):
e16
e27
.
25
Coley
SM.
Crapanzano
JP.
Saqi
A. FNA.
core biopsy, or both for the diagnosis of lung carcinoma: obtaining sufficient tissue for a specific diagnosis and molecular testing
.
Cancer Cytopathol
.
2015
;
123
(
5
):
318
326
.
26
Roy-Chowdhuri
S.
Aisner
DL.
Allen
TC.
et al.
Biomarker testing in lung carcinoma cytology specimens: a perspective from members of the Pulmonary Pathology Society
.
Arch Pathol Lab Med
.
2016
;
140
(
11
):
1267
1272
.
27
Karunamurthy
A.
Cai
G.
Dacic
S.
Khalbuss
WE.
Pantanowitz
L.
Monaco
SE.
Evaluation of endobronchial ultrasound-guided fine-needle aspirations (EBUS-FNA): correlation with adequacy and histologic follow-up
.
Cancer Cytopathol
.
2014
;
122
(
1
):
23
32
.
28
Dong
X.
Qiu
X.
Liu
Q.
Jia
J.
Endobronchial ultrasound-guided transbronchial needle aspiration in the mediastinal staging of non-small cell lung cancer: a meta-analysis
.
Ann Thorac Surg
.
2013
;
96
(
4
):
1502
1507
.
29
Herth
FJ.
Eberhardt
R.
Vilmann
P.
Krasnik
M.
Ernst
A.
Real-time endobronchial ultrasound guided transbronchial needle aspiration for sampling mediastinal lymph nodes
.
Thorax
.
2006
;
61
(
9
):
795
798
.
30
Ye
T.
Hu
H.
Luo
X.
Chen
H.
The role of endobronchial ultrasound guided transbronchial needle aspiration (EBUS-TBNA) for qualitative diagnosis of mediastinal and hilar lymphadenopathy: a prospective analysis
.
BMC Cancer
.
2011
;
11
:
100
.
31
Lee
BE.
Kletsman
E.
Rutledge
JR.
Korst
RJ.
Utility of endobronchial ultrasound-guided mediastinal lymph node biopsy in patients with non-small cell lung cancer
.
J Thorac Cardiovasc Surg
.
2012
;
143
(
3
):
585
590
.
32
Cerfolio
RJ.
Bryant
AS.
Eloubeidi
MA.
et al.
The true false negative rates of esophageal and endobronchial ultrasound in the staging of mediastinal lymph nodes in patients with non-small cell lung cancer
.
Ann Thorac Surg
.
2010
;
90
(
2
):
427
434
.
33
Mehta
HJ.
Tanner
NT.
Silvestri
G.
et al.
Outcome of patients with negative and unsatisfactory cytologic specimens obtained by endobronchial ultrasound-guided transbronchial fine-needle aspiration of mediastinal lymph nodes
.
Cancer Cytopathol
.
2015
;
123
(
2
):
92
97
.
34
VanderLaan
PA.
Wang
HH.
Majid
A.
Folch
E.
Endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA): an overview and update for the cytopathologist
.
Cancer Cytopathol
.
2014
;
122
(
8
):
561
576
.
35
Louw
M.
Brundyn
K.
Schubert
PT.
Wright
CA.
Bolliger
CT.
Diacon
AH.
Comparison of the quality of smears in transbronchial fine-needle aspirates using two staining methods for rapid on-site evaluation
.
Diagn Cytopathol
.
2012
;
40
(
9
):
777
781
.
36
Meena
N.
Jeffus
S.
Massoll
N.
et al.
Rapid onsite evaluation: A comparison of cytopathologist and pulmonologist performance
.
Cancer Cytopathol
.
2016
;
124
(
4
):
279
284
.
37
Shield
PW.
Cosier
J.
Ellerby
G.
Gartrell
M.
Papadimos
D.
Rapid on-site evaluation of fine needle aspiration specimens by cytology scientists: a review of 3032 specimens
.
Cytopathology
.
2014
;
25
(
5
):
322
329
.
38
Bonifazi
M.
Sediari
M.
Ferretti
M.
et al.
The role of the pulmonologist in rapid on-site cytologic evaluation of transbronchial needle aspiration: a prospective study
.
Chest
.
2014
;
145
(
1
):
60
65
.
39
Khurana
KK.
Kovalovsky
A.
Wang
D.
Lenox
R.
Feasibility of dynamic telecytopathology for rapid on-site evaluation of endobronchial ultrasound-guided transbronchial fine needle aspiration
.
Telemed J E Health
.
2013
;
19
(
4
):
265
271
.
40
Bott
MJ.
James
B.
Collins
BT.
et al.
A prospective clinical trial of telecytopathology for rapid interpretation of specimens obtained during endobronchial ultrasound-fine needle aspiration
.
Ann Thorac Surg
.
2015
;
100
(
1
):
201
206
.
41
Vallonthaiel
AG.
Jain
D.
Madan
K.
Arava
S.
Pulmonary adenocarcinoma with signet ring features: detailed cytomorphologic analysis
.
Diagn Cytopathol
.
2016
;
44
(
7
):
607
611
.
42
Kumari
K.
Jain
D.
Kumar
R.
Mohan
A.
Kumar
R.
Metastatic carotid body paraganglioma of lungs and lymph nodes: unsuspected diagnosis on EBUS-TBNA
.
Diagn Cytopathol
.
2017
;
45
(
4
):
327
332
.
43
Agarwal
R.
Srinivasan
A.
Aggarwal
AN.
Gupta
D.
Efficacy and safety of convex probe EBUS-TBNA in sarcoidosis: a systematic review and meta-analysis
.
Respir Med
.
2012
;
106
(
6
):
883
892
.
44
Alsharif
M.
Anrade
RS.
Groth
SS.
Stelow
EB.
Pambuccian
SE.
Endobronchial ultrasound guided transbronchial fine needle aspiration: the University of Minnesota experience, with emphasis on usefulness, adequacy assessment, and diagnostic difficulties
.
Am J Clin Pathol
.
2008
;
130
(
3
):
434
443
.
45
Nayak
A.
Sugrue
C.
Koenig
S.
Wasserman
PG.
Morgenstern
NJ.
Endobronchial ultrasound-guided transbronchial needle aspirate (EBUS-TBNA): a proposal for on-site adequacy criteria
.
Diagn Cytopathol
.
2012
;
40
(
2
):
128
137
.
46
Choi
SM.
Lee
AR.
Choe
JY.
et al.
Adequacy criteria of rapid on-site evaluation for endobronchial ultrasound-guided transbronchial needle aspiration: a simple algorithm to assess the adequacy of ROSE
.
Ann Thorac Surg
.
2016
;
101
(
2
):
444
450
.
47
Sørhaug
S.
Hjelde
H.
Hatlen
P.
et al.
Learning EBUS-TBNA - a 6-year experience at a single institution [published online ahead of print
March
8,
2016]
.
Clin Respir J
. doi:.
48
Griffin
AC.
Schwartz
LE.
Baloch
ZW.
Utility of on-site evaluation of endobronchial ultrasound-guided transbronchial needle aspiration specimens
.
Cytojournal
.
2011
;
8
:
20
.
49
Joseph
M.
Jones
T.
Lutterbie
Y.
et al.
Rapid on-site pathologic evaluation does not increase the efficacy of endobronchial ultrasonographic biopsy for mediastinal staging
.
Ann Thorac Surg
.
2013
;
96
(
2
):
403
410
.
50
Oki
M.
Saka
H.
Kitagawa
C.
et al.
Rapid on-site cytologic evaluation during endobronchial ultrasound-guided transbronchial needle aspiration for diagnosing lung cancer: a randomized study
.
Respiration
.
2013
;
85
(
6
):
486
492
.
51
Collins
BT.
Chen
AC.
Wang
JF.
Bernadt
CT.
Sanati
S.
Improved laboratory resource utilization and patient care with the use of rapid on-site evaluation for endobronchial ultrasound fine-needle aspiration biopsy
.
Cancer Cytopathol
.
2013
;
121
(
10
):
544
551
.
52
Murakami
Y.
Oki
M.
Saka
H.
et al.
Endobronchial ultrasound-guided transbronchial needle aspiration in the diagnosis of small cell lung cancer
.
Respir Investig
.
2014
;
52
(
3
):
173
178
.
53
Trisolini
R.
Cancellieri
A.
Tinelli
C.
et al.
Randomized trial of endobronchial ultrasound-guided transbronchial needle aspiration with and without rapid on-site evaluation for lung cancer genotyping
.
Chest
.
2015
;
148
(
6
):
1430
1437
.
54
Cardoso
AV.
Neves
I.
Magalhães
A.
Sucena
M.
Barroca
H.
Fernandes
G.
The value of rapid on-site evaluation during EBUS-TBNA
.
Rev Port Pneumol
(
2006
).
2015;
21
(
5
):
253
258
.
55
Dyhdalo
KS.
Booth
CN.
Shorie
J.
Underwood
DL.
Mazzone
P.
Brainard
JA.
Rapid on-site evaluation of endobronchial ultrasound guided fine-needle aspirates: correlation of adequacy assessment and final diagnosis in patients with bronchogenic carcinoma
.
J Am Soc Cytopathol
.
2014
;
3
(
4
):
199
205
.
56
Lindeman
NI.
Cagle
PT.
Beasley
MB.
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
.
Arch Pathol Lab Med
.
2013
;
137
(
6
):
828
860
.
57
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
(
6
):
356
371
.
58
Askoxylakis
V.
Thieke
C.
Pleger
ST.
et al.
Long-term survival of cancer patients compared to heart failure and stroke: a systematic review
.
BMC Cancer
.
2010
;
10
:
105
.
59
Travis
WD.
Brambilla
E.
Noguchi
M.
et al.
International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society international multidisciplinary classification of lung adenocarcinoma
.
J Thorac Oncol
.
2011
;
6
(
2
):
244
285
.
60
Hopkins
E.
Moffat
D.
Parkinson
I.
et al.
Cell block samples from endobronchial ultrasound transbronchial needle aspiration provide sufficient material for ancillary testing in lung cancer-a quaternary referral centre experience
.
J Thorac Dis
.
2016
;
8
(
9
):
2544
2550
.
61
Fernandez-Bussy
S.
Labarca
G.
Pires
Y.
Caviedes
I.
Burotto
M.
Molecular testing of EGFR, EGFR resistance mutation, ALK and ROS1 achieved by EBUS-TBNA in Chile
.
Arch Bronconeumol
.
2017
;
53
(
3
):
172
174
.
62
Guisier
F.
Salaün
M.
Lachkar
S.
et al.
Molecular analysis of peripheral non-squamous non-small cell lung cancer sampled by radial EBUS
.
Respirology
.
2016
;
21
(
4
):
718
726
.
63
Kirita
K.
Izumo
T.
Matsumoto
Y.
Hiraishi
Y.
Tsuchida
T.
Bronchoscopic re-biopsy for mutational analysis of non-small cell lung cancer
.
Lung
.
2016
;
194
(
3
):
371
378
.
64
Schmid-Bindert
G.
Wang
Y.
Jiang
H.
et al.
EBUS-TBNA provides highest RNA yield for multiple biomarker testing from routinely obtained small biopsies in non-small cell lung cancer patients – a comparative study of three different minimal invasive sampling methods
.
PLoS One
.
2013
;
8
(
10
):
e77948
.
65
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
.
66
van der Heijden
EH.
Casal
RF.
Trisolini
R.
et al
World Association for Bronchology and Interventional Pulmonology, Task Force on Specimen Guidelines. Guideline for the acquisition and preparation of conventional and endobronchial ultrasound-guided transbronchial needle aspiration specimens for the diagnosis and molecular testing of patients with known or suspected lung cancer
.
Respiration
.
2014
;
88
(
6
):
500
517
.
67
Layfield
LJ.
Bentz
JS.
Gopez
EV.
Immediate on-site interpretation of fine-needle aspiration smears: a cost and compensation analysis
.
Cancer
.
2001
;
93
(
5
):
319
322
.
68
Nasuti
JF.
Gupta
PK.
Baloch
ZW.
Diagnostic value and cost-effectiveness of on-site evaluation of fine-needle aspiration specimens: review of 5,688 cases
.
Diagn Cytopathol
.
2002
;
27
(
1
):
1
4
.
69
Detterbeck
FC.
Mazzone
PJ.
Naidich
DP.
Bach
PB.
Screening for lung cancer: diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines
.
Chest
.
2013
;
143
(
suppl 5
):
e78S
92S
.
70
Eapen
GA.
Shah
AM.
Lei
X.
et al
American College of Chest Physicians Quality Improvement Registry, Education. Complications, consequences, and practice patterns of endobronchial ultrasound-guided transbronchial needle aspiration: results of the AQuIRE registry
.
Chest
.
2013
;
143
(
4
):
1044
1053
.
71
Trisolini
R.
Cancellieri
A.
Tinelli
C.
et al.
Rapid on-site evaluation of transbronchial aspirates in the diagnosis of hilar and mediastinaladenopathy: a randomized trial
.
Chest
.
2011
;
139
(
2
):
395
401
.

Author notes

From the Department of Pathology, All India Institute of Medical Sciences, New Delhi, India (Dr Jain); the Department of Pathology, University of Texas Medical Branch, Galveston (Dr Allen); the Department of Pathology, University of Colorado, Aurora (Dr Aisner); the Department of Pathology, Mount Sinai Medical Center, New York, New York (Dr Beasley); the Department of Pathology & Genomic Medicine, Houston Methodist Hospital, Houston, Texas (Drs Cagle and Miller); the Department of Pathology, University of São Paulo, São Paulo, Brazil (Dr Capelozzi); the Department of Pathology, Massachusetts General Hospital, Boston (Drs Hariri and Mino-Kenudson); the Département de Biopathologie, Centre Léon Bérard, Lyon, France (Dr Lantuejoul); Université Joseph Fourier INSERM U 823, Institut A. Bonniot, La Tronche, France (Dr Lantuejoul); the Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (Dr Monaco); the Department of Pathology, New York University Langone Medical Center, New York, New York (Dr Moreira); the Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois (Dr Raparia); the Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (Dr Rekhtman); the Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota (Dr Roden); the Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston (Dr Roy-Chowdhuri); the Department of Laboratory Medicine and Pathobiology, University of Toronto University Health Network, Toronto, Ontario, Canada (Dr Santos); the Department of Pathology, VU Medical Center, Amsterdam, the Netherlands (Dr Thunnissen); the Dipartimento di Sanità Pubblica, Università degli Studi di Napoli Federico II, Naples, Italy (Dr Troncone); and the Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts (Dr Vivero).

The authors have no relevant financial interest in the products or companies described in this article.