The use of immunohistochemistry for the determination of pulmonary carcinoma biomarkers is a well-established and powerful technique. Immunohistochemisty is readily available in pathology laboratories, is relatively easy to perform and assess, can provide clinically meaningful results very quickly, and is relatively inexpensive. Pulmonary predictive biomarkers provide results essential for timely and accurate therapeutic decision making; for patients with metastatic non–small cell lung cancer, predictive immunohistochemistry includes ALK and programmed death ligand-1 (PD-L1) (ROS1, EGFR in Europe) testing. Handling along proper methodologic lines is needed to ensure patients receive the most accurate and representative test outcomes.

Pulmonary predictive biomarkers are defined as markers for which the results are essential for therapeutic decision making. In the setting of metastatic non–small cell lung cancer (NSCLC), predictive immunohistochemistry (IHC) includes ALK and programmed death ligand-1 (PD-L1) (ROS1, EGFR in Europe). In the cases of ALK and ROS1 IHC, positive staining serves either as a screening test or as a surrogate test for fusion in the indicated gene, resulting in aberrant expression of the gene products; and in the case of PD-L1, positive staining indicates the presence of an immunomodulatory molecule that is affected by anti–PD-L1 or anti–programmed death receptor-1 (PD-1, CD279) therapy. The resulting treatment consequences for patients are as follows: for ALK+ NSCLC, an ALK inhibitor; for ROS1+ NSCLC, a ROS1 inhibitor; and for PD-L1+ NSCLC, an inhibitor of PD-1 or PD-L1. Pulmonary predictive biomarker IHC is not the sole therapeutic decision maker; an individual patient's treatment also depends on other factors, such as tumor stage, performance score, access to targeted agents, and patient and clinician preference and experience. For a patient eligible for tyrosine kinase inhibitor and immunotherapy, the first treatment of choice may be the tyrosine kinase inhibitor. It is important to differentiate the therapeutic decision-making role of pulmonary predictive biomarker IHC from IHC performed for diagnostic purposes, which, in contrast, serves a diagnostically supportive or decisive role. The latter can be critical in distinguishing NSCLC subtypes, which itself has therapeutic implications.1,2 

Many IHC antibodies are dependent upon the fixation procedure for success: some work on frozen sections, but not on formalin-fixed paraffin-embedded (FFPE) samples, and vice versa. This article restricts discussion of IHC pulmonary biomarkers to FFPE material and the IHC applications of ALK rearrangement, ROS1 rearrangement, and BRAF V600E mutation, as well as PD-L1 expression in tumor cells and/or adjacent inflammatory cells. Automated IHC protocols for ALK and PD-L1 detection were recently approved by the US Food and Drug Administration (FDA) as companion diagnostics for targeted therapies. Issues related to these companion diagnostics have been recently reviewed.3,4  Immunohistochemistry is a rapid and relatively inexpensive method, preferred by most pathologists, primarily because it allows the simultaneous evaluation of a staining pattern and tissue architecture and tumor cells. Potentially, IHC can be interpreted with fewer malignant cells than are needed for successful interpretation by fluorescence in situ hybridization (FISH) or other molecular technologies. Immunohistochemistry can be performed efficaciously on a variety of different tumor specimens. FFPE tissue blocks, fluid, and fine-needle aspiration cytology cell blocks or smears can be tested successfully as long as there are at least a few clusters of viable tumor cells present in the specimen. However, there are limitations; currently, scarcity of predictive trial data regarding the application of IHC to cytology prohibits any definitive determination regarding the value of cytology specimens for pulmonary predictive biomarker IHC. This may be due to trial designers' lack of familiarity with the advantages and limitations of cytology, and their preference of biopsy material for additional translational research in the trial.5  Nevertheless, excellent studies exist on the application of ALK on cytology.5,6  Also, molecular genetic aberrations with low prevalence, such as ALK- and ROS1-rearranged NSCLC, call for economic screening methods, and a validated and robust IHC assay may provide a cost-effective platform.

Preanalytic variables start at the moment the biopsy or resection specimen is removed from the patient. These variables include delay in fixation, inappropriate fixation time, and issues regarding paraffin embedding.7  Cold ischemia time, the time between specimen removal out of patient and initiation of fixation, should be less than 1 hour. Specimens should immediately be fixed in an adequate amount (10 times the volume of the specimen) of neutral buffered 10% formalin and embedded in melted paraffin.7,8  In cytology, alcohol fixation is sometimes used.5  However, when formalin-optimized IHC protocols are used, the accuracy of IHC may be reduced by causing loss or decrease of immunogenicity. Immunohistochemistry protocol adjustments restore immunogenicity for most but not all of the antigens.9  Formalin fixation times of less than 6 hours are not recommended because conventional staining as well as IHC can be adversely affected, for example, for ALK.10  For practical purposes, a fixation interval of 6 to 24 hours for biopsy specimens, and 24 to 72 hours for resection specimens, is recommended.11,12 

Because formalin diffusion time is approximately 1 to 3 mm/h, at gross dissection tumor should be cut in slices of 3- to 4-mm thickness. FFPE samples are considered stable, preserved, and protected from oxidative influences. FFPE tissue specimens should be cut into 3- to 5-μm sections for placement on glass slides. If unstained sections are not used within a few days, they should be stored in a closed container at 2°C to 8°C to preserve antigenicity from photooxidation and drying, which could cause false-negative results.13  Decalcification can negatively affect antigenicity, especially when highly acidic agents such as hydrochloric acid and nitric acid are used. Therefore, weaker acids such as ethylenediaminetetraacetic acid (EDTA), an effective decalcifying agent, have become more popular, as they do not appear to interfere with IHC14 ; in addition, in many small sample bone biopsies, decalcification can be completely avoided.

For the analytic procedure, several issues need to be controlled and optimized, including the development of adequate antibodies, antigen retrieval, type and concentration of the antibody, incubation time, incubation temperature, and signal enhancement (eg, with a tyramide cascade and intercalation of an antibody-enhanced polymer). Antigen preservation for IHC is epitope dependent, and some epitopes may not be hampered by fixation times—as long as 120 hours. The cross-links due to formalin fixation may hamper the epitope recognition by the primary antibody. To this end different epitope retrieval buffers are tested and the optimal buffer chosen for the standard staining procedure.

The postanalytic phase starts with the glass slide's microscopic evaluation. Staining intensity is dependent on the enhancement system used. The relationship between the enhancement system and signal intensity was shown in 2003 by Prinsen et al15  and is graphically illustrated in Figure 1. The subjectivity of staining intensity assessment can be reduced with the use of successive microscope objective lenses with inherent related spatial resolution as a physical aid in establishing the intensity level. This approach, first applied to HER2 testing, may lead to more uniform intensity scoring.16  Strong staining (+++) is clearly visible with use of an ×2 or ×4 microscope objective lens; moderate staining (++) requires an ×10 or ×20 objective lens to be clearly seen; and weak staining (+) can be seen only with an ×40 objective lens.17  The histo-score (H-score) is derived by multiplying the percentage of tumor cells that stain positively by different intensities (0, 1 = +, 2 = ++, or 3 = +++), giving a range of 0 to 300. This approach takes greater account of the heterogeneity of the staining. Interestingly, with additional tyramide enhancement, the difference in epitope concentration between a negative and a strong positive staining intensity is reduced to the extent that scoring is restricted to either negative or positive (Figure 1). Strong signal enhancement may have important consequences. Of course, proper titration of primary antibody and enhancement components is a prerequisite. A strong enhancement system may in some cases reveal a positive test result, while an approach using a less strong signal enhancement may be negative. This implies that once a test is clinically validated, only tests with equal test performance can be used. Otherwise, the generalization of a phase III trial toward a presumed predicted chance of a response to a given drug will not hold.

Figure 1

Relationship between immunohistochemistry epitope concentration and intensity, modified from Prinsen et al15  with permission from Wolters Kluwer Health, Inc. Blue = direct labeling of primary antibody; red = streptavidin-biotin signal enhancement; green = polymer plus tyramide amplification signal enhancement. Abbreviation: A.U., arbitrary unit.

Figure 2The critical sample set is determined by testing (1) originally positive samples with a 2-fold reduced concentration of the primary antibody and (2) originally negative samples with a 2-fold increase in concentration. Cases reversing in the test outcome are critical samples (green) with an epitope concentration close to the positivity threshold, while samples outside this range are not informative (red) for clinical validation of different immunohistochemistry (IHC) tests. Abbreviation: A.U., arbitrary unit.

Figure 1

Relationship between immunohistochemistry epitope concentration and intensity, modified from Prinsen et al15  with permission from Wolters Kluwer Health, Inc. Blue = direct labeling of primary antibody; red = streptavidin-biotin signal enhancement; green = polymer plus tyramide amplification signal enhancement. Abbreviation: A.U., arbitrary unit.

Figure 2The critical sample set is determined by testing (1) originally positive samples with a 2-fold reduced concentration of the primary antibody and (2) originally negative samples with a 2-fold increase in concentration. Cases reversing in the test outcome are critical samples (green) with an epitope concentration close to the positivity threshold, while samples outside this range are not informative (red) for clinical validation of different immunohistochemistry (IHC) tests. Abbreviation: A.U., arbitrary unit.

Close modal

Standardization of both positive and negative controls is needed in predictive IHC, since the use of polymer enhancement negative controls are no longer needed for diagnostic IHC in routine practice. In general, the use of IHC-negative tissue controls, irrespective of type, although well established, is not globally standardized. As such, the relevance and applicability of negative tissue controls continues to challenge both pathologists and laboratory budgets. Negative reagent controls (based on a separate slide, where the primary antibody is replaced with another “irrelevant” antibody of same type and concentration, while all other protocol parameters are identical) are virtually useless, particularly after implementation of biotin-free visualization systems. Despite the clear theoretical notion that appropriate tissue controls serve to demonstrate the sensitivity and specificity of the IHC test, it remains unclear which types of positive and negative controls are applicable or useful in day-to-day clinical practice.18,19  Recently, the term immunohistochemistry critical assay performance controls (iCAPCs), was introduced.19  iCAPCs are external positive controls. iCAPCs monitor the overall system performance, but like any other external positive control, they do not fully inform the patient's results because final results also significantly depend on preanalytic variables unique to the patient's specimen. The optimal IHC positive control has an intensity performance at or above the lower limit of detection and is defined by an observed positive reaction (staining) in a tissue/cellular element that is known to express low levels of the evaluated marker.19 

The use of an internal positive control tissue is generally perceived as adequate and an external control might not be needed. However, it is useful to maintain an external positive control for most stains because not every section will contain an internal positive control, and internal positive controls, when present, typically display a relatively high epitope concentration. As such, they rarely provide information as to whether the appropriate sensitivity of the protocol was achieved. This gap is filled by the external positive control with low epitope concentration. External positive controls are increasingly being mounted on the same slide as the test section as a means of improving control of test IHC staining.

As with all IHC stains, artifacts may occur, including nonspecific background changes (in relation to improper drying of the slides, improper deparaffinization, or incomplete rinsing); and edge artifacts (due to drying of the tissue before fixation or during the staining procedure); and crush artifacts (due to necrosis or poor fixation).

Validation

For diagnostic testing according to the College of American Pathologists guidelines, a minimum of 10 samples is required for technical validation.20  In contrast to ALK and ROS1, where the underlying oncogenetic mechanism is gene rearrangement, PD-L1 is a protein with no oncogenetic change. Instead PD-L1 is part of the physiologic immune defense system. The validation of ALK and ROS1 is performed by comparison with tests demonstrating the associated rearrangement (such as FISH or next-generation sequencing–based testing). A practical difficulty is that it takes a long time to acquire 10 positive samples in low prevalent disease (eg, ROS1) for initial setup of the IHC. Thus, careful testing with fewer samples may be a way forward. For PD-L1 the approach is different and is discussed below.

Genomic rearrangements involving ALK occur in 1% to 5% of NSCLCs. (In 2013 crizotinib became second-line treatment for ALK+ NSCLC21  and since 2014 it has generally been used as the first-line treatment option.22) This therapeutic option was based on ALK FISH assay subsequently approved by the FDA. More recently, IHC has been accepted as a screening method for ALK+ NSCLC.

ALK Preanalytic Variables

If preanalytic conditions are appropriately controlled, most tumor cells stain with IHC, paralleling the homogeneous distribution of the ALK gene rearrangement in FISH analysis (Table 1).

Table 1

Distribution of ALK Immunohistochemistry (IHC+) Studies With n > 100 Cases, Using 5A4 and/or D5F3 Antibody in Non–Small Cell Lung Cancer (NSCLC), With Percentage ALK IHC Positive (% ALK+) Cases and Fraction of Discordant Cases With ALK Fluorescence In Situ Hybridization (FISH) (IHC+/FISH)

Distribution of ALK Immunohistochemistry (IHC+) Studies With n > 100 Cases, Using 5A4 and/or D5F3 Antibody in Non–Small Cell Lung Cancer (NSCLC), With Percentage ALK IHC Positive (% ALK+) Cases and Fraction of Discordant Cases With ALK Fluorescence In Situ Hybridization (FISH) (IHC+/FISH−)
Distribution of ALK Immunohistochemistry (IHC+) Studies With n > 100 Cases, Using 5A4 and/or D5F3 Antibody in Non–Small Cell Lung Cancer (NSCLC), With Percentage ALK IHC Positive (% ALK+) Cases and Fraction of Discordant Cases With ALK Fluorescence In Situ Hybridization (FISH) (IHC+/FISH−)

Initially, several different techniques were used for detecting ALK IHC.3  In these studies, the type or source of antibodies, the process of antigen retrieval and antibody detection, and the amplification techniques varied substantially. Eventually, head-to-head comparison of different antibodies showed that D5F3 (Cell Signaling Technology, Danvers, Massachusetts) and 5A4 (Novocastra, Leica Biosystems, Buffalo Grove, Illinois) with the ADVANCE system (Dako, Santa Clara, California) were equally sensitive.23  D5F3 is part of a commercial companion diagnostic ALK IHC assay kit, whereas 5A4 is commonly used as a laboratory-developed test (LDT).24 

The ALK1 antibody (Dako) is less accurate and should not be used. A novel monoclonal anti-ALK antibody, 1A4 (Origene, Rockville, Maryland), was compared with D5F3 and described as a promising candidate for screening lung tumors for the presence of ALK rearrangements25 ; however, the 1A4 antibody examined in an independent cohort showed comparable sensitivity to D5F3, but lower specificity (70%).26  Therefore, tumors that are positive for ALK on testing with 1A4 IHC will require an additional predictive technique before treatment advice can be rendered.

The D5F3-based immunoassay (Ventana ALK [D5F3] CDx Assay, Tucson, Arizona) was developed and standardized on the automated immunostaining platform BenchMark XT combined with the OptiView Amplification Kit. Interestingly, with tyramide enhancement the difference in epitope concentration between a negative and a strong positive staining intensity is reduced to the extent that scoring is either negative or positive. This tyramide enhancement works similarly for both of the currently used antibodies, D5F3 and 5A4.27,28  The reproducibility of ALK IHC results among different laboratories and pathologists is high for the 2 validated protocols.24,28 

ALK Postanalytic Variables

Because ALK protein is not expressed in normal, mature lung tissue, strong IHC amplification systems can generally be used as a marker for tumor ALK positivity; however, pathologists should be familiar with various artifacts that may lead to false-positive staining. Positive ALK IHC typically shows strong granular cytoplasmic staining. Cytoplasmic staining may occur in alveolar macrophages, cells of neural origin (nerve and ganglion cells including within tumors), glandular epithelium, extracellular mucin, and areas of necrotic tumor.29  Background staining is rarely observed within normal lung parenchyma. False-positive cytoplasmic staining in NSCLC has been noted with the tyramide amplification system using D5F3.28  This stain may show weaker than usual positivity in lung cancers that are ALK+ on IHC.

Histologically, mucin-containing cells such as signet ring cells require careful interpretation of ALK immunoreactivity, which is particularly relevant given frequent signet ring cell morphology of ALK-rearranged adenocarcinomas. A thin membranous positive pattern on ALK IHC may be masked by an intracellular mucin vacuole, and the positive pattern may then be difficult to detect in the signet ring cells.3032  This finding is not specific to cancer cells and has also been seen in some nontumor cells, such as reactive type II pneumocytes. Thus, it is important to recognize background staining in the stained specimens. In addition, some neuroendocrine carcinomas have also been associated with positive reactions in the absence of ALK rearrangements.33,34  Merkel cell tumors of the skin may also be ALK+ on IHC but have no ALK rearrangement detected by FISH or next-generation sequencing. The paranuclear dotlike pattern reported as typical of the KIF5B-ALK rearrangement may require further confirmation.35 

The staining may appear heterogeneous in some tumors, particularly in surgical specimens. This heterogeneity is likely related to delay in the fixation of more deeply situated tissues, due to slow fixative diffusion and is not indicative of a different histologic pattern. The sensitivity of ALK protein to fixation delay is typically not an issue for biopsy specimens, but may be an issue when using tissue microarrays for ALK screening of archived specimens.

ALK Validation

Initially in the United States when the FDA approved the ALK FISH assay, patients with positive results on ALK FISH were eligible for treatment with an ALK inhibitor. The European Medicine Agency (EMA) approved patients with ALK+ metastatic NSCLC as eligible for treatment with an ALK inhibitor, thus not specified to FISH or IHC. More recently, the FDA has approved the IHC CDx Assay, and patients with positive results on ALK D5F3 IHC are now also eligible for treatment with an ALK inhibitor.

In several studies, D5F3 IHC was compared with ALK FISH and showed high concordance rates.3646  The sensitivity was shown to range from 81% to 100%, and specificity from 82% to 100%. The interobserver reproducibility for D5F3 in a selected series of adenocarcinomas is high.47 

Several studies4856  compared 5A4 IHC with ALK FISH. The sensitivity ranged from 93% to 100% and specificity from 96% to 100%.

In a recent analysis of pooled data, the diagnostic operating characteristics in 12 studies (3754 NSCLC specimens) were analyzed, taking the different scoring systems into account.57  The IHC 3+ binary ALK+ category matched for both antibody procedures with ALK FISH+ cases, and the IHC cases matched FISH cases. The nearly 100% concordance in these IHC categories favors the use of IHC as a screening method to identify ALK+ NSCLC. However, for the lower-intensity staining in the 4-tiered IHC approach, tumors with 1+ and 2+ intensity need additional validation with ALK FISH testing.

Discordant NSCLC cases, with ALK protein expression (for D5F3/5A4) but no detectable ALK rearrangement on FISH, have been reported.* Explanations for this discordance include (1) false-negative interpretation of FISH results, especially for results that are close to the threshold of 15% and below 20%46,61,65; (2) amplification of the ALK gene (which has been associated with ALK protein expression in some but not all cases), possibly leading to 1+ or 2+ staining67,68; (3) false-positive interpretation of ALK IHC results; (4) complex rearrangement involving the ALK gene, reducing the visible distance of the 2 FISH probes, leading to false-negative FISH findings; (5) de novo alternative transcription initiation69 ; and (6) additional yet undetermined mechanism(s). Some of the ALK IHC+, FISH cases respond to crizotinib.59,70 

ALK IHC assays, already recommended by organizations in Europe, Japan, Asia, and the United States, are validated and standardized and are clinical tools for cost-effective screening for the presence of ALK rearrangement in NSCLC. To maintain the reliability of assays for detecting ALK positivity, laboratories should participate regularly in external quality assessment programs.

The pathologic significance of ROS1 expression has recently been reviewed.34  Genomic rearrangements involving ROS1 occur in 1% to 2% of unselected NSCLCs.7173  ROS1 fusion partners in lung cancer include FIG, CD74, SLC34A2, and SDC4.7477  Crizotinib was recently approved by the FDA for patients with advanced ROS1+ NSCLC.78  ROS1 may be detected by a variety of techniques, including FISH, reverse transcription–polymerase chain reaction (RT-PCR), next-generation sequencing, and IHC. This discussion focuses on IHC. Predictive testing for ROS1 rearrangement started recently, and as such, less information is currently available for ROS1 than for ALK testing. ROS1 IHC may be an effective screening tool to detect ROS1+ NSCLC.

ROS1 Preanalytic Variables

It is unclear as to whether or not the protein stability is affected by preanalytic variables.4  Most studies on ROS1 IHC use the D4D6 rabbit monoclonal antibody (Cell Signaling Technology) applied at dilutions ranging from 1:50 to 1:1000 with various antigen retrieval methods, with use of different amplification and detection systems, in automated instruments or with manual testing.79  In contrast to ALK, where the ganglion cells of the appendix serve as an adequate external control, there is currently no good external benign tissue control for ROS1.3,4  Tumor specimens with known ROS1 rearrangement, or a cell block of the HCC78 cell line harboring the SLC34A2-ROS1 fusion gene, can serve as external positive controls.4,74 

ROS1 Postanalytic Variables

Positive ROS1 IHC typically shows finely granular cytoplasmic staining; however, the staining pattern may depend on the function and subcellular location of the gene fusion partner.80  Globular ROS1 immunoreactivity has been described in tumor specimens with the CD74-ROS1 fusion, and membranous staining has been observed in tumors with the EZR-ROS1 fusion.79,80  Interestingly, ROS1 expression levels in ROS1-rearranged lung cancers and cell lines can vary from cell to cell, suggesting dynamic ROS1 protein expression despite homogeneous presence of ROS1 gene rearrangement. Detection of ROS1 protein expression in ROS1-rearranged adenocarcinomas with signet ring cells is challenging, since the cytoplasm may contain nonreactive mucin,80  similar to the situation with ALK IHC.4 

Studies reporting on ROS1 immunohistochemistry are shown in Table 2. Frequently, with D4D6, weak (1+ to 2+) to strong (3+) staining intensity has been reported. In the cases with strong intensity (88% [49 of 56]), rearrangement was confirmed with ROS1 FISH, while in the 1+ to 2+ positive cases, rearrangement could only be demonstrated in 14% (31 of 228) of the cases. Therefore, current understanding suggests screening with IHC and subsequent confirmation of the positive IHC cases with FISH. ROS1 inhibitors should only be given in the cases that are both IHC and FISH positive. Little is understood regarding ROS1 FISH+ cases with negative ROS1 IHC.

Table 2

ROS 1 Immunohistochemistry (IHC) Studies With D4D6 ROS1 Monoclonal Antibody in Non–Small Cell Lung Carcinoma (NSCLC) Showing the Relation With ROS1 Fluorescence In Situ Hybridization (FISH) for Strong IHC Positivity (IHC 3+) and Lower Intensity (IHC 1+/2+)

ROS 1 Immunohistochemistry (IHC) Studies With D4D6 ROS1 Monoclonal Antibody in Non–Small Cell Lung Carcinoma (NSCLC) Showing the Relation With ROS1 Fluorescence In Situ Hybridization (FISH) for Strong IHC Positivity (IHC 3+) and Lower Intensity (IHC 1+/2+)
ROS 1 Immunohistochemistry (IHC) Studies With D4D6 ROS1 Monoclonal Antibody in Non–Small Cell Lung Carcinoma (NSCLC) Showing the Relation With ROS1 Fluorescence In Situ Hybridization (FISH) for Strong IHC Positivity (IHC 3+) and Lower Intensity (IHC 1+/2+)

Weak ROS1 expression is occasionally detectable in nonneoplastic hyperplastic type II pneumocytes and in alveolar macrophages.81  In bone metastases, there is strong granular cytoplasmic staining of osteoclast-type giant cells.81  In most cases, the expression in these cells is weak to moderate (1+/2+ in intensity).3,4 

ROS1 Validation

As mentioned above, in cases with strong ROS1 staining with clone D4D6 IHC using a highly sensitive amplification kit, there is a high correlation with ROS1 FISH+. Although some discrepant cases have been reported,82  ROS1 testing by IHC is highly sensitive, but less specific, when compared with ALK IHC for detection of the corresponding gene rearrangement. Some authors3,4  suggest that IHC testing of specimens containing at least 20 tumor cells and application of an H-score cutoff of greater than 100 is highly concordant with ROS1 rearrangement by FISH or RT-PCR.

PD-L1 is a biomarker with some predictive value for immunotherapy, which is determined with different but not necessarily equal technologies.83  The pharmaceutical interaction is via PD-1 or PD-L1 inhibition, with 5 different companies having separate but associated proprietary anti–PD-L1 antibodies for use in IHC assays: nivolumab with 28-8 rabbit,84  pembrolizumab with 22C3 mouse,85  atezolizumab with SP142 rabbit, durvalumab with SP 263 rabbit,86  and avelumab with rabbit 73-10.87  The commercial companion PD-L1 test for pembrolizumab (Dako 22C3 pharmDx) and complementary diagnostic PD-L1 tests for nivolumab (Dako 28-8 pharmDx) and atezolizumab (Ventana PD-L1 SP142) are now FDA approved for use in NSCLC.

Pathology laboratories need at least 1 affordable, validated test, probably based on relatively high volume. It is not practical for pathology laboratories to perform 5 or more specific assays for 1 protein with the test used being dependent upon the drug that might possibly be prescribed in 1 facility.

In one study,88  comparison of the primary antibodies with immunohistochemical assessment did not reveal major differences, except for E1L3N, which is not included in one of the abovementioned tests.88  However, SP142 has lower proportion of positivity in another study (see below). Preferably, 1 PD-L1 IHC assay should be suitable for all pharmaceutical drugs that potentially could be prescribed, as that test essentially determines 1 PD-L1 protein within 1 NSCLC case. In addition, the use of multiple IHC cutoffs and proprietary PD-L1 IHC detection systems represents an immense barrier to interpretation of clinical trial biomarker data across trials. Laboratory feasibility issues are further complicated by the application of PD-L1 IHC staining approaches in other tumor types.

The staining procedures of the different commercially available assays are currently being compared to each other. One comparison based on technical validation with E1L3N and SP142 demonstrated prominent interassay variability or discordance.89  The methodology for clinical validation of laboratory-developed assays is discussed below.

Analytic Issues

For PD-L1, it is unknown whether fixation delay affects test results. Initially, it was advised that the tissue-handling procedure for PD-L1 be the same as for other diagnostic or predictive markers such as ALK.90  From the 22C3 and 28-8 pharmDx (Dako 22C3 pharmDx and Dako 28-8 pharmDx) interpretation manuals, a critical step for PD-L1 IHC is a short fixation time of less than 3 hours; other parameters such as prolonged fixation, paraffin embedding, dehydration, and use of unstained slides are not critical issues, as long as unstained slides are less than 6 months old. Specimen age for PD-L1 testing should be less than 3 years, as advanced age of tissue blocks in one PD-L1 study91  for the ATLANTIC phase II trial showed a similar percentage for tumor proportion score that was greater than 25% (around 30%), which dropped for specimens older than 3 years (14%).

The use of PD-L1 IHC on decalcified tissues should be interpreted with caution until further validation studies on PD-L1 IHC have become available. For LDTs, besides essential analytic validation, adequate clinical validation is of critical importance.

PD-L1 Interpretation

PD-L1 expression may be present on dendritic cells, macrophages, mast cells, T and B lymphocytes, endothelial cells, human placenta, and tumor cells.92,93  PD-L1 has 2 small hydrophilic regions for binding of IHC detection antibodies.94  The biological consequences of PD-L1 expression depend on cell membrane localization because it is presumed that PD-L1 is functional only when it ligates a counterreceptor.95 

Before determining a specimen's PD-L1 staining result, it is important to examine the hematoxylin-eosin–stained section to assess preservation and staining quality. If any staining of the PD-L1 IHC external control slide is unsatisfactory, all related patient specimen results should be considered invalid. For PD-L1, an external positive control FFPE cell block of human tonsils, placenta, and/or cell lines with variable PD-L1 expression (www.histocyte.com) may be used to develop the staining conditions. Care should be taken to use only cell lines with high epitope concentration, as the optimal staining control has weak epitope concentration, which allows detection of minor deviations in the staining protocol, whereas those with high epitope concentrations will not reveal a difference and remain positive.

Examination of PD-L1 NSCLC tissue helps to evaluate potential background staining. Any background staining should be of less than 1+ staining intensity. If plasma membrane staining of malignant cells occurs in the negative control tissue, all patient specimen results should be considered invalid. In any case, an NSCLC without staining for PD-L1 in tumor cells may still be good staining, representing a valid result.

When examining a patient's slide, necrotic or degenerate malignant cells should be excluded from evaluation. A minimum of 100 viable tumor cells should be present in the PD-L1–stained patient specimen slide for determination of the percentage of stained cells. Immune cells, as positive internal controls (eg, such as infiltrating lymphocytes or macrophages), may also stain with PD-L1. Any background staining should be of less than 1+ staining intensity.

Definition of Positive PD-L1 Staining in NSCLC

Unfortunately, the definition of “PD-L1 positivity” is not the same for the 5 potential PD-L1 assays. In 4 commercial assays, PD-L1 staining is defined as complete circumferential or partial linear plasma membrane staining of tumor cells at any intensity. Cytoplasmic staining only in tumor cells is not considered positive for scoring purposes. In the assay using SP142, the presence of PD-L1+ immune cells is also considered in determining the PD-L1 staining positivity. This seems to hamper the attempt to establish a single PD-L1 IHC test that will be considered equivalent to all clinically validated tests. However, other antibodies also stain immune cells. Whether or not this will ultimately lead to the use of 1 or 2 tests for broader application is unclear at present.

PD-L1 Scoring

It is important to score viable tumor cells exhibiting complete circumferential or partial linear plasma membrane staining at any intensity, and to determine the percentage of stained viable tumor cells in the entire specimen. PD-L1 protein expression is defined as the percentage of tumor cells exhibiting positive membranous staining at any intensity.

As a general scoring procedure, scan for an overview at ×4 objective magnification of the entire specimen. Subsequently, evaluate all areas regardless of whether well or poorly stained. Then, examine and score, at ×10 to ×40 objective magnification, viable tumor cells exhibiting complete circumferential or partial linear plasma membrane staining at any intensity. Exclude immune cells (except for SP142), normal cells, and necrotic cells from scoring. The absence of weak staining in tumor cells should be confirmed with ×20 objective examination.

Because PD-L1 heterogeneity may occur, scoring may be facilitated by dividing the slide into areas with equal amounts of “neoplastic cells” at low magnification, with evaluation of each area for percentage of PD-L1 positivity. The percentage of positivity from each area is then added, and divided by the total number of areas.

The immune activation response in the tumor cells at the tumor-stroma interface may lead to preferential expression of PD-L1 in this region. Small tumor biopsies may, however, miss the pertinent tumor-immune interface. In one study 92% concordance between biopsy and resection was reported,96  while in another the concordance rate was much lower (52%) with underestimations found with the biopsies.97  However, the latter study was unclear with regard to reporting the preanalytic variation. The authors used SP142, a slightly less sensitive antibody, and evaluated immune cells. These aspects clearly need more study.

Because in practice more biopsy specimens than resection specimens are stained for PD-L1, an inaccuracy in PD-L1 testing due to sampling of heterogenous tumors is unavoidable. To put the situation into context, the number of patients who respond to PD-1/PD-L1 treatment, whose tumors have negative PD-L1 IHC staining, is approximately 9%.98,99  If this is due to sampling error (PD-L1 tumor in the biopsy, while another part of the tumor is PD-L+) and if the variation in scoring around the predictive threshold is incorporated, then the effect of biological heterogeneity and variation in scoring seems lower than 10%. This notion is of great importance as this is the maximally attainable accuracy for PD-L1 IHC in daily practice.

PD-L1 Interpretation Cytology/Cell Blocks

Although 30% to 40% of all advanced NSCLCs are diagnosed by cytology alone, the use of cytology samples for determination of PD-L1 is currently not advocated, as none of the assays has been validated for this purpose. However, since immunostaining of tumor cells is standard diagnostic practice in many institutions,100  PD-L1 staining and quantitation should be technically feasible in principle, provided that appropriate protocols and quality control measures are in place. In contrast, quantitation of PD-L1+ immune cells with the SP142 assay approach will likely be more challenging. The lack of tissue architecture precludes distinction of the relevant immune cells between the tumor cells and at the epithelial-stromal interface from immune cells outside of the tumor boundaries that are considered as being irrelevant for PD-L1 scoring. Emerging data from cell blocks and matched histologic specimens suggest that cytologic material is as good as histologic material for PD-L1 IHC tumor cell analysis.101  However, preexisting lymphocytes in a fine-needle aspirate of a lymph node will remain a major confounding factor and this aspect of PD-L1 testing, especially regarding SP142, will not be solved.

For PD-L1 reporting, the predictive IHC test and threshold used should be reported as well as the controls in case of absence of PD-L1 staining in the NSCLC.

Pitfalls With PD-L1

PD-L1 IHC false positivity is generally due to 2 causes. First, PD-L1+ lymphocytes and histiocytes may lie between PD-L1 tumor cells and be misinterpreted as positive. Careful attention to the nuclear to cytoplasmic ratio and thin nuclear membrane of the macrophages is necessary. Second, cytoplasmic staining of tumor cells may be granular, but not truly membranous, and misinterpreted as positive.

PD-L1 Clinical Validation

As well as technical validation, clinical validation of selected thresholds is necessary to prove the predictive power and validity of the assay and the selected thresholds. The selected thresholds, validated in clinical trials, were at least in part derived from the most effective selection of patients who would, or would not, benefit from therapy, in a given clinical scenario, when compared to standard-of-care treatment (Table 3). This threshold forms the basis of the predictive IHC characteristic and is associated with a difference in response rates to the intended treatment. Establishing the threshold can only be achieved by comparison of a study group, in which (1) treatment with the intended drug is performed, (2) outcome data are known, and (3) (preferably graded) IHC biomarker data are known.

Table 3

Comparison Between Technical and Clinical Validation in Immunohistochemistry According to the College of American Pathologists (CAP) Guidelines

Comparison Between Technical and Clinical Validation in Immunohistochemistry According to the College of American Pathologists (CAP) Guidelines
Comparison Between Technical and Clinical Validation in Immunohistochemistry According to the College of American Pathologists (CAP) Guidelines

The predictive procedure, including preanalytic, analytic, and interpretation, needs to be robust in time to maintain the predictive value. In establishing the clinical validity of an LDT, IHC performance should be the same as that of the commercial clinically validated test. This implies that (1) the antibody should be titrated and/or incubation time varied to obtain the same signal; (2) the effect of the signal enhancement system should be equal; (3) in serial sections the PD-L1+ areas should be the same; (4) in approximately 10 PD-L1, 10 PD-L1+ samples and 20 critical predictive samples covering the linear dynamic range, the clinical validated PD-L1 IHC test should be the same; (5) as in general there is no calibration standard for IHC, thus, for selection of critical predictive samples the starting point is the intensity threshold of the initially validated test. Conceptually, those samples that are positive in a clinically validated test and become negative after dilution of the primary antibody to 50% fall within the linear dynamic range.15  Samples that remain positive are uninformative for comparison of different clinically validated assays on the same protein (ie, PD-L1). Samples that are negative in a clinically validated test and turn positive when the concentration of the primary antibody is increased 2-fold also fall within the linear dynamic range (Figure 2). However, if such a sample remains negative, it is not informative for comparison of different clinically validated assays on the same protein (ie, PD-L1). Only informative samples with an epitope/antigen concentration close to both sides of the clinically validated threshold are suitable for the critical predictive sample set. For comparison of the LDT with the clinically validated test, 90% of the samples should have an equal outcome, that is, similar to the External Quality Assessment standard in molecular pathology.102  Importantly, after selection of critical samples, consecutive sections should be stained with the clinically validated assay and the test to be compared (commercial or LDT), and the pattern of staining (including proportion of PD-L1+ neoplastic cells) should be more or less equal. Lastly, the laboratory should be certified and have standard operating procedures (including validation reports) for commercial test/LDT in place to maintain robust testing in time; and (6) the laboratory should regularly and successfully participate in external quality assessment program. If any of these requirements are not fulfilled, generalization of the predictive biomarker cannot be guaranteed and performance for clinical testing should be immediately stopped.

The recent studies on comparison for different commercial PD-L1 IHC assays will be discussed along the methodologic notions discussed above.

In the “Blueprint 1” study, cases were selected to “represent the respective dynamic range of each of the 4 PD-L1 assays,” but the authors103  do not mention how the cases were actually selected. Staining was performed centrally and interpreted separately by 3 companies' pathologists using assay-specific cutoff values. In fact, 14 cases show discordances with assay-specific cutoffs. In comparing 22-8 with 22C3, 2 of 14 (14%) are discordant; 22-8 with Sp263, 5 of 14 (36%) are discordant; and 22C3 with SP263, 6 of 14 (43%) are discordant. This comparison does not attempt to adjust one assay to achieve validation from one assay to the other. From the remaining 24 cases (19 positive by all 4 assays and 5 negative by all 4 assays), it is unclear whether these are really critical samples or not.

The German “Harmonization study”129  examined 15 cases most with high concordance but 4 with systematic differences. To quantify potential differences in the proportions of stained carcinoma cells, the 6 pairwise comparisons of the 4 assays were calculated. For 28-8 versus 22C3, 72% of the pairs were concordant; 13% showed higher proportions for 28-8 and 16% higher proportions for 22C3. Comparisons of SP263 with the other assays indicated higher proportions for SP263 in 46% (28-8), 44% (22C3), and 59% (SP142) of the pairs. Comparisons of SP142 indicated lower proportions in 36% (28-8), 39% (22C3), and 59% (SP263) of the pairs. In this study the selection of cases was from a comprehensively annotated patient cohort to include both adenocarcinoma and squamous cell carcinomas to mimic the composition of clinical trials. A relation to the dynamic range of the assays was not provided.

To establish a representative value for interobserver variation, studies examining consecutive series of NSCLCs should be performed. Currently, in a German study with highly selected cases the interobserver variation at the 1% threshold varied for all 4 assays from 2.8% to 9.6%, and at the 50% threshold ranged from 5.2% to 8.5%.104  At the 16th World Conference on Lung Cancer in Vienna, an Australian study, also in a highly selected set of cases, showed interobserver variation of 15.8% at the 1% threshold and of 18% at the 50% threshold.105  Rehman and colleagues106  examined, with the SP142 protocol, the reproducibility of 5 pathologists' scores on a selected set of cases and showed an intraclass correlation coefficient of 94% agreement among the pathologists for the assessment of PD-L1 in tumor cells, but only 27% agreement in stromal/immune cell PD-L1 expression. The possible lack of consensus on the exact method for reading stromal cells was one of the explanations.

The recently published study of Ratcliffe and colleagues107  examined 493 selected cases stained with 3 protocols (22C3, 28-8, and SP263) of which 200 were read by a second pathologist. The intraobserver overall percentage of agreement between the 3 protocols varied for the 1% threshold between 88.7% and 91.7% and for the 50% threshold between 91.5% and 95.6%. Remarkably, the interobserver overall percentage of agreement between the 3 protocols varied for the 1% threshold between 75.9% and 77% and for the 50% threshold between 94.5% and 97.5%. It is likely that a relevant part of the examined cases will consist of critical samples. If we assume that approximately 150 of the more than 200+ negative cases and more than 50 of the highly positive cases may be looked upon, from a methodologic point of view, as not informative, then the denominator will be smaller and the comparison in samples that really matter will increase: for example, from 10% to 16%. It would be useful to know the actual critical samples of this study and establish the clinical validity. Interestingly, the size of this study sheds light on the interobserver variation, being well within the 9% range at the 50% threshold. However, at the 1% threshold there is room for improvement. To what extend training may improve this is currently unclear.

A French PD-L1 comparison study, presented at the 16th World Conference on Lung Cancer in Vienna, was performed by 7 centers, contributing 6 NSCLC cases distributed over adenocarcinoma and squamous cell carcinoma as well as more than 3 categories of PD-L1 positivity (<5%, 5%–25%, and ≥50%).108  Each center used its own staining platform (3 with Dako AS Linker 48, 2 with Ventana Benchmark Ultra, and 2 with Leica Bond) and all 7 used 5 staining protocols: 28-8, 22C3, SP142, SP263, and E1L3N. Thus, at least 3 of the 5 sections per case per laboratory were stained by an LDT and minimally 35 sections were required per case, leaving 41 tested cases. The LDT was locally established, aiming for concordance with 28-8 or 22C3 (reference images and tonsil tissue). The slides were centrally relabeled after staining and read once (each pathologist read 6 cases, 35 PD-L1–stained sections) after a preliminary discussion of the PD-L1 scoring method. Thus, interobserver variability was not assessed. Weighted κ was used to compare the tests, based on the 1% and 50% thresholds.

Based on an arbitrary threshold of κ = 0.75, the number of acceptable concordance, according to the authors of the 7 laboratories, was 4, 5, 7, 2, and 4 for 28-8, 22C3, SP263, SP142, and E1L3N, respectively.108  For the LDTs performed on an instrument with different antibodies from those of the commercial kit, 13 of 27 comparisons were below the κ threshold. The agreement/discordant rates and details of signal enhancement systems used are not available yet. It is not clear whether the samples used were critical samples or not. Thus, care has to be taken before implementing LDT for PD-L1 testing.

Overall, the comparison of different PD-L1 assays suggests hopeful data, but more research is needed. According to the FDA a high number of samples need to be examined for noninferiority testing.109  For optimal determination of the interobserver variability in daily practice, a consecutive series of cases is mandatory.

EGFR mutation–specific antibodies have been developed, which recognize the protein conformation change due to the mutation, but do not bind to the wild-type EGFR protein.

Different antibodies identify a 15-bp deletion in exon 19 and an L858R point mutation in exon 21. Currently, these are not recommended for predictive testing, as the sensitivity, in particular for other exon 19 deletions, is moderate: 15-bp deletion is better recognized than 18- or 12-bp deletion.110115  However, when limited tissue is available, such as in cytologic samples, application of IHC with these mutation-specific antibodies may be useful.

In Europe, EMA approved the monoclonal anti-EGFR drug necitumumab for patients with squamous NSCLC in advanced stage of disease that expresses the wild-type EGFR protein by IHC.

A BRAF V600E mutation–specific antibody exists, but hardly detects any of the proteins with non-V600E mutations.116,117  In NSCLC a relevant part of the mutations are non-V600E mutations.118121  In small samples IHC may be used to detect V600E mutations.

The use of IHC for the determination of pulmonary carcinoma biomarkers is a well-established and powerful technique (Table 4). Immunohistochemistry is readily available in pathology laboratories, it is relatively easy to perform and assess, can provide clinically meaningful results very quickly, and is relatively inexpensive. The lung oncology community may be somewhat suspicious of IHC biomarkers, since there have been a number of failed trials that have occasionally faltered owing to the quality and nature of the IHC biomarker (ERCC1), or the biomarker was perhaps wrongly blamed (METMAb, EGFR/cetuximab). This should not be used as evidence against the use of IHC biomarkers. The IHC biomarkers discussed in this article, however, highlight the importance of understanding the practice of IHC, and how the particular chemistry used in any assay may influence the test outcome. This can be used to the patient's advantage; with appropriate usage, for example, ALK IHC protocols may or may not require FISH confirmation, depending on the way in which the ALK IHC is conducted. The complex story surrounding PD-L1 IHC highlights the need for understanding also. The currently described literature on PD-L1 IHC has not yet shown, from a methodologic point of view, that different assays are comparable. The route to LDT and commercial test validation is challenging and complex. Handling along proper methodologic lines is needed to ensure patients receive the most accurate and representative test outcomes.

Table 4

Summary

Summary
Summary
1
Dietel
M.
Bubendorf
L.
Dingemans
A-MC.
et al.
Diagnostic procedures for non-small-cell lung cancer (NSCLC): recommendations of the European Expert Group
.
Thorax
.
2016
;
71
(
2
):
177
184
. doi:.
2
Travis
WD.
Brambilla
E.
Nicholson
AG.
et al.
The 2015 World Health Organization Classification of Lung Tumors: impact of genetic, clinical and radiologic advances since the 2004 classification
.
J Thorac Oncol
.
2015
;
10
(
9
):
1243
1260
. doi:.
3
Tsao
MS.
Hirsch
FR.
Yatabe
Y.
eds
.
IASLC Atlas of ALK and ROS1 Testing in Lung Cancer. 2nd ed
.
North Forth Myers, FL
:
Editorial Rx Press;
2016
.
4
Bubendorf
L.
Büttner
R.
Al-Dayel
F.
et al.
Testing for ROS1 in non-small cell lung cancer: a review with recommendations
.
Virchows Arch
.
2016
;
469
(
5
):
489
503
. doi:.
5
Zhou
F.
Moreira
AL.
Lung carcinoma predictive biomarker testing by immunoperoxidase stains in cytology and small biopsy specimens: advantages and limitations
.
Arch Pathol Lab Med
.
2016
;
140
(
12
):
1331
1337
. doi:.
6
Savic
S.
Bode
B.
Diebold
J.
et al.
Detection of ALK-positive non-small-cell lung cancers on cytological specimens: high accuracy of immunocytochemistry with the 5A4 clone
.
J Thorac Oncol
.
2013
;
8
(
8
):
1004
1011
. doi:.
7
Bass
BP.
Engel
KB.
Greytak
SR.
Moore
HM.
A review of preanalytical factors affecting molecular, protein, and morphological analysis of formalin-fixed, paraffin-embedded (FFPE) tissue: how well do you know your FFPE specimen?
Arch Pathol Lab Med
.
2014
;
138
(
11
):
1520
1530
. doi:.
8
Pollard
K.
Lunny
D.
Holgate
CS.
Jackson
P.
Bird
CC.
Fixation, processing, and immunochemical reagent effects on preservation of T-lymphocyte surface membrane antigens in paraffin-embedded tissue
.
J Histochem Cytochem
.
1987
;
35
(
11
):
1329
1338
.
9
Sauter
JL.
Grogg
KL.
Vrana
JA.
Law
ME.
Halvorson
JL.
Henry
MR.
Young investigator challenge: validation and optimization of immunohistochemistry protocols for use on cellient cell block specimens
.
Cancer Cytopathol
.
2016
;
124
(
2
):
89
100
. doi:.
10
Loftin
I.
Miller
R.
Thorne-Nuzzo
P.
et al.
ALK IHC is highly sensitive to fixation parameters
.
J Thorac Oncol
.
2017
;
12
(
S1
):
S265
.
11
Kerr
KM.
Bubendorf
L.
Edelman
MJ.
et al.
Second ESMO consensus conference on lung cancer: pathology and molecular biomarkers for non-small-cell lung cancer
.
Ann Oncol
.
2014
;
25
(
9
):
1681
1690
. doi:.
12
Thunnissen
E.
Kerr
KM.
Herth
FJ.
et al.
The challenge of NSCLC diagnosis and predictive analysis on small samples: practical approach of a working group
.
Lung Cancer
.
2012
;
76
(
1
):
1
18
. doi:.
13
Blind
C.
Koepenik
A.
Pacyna-Gengelbach
M.
et al.
Antigenicity testing by immunohistochemistry after tissue oxidation
.
J Clin Pathol
.
2008
;
61
(
1
):
79
83
. doi:.
14
Schrijver
WAME.
van der Groep
P.
Hoefnagel
LD.
et al.
Influence of decalcification procedures on immunohistochemistry and molecular pathology in breast cancer
.
Mod Pathol
.
2016
;
29
(
12
):
1460
1470
. doi:.
15
Prinsen
CFM.
Klaassen
CHW.
Thunnissen
FBJM.
Microarray as a model for quantitative visualization chemistry
.
Appl Immunohistochem Mol Morphol
.
2003
;
11
(
2
):
168
173
.
16
Rüschoff
J.
Dietel
M.
Baretton
G.
et al.
HER2 diagnostics in gastric cancer-guideline validation and development of standardized immunohistochemical testing
.
Virchows Arch
.
2010
;
457
(
3
):
299
307
. doi:.
17
Rüschoff
J.
Kerr
K.
Grote
H.
et al.
the Reproducibility of immunohistochemical scoring for epidermal growth factor receptor expression in non-small cell lung cancer
.
Arch Pathol Lab Med
.
2013
;
137
(
9
):
1255
1261
.
18
Torlakovic
EE.
Francis
G.
Garratt
J.
et al.
Standardization of negative controls in diagnostic immunohistochemistry: recommendations from the international ad hoc expert panel
.
Appl Immunohistochem Mol Morphol
.
2014
;
22
(
4
):
241
252
. doi:.
19
Torlakovic
EE.
Nielsen
S.
Francis
G.
et al.
Standardization of positive controls in diagnostic immunohistochemistry: recommendations from the International Ad Hoc Expert Committee
.
Appl Immunohistochem Mol Morphol
.
2015
;
23
(
1
):
1
18
. doi:.
20
Fitzgibbons
PL.
Bradley
LA.
Fatheree
LA.
et al.
Principles of analytic validation of immunohistochemical assays: Guideline From the College of American Pathologists Pathology and Laboratory Quality Center
.
Arch Pathol Lab Med
.
2014
;
138
(
11
):
1432
1443
. doi:.
21
Shaw
AT.
Kim
D-W.
Nakagawa
K.
et al.
Crizotinib versus chemotherapy in advanced ALK-positive lung cancer
.
N Engl J Med
.
2013
;
368
(
25
):
2385
2394
. doi:.
22
Solomon
BJ.
Mok
T.
Kim
D-W.
et al.
First-line crizotinib versus chemotherapy in ALK-positive lung cancer
.
N Engl J Med
.
2014
;
371
(
23
):
2167
2177
. doi:.
23
Conklin
CMJ.
Craddock
KJ.
Have
C.
Laskin
J.
Couture
C.
Ionescu
DN.
Immunohistochemistry is a reliable screening tool for identification of ALK rearrangement in non-small-cell lung carcinoma and is antibody dependent
.
J Thorac Oncol
.
2013
;
8
(
1
):
45
51
. doi:.
24
Blackhall
FH.
Peters
S.
Bubendorf
L.
et al.
Prevalence and clinical outcomes for patients with ALK-positive resected stage I to III adenocarcinoma: results from the European Thoracic Oncology Platform Lungscape Project
.
J Clin Oncol
.
2014
;
32
(
25
):
2780
2787
. doi:.
25
Gruber
K.
Kohlhäufl
M.
Friedel
G.
Ott
G.
Kalla
C.
A novel, highly sensitive ALK antibody 1A4 facilitates effective screening for ALK rearrangements in lung adenocarcinomas by standard immunohistochemistry
.
J Thorac Oncol
.
2015
;
10
(
4
):
713
716
. doi:.
26
Wang
Q.
Zhao
L.
Yang
X.
et al.
Antibody 1A4 with routine immunohistochemistry demonstrates high sensitivity for ALK rearrangement screening of Chinese lung adenocarcinoma patients: a single-center large-scale study
.
Lung Cancer
.
2016
;
95
:
39
43
. doi:.
27
Savic
S.
Diebold
J.
Zimmermann
A-K.
et al.
Screening for ALK in non-small cell lung carcinomas: 5A4 and D5F3 antibodies perform equally well, but combined use with FISH is recommended
.
Lung Cancer
.
2015
;
89
(
2
):
104
109
. doi:.
28
Ibrahim
M.
Parry
S.
Wilkinson
D.
et al.
ALK Immunohistochemistry in NSCLC: discordant staining can impact patient treatment regimen
.
J Thorac Oncol
.
2016
;
11
(
12
):
2241
2247
. doi:.
29
VENTANA ALK Scoring Interpretation Guide for Non-Small Cell Lung Carcinoma (NSCLC)
.
March
26,
2017
.
30
Rodig
SJ.
Mino-Kenudson
M.
Dacic
S.
et al.
Unique clinicopathologic features characterize ALK-rearranged lung adenocarcinoma in the western population
.
Clin Cancer Res
.
2009
;
15
(
16
):
5216
5223
. doi:.
31
Yoshida
A.
Tsuta
K.
Watanabe
S.
et al.
Frequent ALK rearrangement and TTF-1/p63 co-expression in lung adenocarcinoma with signet-ring cell component
.
Lung Cancer
.
2011
;
72
(
3
):
309
315
. doi:.
32
Popat
S.
Gonzalez
D.
Min
T.
et al.
ALK translocation is associated with ALK immunoreactivity and extensive signet-ring morphology in primary lung adenocarcinoma
.
Lung Cancer
.
2012
;
75
(
3
):
300
305
. doi:.
33
Murakami
Y.
Mitsudomi
T.
Yatabe
Y.
A screening method for the ALK fusion gene in NSCLC
.
Front Oncol
.
2012
;
2
(
March
):
24
. doi:.
34
Nakamura
H.
Tsuta
K.
Yoshida
A.
et al.
Aberrant anaplastic lymphoma kinase expression in high-grade pulmonary neuroendocrine carcinoma
.
J Clin Pathol
.
2013
;
66
(
8
):
705
707
. doi:.
35
Takeuchi
K.
Choi
YL.
Togashi
Y.
et al.
KIF5B-ALK, a novel fusion oncokinase identified by an immunohistochemistry-based diagnostic system for ALK-positive lung cancer
.
Clin Cancer Res
.
2009
;
15
(
9
):
3143
3149
. doi:.
36
Mino-Kenudson
M.
Chirieac
LR.
Law
K.
et al.
A novel, highly sensitive antibody allows for the routine detection of ALK-rearranged lung adenocarcinomas by standard immunohistochemistry
.
Clin Cancer Res
.
2010
;
16
(
5
):
1561
1571
. doi:.
37
Martinez
P.
Hernández-Losa
J.
Montero
MÁ.
et al.
Fluorescence in situ hybridization and immunohistochemistry as diagnostic methods for ALK positive non-small cell lung cancer patients
.
PLoS One
.
2013
;
8
(
1
):
e52261
. doi:.
38
Minca
EC.
Portier
BP.
Wang
Z.
et al.
ALK status testing in non-small cell lung carcinoma: correlation between ultrasensitive IHC and FISH
.
J Mol Diagn
.
2013
;
15
(
3
):
341
346
. doi:.
39
Ying
J.
Guo
L.
Qiu
T.
et al.
Diagnostic value of a novel fully automated immunochemistry assay for detection of ALK rearrangement in primary lung adenocarcinoma
.
Ann Oncol
.
2013
;
24
(
10
):
2589
2593
. doi:.
40
Wu
Y-C.
Chang
I-C.
Wang
C-L.
et al.
Comparison of IHC, FISH and RT-PCR methods for detection of ALK rearrangements in 312 non-small cell lung cancer patients in Taiwan
.
PLoS One
.
2013
;
8
(
8
):
e70839
. doi:.
41
Tantraworasin
A.
Lertprasertsuke
N.
Kongkarnka
S.
Euathrongchit
J.
Wannasopha
Y.
Saeteng
S.
Retrospective study of ALK rearrangement and clinicopathological implications in completely resected non- small cell lung cancer patients in Northern Thailand: role of screening with D5F3 antibodies
.
Asian Pac J Cancer Prev
.
2014
;
15
(
7
):
3057
3063
.
42
Wang
J.
Cai
Y.
Dong
Y.
et al.
Clinical characteristics and outcomes of patients with primary lung adenocarcinoma harboring ALK rearrangements detected by FISH, IHC, and RT-PCR
.
PLoS One
.
2014
;
9
(
7
):
e101551
. doi:.
43
Zhou
J.
Zhao
J.
Sun
K.
et al.
Accurate and economical detection of ALK positive lung adenocarcinoma with semiquantitative immunohistochemical screening
.
PLoS One
.
2014
;
9
(
3
):
e92828
. doi:.
44
Shan
L.
Lian
F.
Guo
L.
Yang
X.
Ying
J.
Lin
D.
Combination of conventional immunohistochemistry and qRT-PCR to detect ALK rearrangement
.
Diagn Pathol
.
2014
;
9
(
1
):
3
. doi:.
45
Alì
G.
Proietti
A.
Pelliccioni
S.
et al.
ALK rearrangement in a large series of consecutive non–small cell lung cancers: comparison between a new immunohistochemical approach and fluorescence in situ hybridization for the screening of patients eligible for crizotinib treatment
.
Arch Pathol Lab Med
.
2014
;
138
(
11
):
1449
1458
. doi:.
46
Ilie
MI.
Bence
C.
Hofman
V.
et al.
Discrepancies between FISH and immunohistochemistry for assessment of the ALK status are associated with ALK “borderline”-positive rearrangements or a high copy number: a potential major issue for anti-ALK therapeutic strategies
.
Ann Oncol
.
2015
;
26
(
1
):
238
244
. doi:.
47
Wynes
MW.
Sholl
LM.
Dietel
M.
et al.
An international interpretation study using the ALK IHC antibody D5F3 and a sensitive detection kit demonstrates high concordance between ALK IHC and ALK FISH and between evaluators
.
J Thorac Oncol
.
2014
;
9
(
5
):
631
638
. doi:.
48
Paik
JH.
Choe
G.
Kim
H.
et al.
Screening of anaplastic lymphoma kinase rearrangement by immunohistochemistry in non-small cell lung cancer: correlation with fluorescence in situ hybridization
.
J Thorac Oncol
.
2011
;
6
(
3
):
466
472
. doi:.
49
Jokoji
R.
Yamasaki
T.
Minami
S.
et al.
Combination of morphological feature analysis and immunohistochemistry is useful for screening of EML4-ALK-positive lung adenocarcinoma
.
J Clin Pathol
.
2010
;
63
(
12
):
1066
1070
. doi:.
50
Kim
H.
Yoo
S-B.
Choe
J-Y.
et al.
Detection of ALK gene rearrangement in non-small cell lung cancer: a comparison of fluorescence in situ hybridization and chromogenic in situ hybridization with correlation of ALK protein expression
.
J Thorac Oncol
.
2011
;
6
(
8
):
1359
1366
. doi:.
51
McLeer-Florin
A.
Moro-Sibilot
D.
Melis
A.
et al.
Dual IHC and FISH testing for ALK gene rearrangement in lung adenocarcinomas in a routine practice: a French study
.
J Thorac Oncol
.
2012
;
7
(
2
):
348
354
. doi:.
52
Lopes
L.
Bacchi
C.
Anaplastic lymphoma kinase gene rearrangement in non-small-cell lung cancer in a Brazilian population
.
Clinics
.
2012
;
67
(
7
):
845
847
. doi:.
53
Park
HS.
Lee
JK.
Kim
D-W.
et al.
Immunohistochemical screening for anaplastic lymphoma kinase (ALK) rearrangement in advanced non-small cell lung cancer patients
.
Lung Cancer
.
2012
;
77
(
2
):
288
292
. doi:.
54
Sholl
LM.
Weremowicz
S.
Gray
SW.
et al.
Combined use of ALK immunohistochemistry and FISH for optimal detection of ALK-rearranged lung adenocarcinomas
.
J Thorac Oncol
.
2013
;
8
(
3
):
322
328
. doi:.
55
To
K-F.
Tong
JH.
Yeung
KS.
et al.
Detection of ALK rearrangement by immunohistochemistry in lung adenocarcinoma and the identification of a novel EML4-ALK variant
.
J Thorac Oncol
.
2013
;
8
:
883
891
. doi:.
56
Cabillic
F.
Gros
A.
Dugay
F.
et al.
Parallel FISH and immunohistochemical studies of ALK status in 3244 non-small-cell lung cancers reveal major discordances
.
J Thorac Oncol
.
2014
;
9
(
3
):
295
306
. doi:.
57
Jiang
L.
Yang
H.
He
P.
et al.
Improving selection criteria for ALK inhibitor therapy in non-small cell lung cancer: a pooled-data analysis on diagnostic operating characteristics of immunohistochemistry
.
Am J Surg Pathol
.
2016
;
40
(
5
):
697
703
. doi:.
58
Conde
E.
Suárez-Gauthier
A.
Benito
A.
et al.
Accurate identification of ALK positive lung carcinoma patients: novel FDA-cleared automated fluorescence in situ hybridization scanning system and ultrasensitive immunohistochemistry
.
PLoS One
.
2014
;
9
(
9
):
e107200
. doi:.
59
Marchetti
A.
Di Lorito
A.
Pace
MV.
et al.
ALK protein analysis by IHC staining after recent regulatory changes: a comparison of two widely used approaches, revision of the literature, and a new testing algorithm
.
J Thorac Oncol
.
2016
;
11
(
4
):
487
495
. doi:.
60
Zwaenepoel
K.
Van Dongen
A.
Lambin
S.
Weyn
C.
Pauwels
P.
Detection of ALK expression in non-small-cell lung cancer with ALK gene rearrangements–comparison of multiple immunohistochemical methods
.
Histopathology
.
2014
;
65
(
4
):
539
548
. doi:.
61
Lantuejoul
S.
Rouquette
I.
Blons
H.
et al.
French multicentric validation of ALK rearrangement diagnostic in 547 lung adenocarcinomas
.
Eur Respir J
.
2015
;
46
(
1
):
207
218
. doi:.
62
Jurmeister
P.
Lenze
D.
Berg
E.
et al.
Parallel screening for ALK, MET and ROS1 alterations in non-small cell lung cancer with implications for daily routine testing
.
Lung Cancer
.
2015
;
87
(
2
):
122
129
. doi:.
63
Gao
X.
Sholl
LM.
Nishino
M.
Heng
JC.
Jänne
PA.
Oxnard
GR.
Clinical implications of variant ALK FISH rearrangement patterns
.
J Thorac Oncol
.
2015
;
10
(
11
):
1648
1652
. doi:.
64
Pekar-Zlotin
M.
Hirsch
FR.
Soussan-Gutman
L.
et al.
Fluorescence in situ hybridization, immunohistochemistry, and next-generation sequencing for detection of EML4-ALK rearrangement in lung cancer
.
Oncologist
.
2015
;
20
(
3
):
316
322
. doi:.
65
von Laffert
M.
Stenzinger
A.
Hummel
M.
et al.
ALK-FISH borderline cases in non-small cell lung cancer: implications for diagnostics and clinical decision making
.
Lung Cancer
.
2015
;
90
:
465
471
. doi:.
66
Selinger
CI.
Rogers
T-M.
Russell
PA.
et al.
Testing for ALK rearrangement in lung adenocarcinoma: a multicenter comparison of immunohistochemistry and fluorescent in situ hybridization
.
Mod Pathol
.
2013
;
26
(
12
):
1545
1553
. doi:.
67
Kim
H.
Xu
X.
Yoo
S-B.
et al.
Discordance between anaplastic lymphoma kinase status in primary non-small-cell lung cancers and their corresponding metastases
.
Histopathology
.
2013
;
62
(
2
):
305
314
. doi:.
68
Salido
M.
Pijuan
L.
Martínez-Avilés
L.
et al.
Increased ALK gene copy number and amplification are frequent in non-small cell lung cancer
.
J Thorac Oncol
.
2011
;
6
(
1
):
21
27
. doi:.
69
Wiesner
T.
Lee
W.
Obenauf
AC.
et al.
Alternative transcription initiation leads to expression of a novel ALK isoform in cancer
.
Nature
.
2015
;
526
(
7573
):
453
457
. doi:.
70
Rosoux
A.
Pauwels
P.
Duplaquet
F.
et al.
Effectiveness of crizotinib in a patient with ALK IHC-positive/FISH-negative metastatic lung adenocarcinoma
.
Lung Cancer
.
2016
;
98
:
118
121
. doi:.
71
Bergethon
K.
Shaw
AT.
Ou
S-HI.
et al.
ROS1 rearrangements define a unique molecular class of lung cancers
.
J Clin Oncol
.
2012
;
30
(
8
):
863
870
. doi:.
72
Li
S.
Choi
Y-L.
Gong
Z.
et al.
Comprehensive characterization of oncogenic drivers in Asian lung adenocarcinoma
.
J Thorac Oncol
.
2016
;
11
(
12
):
2129
2140
. doi:.
73
Govindan
R.
Ding
L.
Griffith
M.
et al.
Genomic landscape of non-small cell lung cancer in smokers and never-smokers
.
Cell
.
2013
;
150
(
6
):
1121
1134
. doi:.
74
Rimkunas
VM.
Crosby
KE.
Li
D.
et al.
Analysis of receptor tyrosine kinase ROS1-positive tumors in non-small cell lung cancer: identification of a FIG-ROS1 fusion
.
Clin Cancer Res
.
2012
;
18
(
16
):
4449
4457
. doi:.
75
Seo
J-S.
Ju
YS.
Lee
W-C.
et al.
The transcriptional landscape and mutational profile of lung adenocarcinoma
.
Genome Res
.
2012
;
22
(
11
):
2109
2119
.
76
Takeuchi
K.
Soda
M.
Togashi
Y.
et al.
RET, ROS1 and ALK fusions in lung cancer
.
Nat Med
.
2012
;
18
(
3
):
378
381
. doi:.
77
Yoshida
A.
Kohno
T.
Tsuta
K.
et al.
ROS1-rearranged lung cancer: a clinicopathologic and molecular study of 15 surgical cases
.
Am J Surg Pathol
.
2013
;
37
(
4
):
554
562
.
78
FDA expands use of Xalkori to treat rare form of advanced non-small cell lung cancer
.
March
26,
2017
.
79
Boyle
TA.
Masago
K.
Ellison
KE.
Yatabe
Y.
Hirsch
FR.
ROS1 immunohistochemistry among major genotypes of non-small-cell lung cancer
.
Clin Lung Cancer
.
2015
;
16
(
2
):
106
111
. doi:.
80
Yoshida
A.
Tsuta
K.
Wakai
S.
et al.
Immunohistochemical detection of ROS1 is useful for identifying ROS1 rearrangements in lung cancers
.
Mod Pathol
.
2014
;
27
(
5
):
711
720
. doi:.
81
Sholl
LM.
Sun
H.
Butaney
M.
et al.
ROS1 immunohistochemistry for detection of lung adenocarcinomas
.
Am J Surg Pathol
.
2013
;
37
(
9
):
1441
1449
.
82
Mescam-Mancini
L.
Lantuéjoul
S.
Moro-Sibilot
D.
et al.
On the relevance of a testing algorithm for the detection of ROS1-rearranged lung adenocarcinomas
.
Lung Cancer
.
2014
;
83
(
2
):
168
173
. doi:.
83
Sholl
LM.
Aisner
DL.
Allen
TC.
et al.
Programmed death ligand 1 immunohistochemistry—a new challenge for pathologists: a perspective from members of the Pulmonary Pathology Society
.
Arch Pathol Lab Med
.
2016
;
140
(
4
):
341
344
. doi:.
84
Weber
JS.
Kudchadkar
RR.
Yu
B.
et al.
Safety, efficacy, and biomarkers of nivolumab with vaccine in ipilimumab-refractory or -naive melanoma
.
J Clin Oncol
.
2013
;
31
(
34
):
4311
4318
. doi:.
85
Tumeh
PC.
Harview
CL.
Yearley
JH.
et al.
PD-1 blockade induces responses by inhibiting adaptive immune resistance
.
Nature
.
2014
;
515
(
7528
):
568
571
. doi:.
86
Planchard
D.
Yokoi
T.
McCleod
MJ.
et al.
A phase III study of durvalumab (MEDI4736) with or without tremelimumab for previously treated patients with advanced NSCLC: rationale and protocol design of the ARCTIC study
.
Clin Lung Cancer
.
2016
;
17
(
3
):
232
236.e1
. doi:.
87
Kaufman
HL.
Russell
J.
Hamid
O.
et al.
Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial
.
Lancet Oncol
.
2016
;
17
(
10
):
1374
1385
. doi:.
88
Gaule
P.
Smithy
JW.
Toki
M.
et al.
A quantitative comparison of antibodies to programmed cell death 1 ligand 1
.
JAMA Oncol
.
2017
;
3
(
2
):
256
. doi:.
89
McLaughlin
J.
Han
G.
Schalper
KA.
et al.
Quantitative assessment of the heterogeneity of PD-L1 expression in non-small-cell lung cancer
.
JAMA Oncol
.
2016
;
2
(
1
):
46
54
. doi:.
90
Cree
IA.
Booton
R.
Cane
P.
et al.
PD-L1 testing for lung cancer in the UK: recognizing the challenges for implementation
.
Histopathology
.
2016
;
69
(
2
):
177
186
. doi:.
91
Midha
A.
PD-L1 expression in advanced NSCLC: primary lesions versus metastatic sites and impact of sample age. Poster presented at: ASCO Chicago, IL; June 5, 2016
.
J Clin Oncol
.
2016
;
34
(
suppl
):
abstract 3025
.
92
Yu
H.
Boyle
TA.
Zhou
C.
Rimm
DL.
Hirsch
FR.
PD-L1 expression in lung cancer
.
J Thorac Oncol
.
2016
;
11
(
7
):
964
975
. doi:.
93
Velcheti
V.
Schalper
KA.
Carvajal
DE.
et al.
Programmed death ligand-1 expression in non-small cell lung cancer
.
Lab Invest
.
2013
;
94
(
1
):
107
116
. doi:.
94
Patel
SP.
Kurzrock
R.
PD-L1 expression as a predictive biomarker in cancer immunotherapy
.
Mol Cancer Ther
.
2015
;
14
(
4
):
847
856
. doi:.
95
Sznol
M.
Chen
L.
Antagonist antibodies to PD-1 and B7-H1 (PD-L1) in the treatment of advanced human cancer
.
Clin Cancer Res
.
2013
;
19
(
5
):
1021
1034
. doi:.
96
Kitazono
S.
Fujiwara
Y.
Tsuta
K.
et al.
Reliability of small biopsy samples compared with resected specimens for the determination of programmed death-ligand 1 expression in non–small-cell lung cancer
.
Clin Lung Cancer
.
2015
;
16
(
5
):
385
390
. doi:.
97
Ilie
M.
Long-Mira
E.
Bence
C.
et al.
Comparative study of the PD-L1 status between surgically resected specimens and matched biopsies of NSCLC patients reveal major discordances: a potential issue for anti-PD-L1 therapeutic strategies
.
Ann Oncol
.
2016
;
27
(
1
):
147
153
. doi:.
98
Borghaei
H.
Paz-Ares
L.
Horn
L.
et al.
Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer
.
N Engl J Med
.
2015
;
373
(
17
):
1627
1639
. doi:.
99
Rittmeyer
A.
Barlesi
F.
Waterkamp
D.
et al.
Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial
.
Lancet
.
2017
;
389
(
10066
):
255
265
. doi:.
100
Fischer
AH.
Schwartz
MR.
Moriarty
AT.
et al.
Immunohistochemistry practices of cytopathology laboratories: a survey of participants in the College of American Pathologists Nongynecologic Cytopathology Education Program
.
Arch Pathol Lab Med
.
2014
;
138
(
9
):
1167
1172
. doi:.
101
Skov
BG.
Skov
T.
Paired comparison of PD-L1 expression on cytologic and histologic specimens from malignancies in the lung assessed with PD-L1 IHC 28-8pharmDx and PD-L1 IHC 22C3pharmDx
[published online ahead of print May 25, 2017]
.
Appl Immunohistochem Mol Morphol
. doi:.
102
van Krieken
JH.
Normanno
N.
Blackhall
F.
et al.
Guideline on the requirements of external quality assessment programs in molecular pathology
.
Virchows Arch
.
2013
;
462
(
1
):
27
37
. doi:.
103
Hirsch
FR.
McElhinny
A.
Stanforth
D.
et al.
PD-L1 Immunohistochemistry assays for lung cancer: results from phase 1 of the Blueprint PD-L1 IHC Assay Comparison Project
.
J Thorac Oncol
.
2017
;
12
(
2
):
208
222
. doi:.
104
Scheel
AH.
Dietel
M.
Heukamp
LC.
et al.
Harmonized PD-L1 immunohistochemistry for pulmonary squamous-cell and adenocarcinomas
.
Mod Pathol
.
2016
;
29
(
10
):
1
8
. doi:.
105
Cooper
WA.
Russell
PA.
Huot-Marhand
P.
et al.
Intra- and interobserver reproducibility study of PD-L1 biomarker in NSCLC: The Dream Study
.
J Thorac Oncol
.
2017
;
12
(
s1
):
424
.
106
Rehman
JA.
Han
G.
Carvajal-Hausdorf
DE.
et al.
Quantitative and pathologist-read comparison of the heterogeneity of programmed death-ligand 1 (PD-L1) expression in non-small cell lung cancer
.
Mod Pathol
.
2017
;
30
(
3
):
340
349
. doi:.
107
Ratcliffe
MJ.
Sharpe
A.
Midha
A.
et al.
Agreement between programmed cell death ligand-1 diagnostic assays across multiple protein expression cut-offs in non-small cell lung cancer
[published online ahead of print
January
10,
2017]
. Clin Cancer Res. doi:.
108
Adam
J.
Rouquette
I.
Damotte
D.
et al.
Multicentric French Harmonization study for PD-L1 testing in NSCLC
.
J Thorac Oncol
.
2017
;
12
(
S1
):
S4
S5
.
109
Li
M.
Statistical methods for clinical validation of follow-on companion diagnostic devices via an external concordance study
.
Stat Biopharm Res
.
2016
;
8
(
3
):
355
363
. doi:.
110
Brevet
M.
Arcila
M.
Ladanyi
M.
Assessment of EGFR mutation status in lung adenocarcinoma by immunohistochemistry using antibodies specific to the two major forms of mutant EGFR
.
J Mol Diagn
.
2010
;
12
(
2
):
169
176
. doi:.
111
Yu
J.
Kane
S.
Wu
J.
et al.
Mutation-specific antibodies for the detection of EGFR mutations in non-small-cell lung cancer
.
Clin Cancer Res
.
2009
;
15
(
9
):
3023
3028
. doi:.
112
Simonetti
S.
Molina
MA.
Queralt
C.
et al.
Detection of EGFR mutations with mutation-specific antibodies in stage IV non-small-cell lung cancer
.
J Transl Med
.
2010
;
8
:
135
. doi:.
113
Kawahara
A.
Yamamoto
C.
Nakashima
K.
et al.
Molecular diagnosis of activating EGFR mutations in non-small cell lung cancer using mutation-specific antibodies for immunohistochemical analysis
.
Clin Cancer Res
.
2010
;
16
(
12
):
3163
3170
. doi:.
114
Kitamura
A.
Hosoda
W.
Sasaki
E.
Mitsudomi
T.
Yatabe
Y.
Immunohistochemical detection of EGFR mutation using mutation-specific antibodies in lung cancer
.
Clin Cancer Res
.
2010
;
16
(
13
):
3349
3355
. doi:.
115
Jiang
G.
Fan
C.
Zhang
X.
et al.
Ascertaining an appropriate diagnostic algorithm using EGFR mutation-specific antibodies to detect EGFR status in non-small-cell lung cancer
.
PLoS One
.
2013
;
8
(
3
):
e59183
. doi:.
116
Sasaki
H.
Shimizu
S.
Tani
Y.
et al.
Usefulness of immunohistochemistry for the detection of the BRAF V600E mutation in Japanese lung adenocarcinoma
.
Lung Cancer
.
2013
;
82
(
1
):
51
54
. doi:.
117
Ilie
M.
Long
E.
Hofman
V.
et al.
Diagnostic value of immunohistochemistry for the detection of the BRAFV600E mutation in primary lung adenocarcinoma Caucasian patients
.
Ann Oncol
.
2013
;
24
(
3
):
742
748
. doi:.
118
Cardarella
S.
Ogino
A.
Nishino
M.
et al.
Clinical, pathologic, and biologic features associated with BRAF mutations in non-small cell lung cancer
.
Clin Cancer Res
.
2013
;
19
(
16
):
4532
4540
. doi:.
119
Davies
H.
Bignell
GR.
Cox
C.
et al.
Mutations of the BRAF gene in human cancer
.
Nature
.
2002
;
417
(
6892
):
949
954
. doi:.
120
Marchetti
A.
Felicioni
L.
Malatesta
S.
et al.
Clinical features and outcome of patients with non-small-cell lung cancer harboring BRAF mutations
.
J Clin Oncol
.
2011
;
29
(
26
):
3574
3579
. doi:.
121
Paik
PK.
Arcila
ME.
Fara
M.
et al.
Clinical characteristics of patients with lung adenocarcinomas harboring BRAF mutations
.
J Clin Oncol
.
2011
;
29
(
15
):
2046
2051
. doi:.
122
Rogers
T-M.
Russell
PA.
Wright
G.
et al.
Comparison of methods in the detection of ALK and ROS1 rearrangements in lung cancer
.
J Thorac Oncol
.
2015
;
10
(
4
):
611
618
. doi:.
123
Shan
L.
Lian
F.
Guo
L.
et al.
Detection of ROS1 gene rearrangement in lung adenocarcinoma: comparison of IHC, FISH and real-time RT-PCR
.
PLoS One
.
2015
;
10
(
3
):
e0120422
. doi:.
124
Wu
J.
Lin
Y.
He
X.
et al.
Comparison of detection methods and follow-up study on the tyrosine kinase inhibitors therapy in non-small cell lung cancer patients with ROS1 fusion rearrangement
.
BMC Cancer
.
2016
;
16
(
1
):
599
. doi:.
125
Cao
B.
Wei
P.
Liu
Z.
et al.
Detection of lung adenocarcinoma with ROS1 rearrangement by IHC, FISH, and RT-PCR and analysis of its clinicopathologic features
.
Onco Targets Ther
.
2015
;
9
:
131
. doi:.
126
Selinger
CI.
Li
BT.
Pavlakis
N.
et al.
Screening for ROS1 gene rearrangements in non-small-cell lung cancers using immunohistochemistry with FISH confirmation is an effective method to identify this rare target
.
Histopathology
.
2017
;
70
(
3
):
402
411
. doi:.
127
Kao
H-L.
Yeh
Y-C.
Lin
C-H.
et al.
Diagnostic algorithm for detection of targetable driver mutations in lung adenocarcinomas: comprehensive analyses of 205 cases with immunohistochemistry, real-time PCR and fluorescence in situ hybridization methods
.
Lung Cancer
.
2016
;
101
:
40
47
. doi:.
128
Wiesweg
M.
Eberhardt
WEEE.
Reis
H.
et al.
High prevalence of concomitant oncogene mutations in prospectively identified patients with ROS1-positive metastatic lung cancer
.
J Thorac Oncol
.
2017
;
12
(
1
):
54
64
. doi:.
129
Scheel
AH.
Dietel
M.
Heukamp
LC.
et al.
Predictive PD-L1 immunohistochemistry for non-small cell lung cancer : Current state of the art and experiences of the first German harmonization study
.
Pathologe
.
2016
;
37
(
6
):
557
567
.
*

 References 24, 27, 38, 39, 41–47, 51, 53–55, 58–66.

Author notes

Dr Cooper receives honoraria from Pfizer Oncology for lectures and advisory board membership. The other authors have no relevant financial interest in the products or companies described in this article.