A narrative review of predictive immuno-histo/cyto chemistry in lung cancer: focus on gene fusions and PD-L1 expression
Review Article

A narrative review of predictive immuno-histo/cyto chemistry in lung cancer: focus on gene fusions and PD-L1 expression

Pasquale Pisapia, Gennaro Acanfora, Caterina De Luca, Antonino Iaccarino, Giancarlo Troncone, Elena Vigliar

Department of Public Health, University of Naples Federico II, Naples, Italy

Contributions: (I) Conception and design: All authors; (II) Administrative support: All authors; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Giancarlo Troncone. Department of Public Health, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy. Email: giancarlo.troncone@unina.it.

Background and Objective: Personalized medicine has significant modified the way advanced stage non-small cell lung cancer (NSCLC) patients have been managed. In fact, the development of different target drugs has significantly improved the clinical outcomes of these patients. In this scenario, gene rearrangements play a crucial role. Gene fusions, including those involving anaplastic lymphoma receptor tyrosine kinase (ALK), ROS proto-oncogene 1, receptor tyrosine kinase (ROS1), rearranged during transfection (RET) and neurotrophic receptor tyrosine kinase (NTRK) genes, occur very rarely in advanced stage NSCLC patients. Remarkably, these genomic alterations represent an important target for treatment decision algorithm in these patients. In addition, programmed death-ligand 1 (PD-L1) protein expression evaluation is crucial for immune-checkpoint inhibitors (ICIs) administration.

Methods: We reviewed the recent literature on PubMed focusing the attention on the role of IHC/ICC in advanced stage NSCLC patients harboring gene fusions and for the evaluation of PD-L1 expression.

Key Content and Findings: Thus, immunohistochemistry/immunocytochemistry (IHC/ICC), beyond the traditional role in the assessment of the immunophenotype of neoplastic cells to refine uncertain morphological diagnosis, plays a pivotal role in advanced stage NSCLC patients in order to administrate the best treatment option and avoid to leave any patient behind. IHC/ICC is a cost-effective, easy to use and rapid technique that can be adopted even on very scant neoplastic samples. Conclusions: Here, we focalize the attention on the adoption of IHC/ICC approach for gene fusions detection and PD-L1 protein expression evaluation in advanced stage NSCLC patients.

Keywords: Immunohistochemistry (IHC); immunocytochemistry (ICC); non-small cell lung cancer (NSCLC); molecular pathology; molecular oncology


Received: 31 January 2022; Accepted: 16 June 2022; Published: 30 June 2022.

doi: 10.21037/jxym-22-7


Introduction

Lung cancer is still the leading cause of cancer related deaths worldwide (1). Nevertheless, significant steps forwards have been made during the last decades in the field of cancer treatment for these patients. In particular, the identification of several genomic alterations, as point mutations, insertions/deletions (indels), abnormal splicing events and gene fusions, that are or may be potential target for personalized treatments has significant modified the management of advanced stage non-small cell lung cancer (NSCLC) patients (2-4). To this end, international societies have established a panel of so-called “must test genes” [including epidermal growth factor receptor (EGFR) and V-raf murine sarcoma viral oncogene homolog B (BRAF) gene mutations and anaplastic lymphoma receptor tyrosine kinase (ALK) and ROS proto-oncogene 1, receptor tyrosine kinase (ROS1) gene rearrangements] that represents the minimum standard of care panel of genes that must be necessarily tested in advanced stage NSCLC patients (5-8). This rapid evolving field has significant modified the role of pathologists in the diagnostic algorithm of NSCLC patients. In fact, it is crucial the correct management of cancer tissue specimens for both morph-molecular analysis, in order to avoid to leave any patient behind (9). It should be borne in mind that several molecular platforms are being currently available for predictive purposes in molecular predictive pathology laboratories. Among these, immunohistochemistry/immunocytochemistry (IHC/ICC), beyond the classical role in the assessment of the immunophenotype of neoplastic cells, by evaluating the protein expressed by cells, have acquired a crucial predictive role. As a general role, IHC/ICC has been adopted to make classification of advanced stage NSCLC patients more accurate, in a time-efficient, easy and cost-limited way (10,11). In addition, IHC/ICC play a pivotal role in the identification of aberrant proteins production as a consequence of gene fusion events (12-15). These latter can determine tumor development and progression either with a constitutive kinase activation or an altered transcription process of the involved genes (16). In addition, IHC/ICC is currently the only approved approach for the evaluation of the expression of programmed death-ligand 1 (PD-L1) for immune-checkpoint inhibitors (ICIs) administration (17-20). IHC/ICC can be adopted even on very scant neoplastic samples (12). In addition, this morphological approach has the ability to identify fusion events even in cases with an unknown gene partner. However, IHC/ICC suffers from inter-observer variability and may require orthogonal validation of positive results (13).

Here we reviewed the role of IHC/ICC in advanced stage NSCLC patients harboring gene fusions and for the evaluation of PD-L1 expression. We present the following article in accordance with the Narrative Review reporting checklist (available at https://jxym.amegroups.com/article/view/10.21037/jxym-22-7/rc).


Methods

We reviewed the recent literature on PubMed focusing the attention on the role of IHC/ICC in advanced stage NSCLC patients harboring gene fusions and for the evaluation of PD-L1 expression.

ALK gene

As early as 2007, Soda et al. described the echinoderm microtubule-associated protein-like 4 (EML4)/ALK gene fusion in NSCLC patients (21). Despite a number of different partners have been identified, in all cases the chimeric protein shows the presence of the carboxy-terminal portion with kinase activity of ALK protein (14,21-24). Overall, ALK gene fusions are reported in about 3-5% of advanced stage NSCLC patients, in particular young, light or never-smokers, featuring an adenocarcinoma with extracellular mucin, cribriform pattern and signet ring cell histology (21,25). Advanced stage NSCLC patients harboring ALK rearrangement benefit from ALK tyrosine kinase inhibitors (TKIs) administration (26). Currently, different antibodies are being commercially available for IHC/ICC evaluation of ALK fusion protein (27). Among these, 5A4 clone (Abcam, Cambridge, UK) and D5F3 (Ventana, Tucson, AZ, USA) are the most commonly adopted in molecular predictive pathology laboratories (14). Clone 5A4 staining is evaluated by a 4 scores system (0, 1+, 2+, and 3+) (28). Overall, despite it has been demonstrated a full concordance with fluorescent in situ hybridization (FISH) for scores of 0 and 3+, less reproducibility has been reported for scores 1+ and 2+; thus a FISH confirmatory analysis is required in these cases (29,30). Conversely, D5F3 clone has demonstrated a higher sensitivity and specificity with respect to 5A4 clone (31). D5F3 clone with the Ventana automated immunoassay (Ventana ALK D5F3 CDx Assay, Ventana Medical Systems, Tucson, AZ) has obtained the Food and Drug Administration (FDA) approval as a companion diagnostic assay for ALK TKIs administration (12). Overall, through a tyramide-based amplification step in addition to the polymeric phase (OptiView; Ventana Medical Systems) it is possible the selectively intensification of the specific immunosignal while contemporary reducing the background noise (31,32). This process determines the generation of a positive, strong, granular cytoplasmic staining, or negative result (31). For all these reasons, D5F3 clone was adopted to select advanced stage NSCLC patients in the phase 3 trial comparing alectinib to crizotinib (33). Basing on these results, recent recommendations indicate that a strong positive staining with D5F3 clone is enough to start a treatment with ALK TKIs (34). However, it should be borne in mind that, due to the rarity of ALK rearrangements in advanced staged NSCLC patients, it must be mandatory to adopt ALK positive and negative controls samples (27). Beyond histological specimens, it has been widely demonstrated the suitability of different cytological preparation for ALK ICC analysis (35-41).

ROS1 gene

ROS1 gene fusions were reported in advanced stage NSCLC patients for the first time in 2007 by Rikova et al. (42) Similar to ALK gene fusions, ROS1 rearranged cases occur very rarely (about 1–2%), in particular in young, never-smoker women, with adenocarcinoma histology characterized by extracellular mucin, cribriform pattern, and signet ring cells (22,43). In addition, ROS1 rearranged tumors show a hepatoid cytology, with abundant eosinophilic cytoplasm, round and relatively monomorphic nuclei, and prominent nucleoli (44). As well as for ALK fusions, ROS1 fused advanced stage NSCLC patients showed a significant improvement in clinical outcome after treatment with TKIs (45). IHC/ICC approach may represent a valid option to detect ROS1 fusion events (12-14). A limited number of ROS1 specific antibodies have been developed and adopted into the clinical diagnostic routine practice (46-48). Among these, the most common employed in molecular predictive pathology laboratories is represented by D4D6 clone (Cell Signaling Technology, Leiden, The Netherlands), due to a high sensitivity and specificity (49-51). However, D4D6 clone has not obtained FDA approval as a companion diagnostic due to challenging in interpretation of results. In fact, the major issue of IHC/ICC approach is related to the possibility of “false positive” results. This phenomenon is due to the reactivity of non-neoplastic cells, such as hyperplastic type 2 pneumocytes, alveolar epithelial and basal cells, bronchial epithelial and metaplastic bronchiolar cells, and peribronchial glands (49). As a general role, only tumors with 2+ or 3+ staining signal in more than half neoplastic cells should be scored as ROS1 positive (52,53). Another ROS1 specific clone, namely SP384 (Ventana Medical Systems, Oro Valley, AZ, USA), showed a very high sensitivity and specificity (54). However, it is strongly recommended to confirm all ROS1 IHC/ICC positive cases with FISH, polymerase chain reaction (PCR) based approaches or next generation sequencing (NGS) approaches (12,53).

RET gene

The first report of kinesin family member 5B (KIF5B)/rearranged during transfection (RET) gene fusion in advanced stage NSCLC dates back to 2012 (55). Similar to ROS1 gene fusions, RET gene rearrangements account very rarely (1–2%) in advanced stage NSCLC patients (55-58). Overall, RET gene fusions account more frequently in young, female, non-smoker adenocarcinoma patients (56,58-63). In addition, it seems to be associated with poorly differentiation and solid pattern of growth (64). Despite the rarity, the increasing attention on RET fusions is associated with the recent approval of selpercatinib and pralsetinib for the treatment of advanced stage NSCLC patients (65-71). Regarding RET IHC, a global poor concordance with results obtained by PCR-based and FISH approaches is reported (72,73). However, among the different clones, the most commonly adopted is EPR2871 antibody. Yang et al. demonstrated that the sensitivity of IHC is related to the fusion partner, and in particular the highest sensitivity has been registered for KIF5B (100%), followed by CCDC6 (88.9%) and NCOA4 (50%). Interestingly a specificity of 82% was reached (74).

Neurotrophic receptor tyrosine kinase (NTRK) gene

NTRK1 and NTRK2 gene fusions were identified for the first time in advanced stage NSCLC patients by Vaishnavi et al. in 2013 and by Stransky et al. in 2014, respectively (75,76). Overall, gene fusions involving NTRK1-2-3 are very rarely (<1%) reported in advanced stage NSCLC patients (75,77,78). As for other gene fusions, these genomic alterations seem to occur more frequently in younger, non-smoker adenocarcinoma patients (79). The necessity to detect these very uncommon gene fusion events is related to the approval of larotrectinib and entrectinib in patients harboring NTRK gene fusions, regardless the age and the histotype (80-82). Different immunostaining approaches have been reported in literature (83,84). Among these, the pan-Trk antibody EPR17341 (Abcam, MA, USA) showed the highest performance. This clone is able to detect a homologous region of Trk-A, Trk-B, and Trk-C proteins near the C-terminus (84-86). In particular Hechtman et al. reported a sensitivity of 95.2% and a specificity of 100.0% among different previously genotyped cancer types. Noteworthy, However, the sensitivity differs in Trk-A (96%), Trk-B (100%) and Trk-C (79%) fused proteins. Overall, regarding NSCLC, the EPR17341 clone showed a sensitivity of 87.5% and a specificity of 100% (87). It should be borne in mind that staining intensity may be variable. Thus, it has been proposed that neoplasms with at least 1% of positive neoplastic cells should be considered as positive (88). An important point is represented by the localization (nuclear, peri-nuclear, cytoplasmic, cell membrane) of the staining pattern, that is correlated with the specific fusion partner (89). A crucial issue in NTRK immunostaining is associated with the constitutive expression of Trk proteins in some normal human adult tissues, including testis, colonic ganglia, and nervous tissue (88). However, in lung cancer patients, the European Society for Medical Oncology (ESMO) guideline suggested to adopt IHC/ICC as a screening tool when NGS is not available. Nevertheless, it is recommended to further confirm positive results before treatment administration (83).

PD-L1

ICIs have dramatically changed the management of advanced stage NSCLC patients (90-92). Notably, IHC/ICC approach is the only approved for the evaluation of the expression level of PD-L1 in order to administrate ICIs (93). In particular, both histological (IHC) and cytological (ICC) samples have been demonstrated to be suitable for tumor proportion score (TPS) evaluation of PD-L1 expression (94,95). As a general rule, Reck et al. reported, in advanced stage NSCLC patients who displayed a PD-L1 expression on at least 50% of tumor cells, a significantly higher progression-free survival and overall survival with limited adverse events of pembrolizumab with respect to platinum-based chemotherapy (90). However, despite these encouraging results, discordant data have been reported in literature. In fact, it has been highlighted that patients with a negative PD-L1 IHC/ICC may respond to ICIs whereas other with a high PD-L1 expression can show no responsiveness to immunotherapy (96,97). This may be due to the heterogeneous expression of PD-L1 in different histological sub-types (98,99). Currently, different clones are commercially available for PD-L1 testing (100). Among these, 22C3 clone (pharmDx, Agilent Technologies, Inc., Santa Clara, CA, USA) is the only companion diagnostic test for the administration of pembrolizumab (101). Conversely, 28-8, SP142 and SP263 clones for nivolumab, atezolizumab and durvalumab, respectively are only being adopted as complementary diagnostics (101,102). However, several efforts have been spent to evaluate the reproducibility and interchangeability of the different clones. In particular, the Blueprint phase 1 and 2 projects clearly demonstrated the interchangeability of clones 22C3, 28-8, and SP263 assays and the lower sensitivity of the SP142 assay for the evaluation of PD-L1 expression on both histological and cytological samples (94,95). Beyond histological specimens, in fact, even cytological samples demonstrated to be a suitable starting specimen to assess PD-L1 expression status before ICIs administration (19,103,104).


Conclusions

In conclusion, although NGS should be preferred over the other molecular diagnostic approaches in advanced stage NSLC patients, enabling to simultaneously analyze different DNA- and RNA-based biomarkers (13), IHC/ICC still represents a valid diagnostic tool for gene fusions detection. In addition, IHC/ICC approach is currently the only available diagnostic tool for the assessment of PD-L1 expression in order to administrate ICIs.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, Journal of Xiangya Medicine for the series “Predictive Molecular Pathology in Lung Cancer”. The article has undergone external peer review.

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://jxym.amegroups.com/article/view/10.21037/jxym-22-7/rc

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://jxym.amegroups.com/article/view/10.21037/jxym-22-7/coif). The series “Predictive Molecular Pathology in Lung Cancer” was commissioned by the editorial office without any funding or sponsorship. GT served as the unpaid Guest Editor of the series, and reports personal fees (as speaker bureau or advisor) from Roche, MSD, Pfizer, Boehringer Ingelheim, Eli Lilly, BMS, GSK, Menarini, AstraZeneca, Amgen and Bayer, unrelated to the current work. PP has received personal fees as speaker bureau from Novartis, unrelated to the current work. EV has received personal fees (as consultant and/or speaker bureau) from Diaceutics and AstraZeneca, unrelated to the current work. The authors have no other conflicts of interest to declare.

Ethical Statement:The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2022. CA Cancer J Clin 2022;72:7-33. [Crossref] [PubMed]
  2. Russo A, Lopes AR, McCusker MG, et al. New Targets in Lung Cancer (Excluding EGFR, ALK, ROS1). Curr Oncol Rep 2020;22:48. [Crossref] [PubMed]
  3. Tan AC, Tan DSW. Targeted Therapies for Lung Cancer Patients With Oncogenic Driver Molecular Alterations. J Clin Oncol 2022;40:611-25. [Crossref] [PubMed]
  4. Malapelle U, Muscarella LA, Pisapia P, et al. Targeting emerging molecular alterations in the treatment of non-small cell lung cancer: current challenges and the way forward. Expert Opin Investig Drugs 2020;29:363-72. [Crossref] [PubMed]
  5. Lindeman NI, Cagle PT, Aisner DL, et al. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. Arch Pathol Lab Med 2018;142:321-46. [Crossref] [PubMed]
  6. Kalemkerian GP, Narula N, Kennedy EB, et al. Molecular Testing Guideline for the Selection of Patients With Lung Cancer for Treatment With Targeted Tyrosine Kinase Inhibitors: American Society of Clinical Oncology Endorsement of the College of American Pathologists/International Association for the Study of Lung Cancer/Association for Molecular Pathology Clinical Practice Guideline Update. J Clin Oncol 2018;36:911-9. [Crossref] [PubMed]
  7. Ettinger DS, Wood DE, Aisner DL, et al. NCCN Guidelines Insights: Non-Small Cell Lung Cancer, Version 2.2021. J Natl Compr Canc Netw 2021;19:254-66. [Crossref] [PubMed]
  8. Kerr KM, Bibeau F, Thunnissen E, et al. The evolving landscape of biomarker testing for non-small cell lung cancer in Europe. Lung Cancer 2021;154:161-75. [Crossref] [PubMed]
  9. Angerilli V, Galuppini F, Pagni F, et al. The Role of the Pathologist in the Next-Generation Era of Tumor Molecular Characterization. Diagnostics (Basel) 2021;11:339. [Crossref] [PubMed]
  10. 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;10:1243-60. [Crossref] [PubMed]
  11. Nicholson AG, Tsao MS, Beasley MB, et al. The 2021 WHO Classification of Lung Tumors: Impact of Advances Since 2015. J Thorac Oncol 2022;17:362-87. [Crossref] [PubMed]
  12. Schmitt F, Di Lorito A, Vielh P. Molecular Testing on Cytology for Gene Fusion Detection. Front Med (Lausanne) 2021;8:643113. [Crossref] [PubMed]
  13. Pisapia P, Pepe F, Sgariglia R, et al. Methods for actionable gene fusion detection in lung cancer: now and in the future. Pharmacogenomics 2021;22:833-47. [Crossref] [PubMed]
  14. Pisapia P, Lozano MD, Vigliar E, et al. ALK and ROS1 testing on lung cancer cytologic samples: Perspectives. Cancer Cytopathol 2017;125:817-30. [Crossref] [PubMed]
  15. Bruno R, Fontanini G. Next Generation Sequencing for Gene Fusion Analysis in Lung Cancer: A Literature Review. Diagnostics (Basel) 2020;10:521. [Crossref] [PubMed]
  16. Schram AM, Chang MT, Jonsson P, et al. Fusions in solid tumours: diagnostic strategies, targeted therapy, and acquired resistance. Nat Rev Clin Oncol 2017;14:735-48. [Crossref] [PubMed]
  17. Udall M, Rizzo M, Kenny J, et al. PD-L1 diagnostic tests: a systematic literature review of scoring algorithms and test-validation metrics. Diagn Pathol 2018;13:12. [Crossref] [PubMed]
  18. Scheel AH, Schäfer SC. Current PD-L1 immunohistochemistry for non-small cell lung cancer. J Thorac Dis 2018;10:1217-9. [Crossref] [PubMed]
  19. Tejerina E, Garca Tobar L, Echeveste JI, et al. PD-L1 in Cytological Samples: A Review and a Practical Approach. Front Med (Lausanne) 2021;8:668612. [Crossref] [PubMed]
  20. Akhtar M, Rashid S, Al-Bozom IA. PD-L1 immunostaining: what pathologists need to know. Diagn Pathol 2021;16:94. [Crossref] [PubMed]
  21. Soda M, Choi YL, Enomoto M, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 2007;448:561-6. [Crossref] [PubMed]
  22. Pan Y, Zhang Y, Li Y, et al. ALK, ROS1 and RET fusions in 1139 lung adenocarcinomas: a comprehensive study of common and fusion pattern-specific clinicopathologic, histologic and cytologic features. Lung Cancer 2014;84:121-6. [Crossref] [PubMed]
  23. Kwak EL, Bang YJ, Camidge DR, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med 2010;363:1693-703. [Crossref] [PubMed]
  24. Du X, Shao Y, Qin HF, et al. ALK-rearrangement in non-small-cell lung cancer (NSCLC). Thorac Cancer 2018;9:423-30. [Crossref] [PubMed]
  25. Bubendorf L, Lantuejoul S, de Langen AJ, et al. Nonsmall cell lung carcinoma: diagnostic difficulties in small biopsies and cytological specimens: Number 2 in the Series "Pathology for the clinician" Edited by Peter Dorfmüller and Alberto Cavazza. Eur Respir Rev 2017;26:170007.
  26. Gristina V, La Mantia M, Iacono F, et al. The Emerging Therapeutic Landscape of ALK Inhibitors in Non-Small Cell Lung Cancer. Pharmaceuticals (Basel) 2020;13:474. [Crossref] [PubMed]
  27. Hofman V, Lassalle S, Bence C, et al. Any Place for Immunohistochemistry within the Predictive Biomarkers of Treatment in Lung Cancer Patients? Cancers (Basel) 2018;10:70. [Crossref] [PubMed]
  28. Mino-Kenudson M. Immunohistochemistry for predictive biomarkers in non-small cell lung cancer. Transl Lung Cancer Res 2017;6:570-87. [Crossref] [PubMed]
  29. 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;11:487-95. [Crossref] [PubMed]
  30. Hofman P, Ilie M, Hofman V, et al. Immunohistochemistry to identify EGFR mutations or ALK rearrangements in patients with lung adenocarcinoma. Ann Oncol 2012;23:1738-43. [Crossref] [PubMed]
  31. Conde E, Hernandez S, Prieto M, et al. Profile of Ventana ALK (D5F3) companion diagnostic assay for non-small-cell lung carcinomas. Expert Rev Mol Diagn 2016;16:707-13. [Crossref] [PubMed]
  32. Kerr KM, López-Ríos F. Precision medicine in NSCLC and pathology: how does ALK fit in the pathway? Ann Oncol 2016;27:iii16-24. [Crossref] [PubMed]
  33. Shaw AT, Bauer TM, de Marinis F, et al. First-Line Lorlatinib or Crizotinib in Advanced ALK-Positive Lung Cancer. N Engl J Med 2020;383:2018-29. [Crossref] [PubMed]
  34. von Laffert M, Schirmacher P, Warth A, et al. ALK-Testing in non-small cell lung cancer (NSCLC): Immunohistochemistry (IHC) and/or fluorescence in-situ Hybridisation (FISH)?: Statement of the Germany Society for Pathology (DGP) and the Working Group Thoracic Oncology (AIO) of the German Cancer Society e.V. (Stellungnahme der Deutschen Gesellschaft für Pathologie und der AG Thorakale Onkologie der Arbeitsgemeinschaft Onkologie/Deutsche Krebsgesellschaft e.V.). Lung Cancer 2017;103:1-5. [Crossref] [PubMed]
  35. Lee K, Um SW, Jeong BH, et al. Triple Gene Analysis Using Samples Obtained by Endobronchial Ultrasound-guided Transbronchial Needle Aspiration. Intern Med 2016;55:3105-11. [Crossref] [PubMed]
  36. Minca EC, Lanigan CP, Reynolds JP, et al. ALK status testing in non-small-cell lung carcinoma by FISH on ThinPrep slides with cytology material. J Thorac Oncol 2014;9:464-8. [Crossref] [PubMed]
  37. Liu L, Zhan P, Zhou X, et al. Detection of EML4-ALK in lung adenocarcinoma using pleural effusion with FISH, IHC, and RT-PCR methods. PLoS One 2015;10:e0117032. [Crossref] [PubMed]
  38. Wang W, Tang Y, Li J, et al. Detection of ALK rearrangements in malignant pleural effusion cell blocks from patients with advanced non-small cell lung cancer: a comparison of Ventana immunohistochemistry and fluorescence in situ hybridization. Cancer Cytopathol 2015;123:117-22. [Crossref] [PubMed]
  39. Zhang C, Randolph ML, Jones KJ, et al. Anaplastic Lymphoma Kinase Immunocytochemistry on Cell-Transferred Cytologic Smears of Lung Adenocarcinoma. Acta Cytol 2015;59:213-8. [Crossref] [PubMed]
  40. Rosenblum F, Hutchinson LM, Garver J, et al. Cytology specimens offer an effective alternative to formalin-fixed tissue as demonstrated by novel automated detection for ALK break-apart FISH testing and immunohistochemistry in lung adenocarcinoma. Cancer Cytopathol 2014;122:810-21. [Crossref] [PubMed]
  41. 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;8:1004-11. [Crossref] [PubMed]
  42. Rikova K, Guo A, Zeng Q, et al. Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell 2007;131:1190-203. [Crossref] [PubMed]
  43. Savic S, Rothschild S, Bubendorf L. Lonely Driver ROS1. J Thorac Oncol 2017;12:776-7. [Crossref] [PubMed]
  44. Zhao J, Zheng J, Kong M, et al. Advanced lung adenocarcinomas with ROS1-rearrangement frequently show hepatoid cell. Oncotarget 2016;7:74162-70. [Crossref] [PubMed]
  45. Remon J, Pignataro D, Novello S, et al. Current treatment and future challenges in ROS1- and ALK-rearranged advanced non-small cell lung cancer. Cancer Treat Rev 2021;95:102178. [Crossref] [PubMed]
  46. 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;469:489-503. [Crossref] [PubMed]
  47. Su Y, Goncalves T, Dias-Santagata D, et al. Immunohistochemical Detection of ROS1 Fusion. Am J Clin Pathol 2017;147:77-82. [PubMed]
  48. Viola P, Maurya M, Croud J, et al. A Validation Study for the Use of ROS1 Immunohistochemical Staining in Screening for ROS1 Translocations in Lung Cancer. J Thorac Oncol 2016;11:1029-39. [Crossref] [PubMed]
  49. Sholl LM, Sun H, Butaney M, et al. ROS1 immunohistochemistry for detection of ROS1-rearranged lung adenocarcinomas. Am J Surg Pathol 2013;37:1441-9. [Crossref] [PubMed]
  50. Cha YJ, Lee JS, Kim HR, et al. Screening of ROS1 rearrangements in lung adenocarcinoma by immunohistochemistry and comparison with ALK rearrangements. PLoS One 2014;9:e103333. [Crossref] [PubMed]
  51. Chiari R, Buttitta F, Iacono D, et al. Dramatic response to crizotinib in ROS1 fluorescent in situ hybridization- and immunohistochemistry-positive lung adenocarcinoma: a case series. Clin Lung Cancer 2014;15:470-4. [Crossref] [PubMed]
  52. Rossi G, Ragazzi M, Tamagnini I, et al. Does Immunohistochemistry Represent a Robust Alternative Technique in Determining Drugable Predictive Gene Alterations in Non-Small Cell Lung Cancer? Curr Drug Targets 2017;18:13-26. [Crossref] [PubMed]
  53. 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;70:402-11. [Crossref] [PubMed]
  54. Conde E, Hernandez S, Martinez R, et al. Assessment of a New ROS1 Immunohistochemistry Clone (SP384) for the Identification of ROS1 Rearrangements in Patients with Non-Small Cell Lung Carcinoma: the ROSING Study. J Thorac Oncol 2019;14:2120-32. [Crossref] [PubMed]
  55. Kohno T, Ichikawa H, Totoki Y, et al. KIF5B-RET fusions in lung adenocarcinoma. Nat Med 2012;18:375-7. [Crossref] [PubMed]
  56. Li AY, McCusker MG, Russo A, et al. RET fusions in solid tumors. Cancer Treat Rev 2019;81:101911. [Crossref] [PubMed]
  57. Mendoza L. Clinical development of RET inhibitors in RET-rearranged non-small cell lung cancer: Update. Oncol Rev 2018;12:352. [Crossref] [PubMed]
  58. Lipson D, Capelletti M, Yelensky R, et al. Identification of new ALK and RET gene fusions from colorectal and lung cancer biopsies. Nat Med 2012;18:382-4. [Crossref] [PubMed]
  59. Li F, Feng Y, Fang R, et al. Identification of RET gene fusion by exon array analyses in "pan-negative" lung cancer from never smokers. Cell Res 2012;22:928-31. [Crossref] [PubMed]
  60. Takeuchi K, Soda M, Togashi Y, et al. RET, ROS1 and ALK fusions in lung cancer. Nat Med 2012;18:378-81. [Crossref] [PubMed]
  61. Yoo SS, Jin G, Jung HJ, et al. RET fusion genes in Korean non-small cell lung cancer. J Korean Med Sci 2013;28:1555-8. [Crossref] [PubMed]
  62. Lin C, Wang S, Xie W, et al. The RET fusion gene and its correlation with demographic and clinicopathological features of non-small cell lung cancer: a meta-analysis. Cancer Biol Ther 2015;16:1019-28. [Crossref] [PubMed]
  63. Kato S, Subbiah V, Marchlik E, et al. RET Aberrations in Diverse Cancers: Next-Generation Sequencing of 4,871 Patients. Clin Cancer Res 2017;23:1988-97. [Crossref] [PubMed]
  64. Wang R, Hu H, Pan Y, et al. RET fusions define a unique molecular and clinicopathologic subtype of non-small-cell lung cancer. J Clin Oncol 2012;30:4352-9. [Crossref] [PubMed]
  65. Markham A. Selpercatinib: First Approval. Drugs 2020;80:1119-24. [Crossref] [PubMed]
  66. Bradford D, Larkins E, Mushti SL, et al. FDA Approval Summary: Selpercatinib for the Treatment of Lung and Thyroid Cancers with RET Gene Mutations or Fusions. Clin Cancer Res 2021;27:2130-5. [Crossref] [PubMed]
  67. Markham A. Pralsetinib: First Approval. Drugs 2020;80:1865-70. [Crossref] [PubMed]
  68. Kim J, Bradford D, Larkins E, et al. FDA Approval Summary: Pralsetinib for the Treatment of Lung and Thyroid Cancers With RET Gene Mutations or Fusions. Clin Cancer Res 2021;27:5452-6. [Crossref] [PubMed]
  69. Approves Selpercatinib FDA. Pralsetinib May Soon Follow. Cancer Discov 2020;10:OF1. [Crossref] [PubMed]
  70. Gainor JF, Curigliano G, Kim DW, et al. Pralsetinib for RET fusion-positive non-small-cell lung cancer (ARROW): a multi-cohort, open-label, phase 1/2 study. Lancet Oncol 2021;22:959-69. [Crossref] [PubMed]
  71. Drilon A, Oxnard GR, Tan DSW, et al. Efficacy of Selpercatinib in RET Fusion-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2020;383:813-24. [Crossref] [PubMed]
  72. Go H, Jung YJ, Kang HW, et al. Diagnostic method for the detection of KIF5B-RET transformation in lung adenocarcinoma. Lung Cancer 2013;82:44-50. [Crossref] [PubMed]
  73. Sasaki H, Shimizu S, Tani Y, et al. RET expression and detection of KIF5B/RET gene rearrangements in Japanese lung cancer. Cancer Med 2012;1:68-75. [Crossref] [PubMed]
  74. Yang SR, Aypar U, Rosen EY, et al. A Performance Comparison of Commonly Used Assays to Detect RET Fusions. Clin Cancer Res 2021;27:1316-28. [Crossref] [PubMed]
  75. Vaishnavi A, Capelletti M, Le AT, et al. Oncogenic and drug-sensitive NTRK1 rearrangements in lung cancer. Nat Med 2013;19:1469-72. [Crossref] [PubMed]
  76. Stransky N, Cerami E, Schalm S, et al. The landscape of kinase fusions in cancer. Nat Commun 2014;5:4846. [Crossref] [PubMed]
  77. Harada T, Yatabe Y, Takeshita M, et al. Role and relevance of TrkB mutations and expression in non-small cell lung cancer. Clin Cancer Res 2011;17:2638-45. [Crossref] [PubMed]
  78. Marchetti A, Felicioni L, Pelosi G, et al. Frequent mutations in the neurotrophic tyrosine receptor kinase gene family in large cell neuroendocrine carcinoma of the lung. Hum Mutat 2008;29:609-16. [Crossref] [PubMed]
  79. Farago AF, Taylor MS, Doebele RC, et al. Clinicopathologic Features of Non-Small-Cell Lung Cancer Harboring an NTRK Gene Fusion. JCO Precis Oncol 2018; [Crossref] [PubMed]
  80. Drilon A. TRK inhibitors in TRK fusion-positive cancers. Ann Oncol 2019;30:viii23-30. [Crossref]
  81. Scott LJ. Larotrectinib: First Global Approval. Drugs 2019;79:201-6. [Crossref] [PubMed]
  82. Marcus L, Donoghue M, Aungst S, et al. FDA Approval Summary: Entrectinib for the Treatment of NTRK gene Fusion Solid Tumors. Clin Cancer Res 2021;27:928-32. [Crossref] [PubMed]
  83. Marchiò C, Scaltriti M, Ladanyi M, et al. ESMO recommendations on the standard methods to detect NTRK fusions in daily practice and clinical research. Ann Oncol 2019;30:1417-27. [Crossref] [PubMed]
  84. Penault-Llorca F, Rudzinski ER, Sepulveda AR. Testing algorithm for identification of patients with TRK fusion cancer. J Clin Pathol 2019;72:460-7. [Crossref] [PubMed]
  85. Solomon JP, Hechtman JF. Detection of NTRK Fusions: Merits and Limitations of Current Diagnostic Platforms. Cancer Res 2019;79:3163-8. [Crossref] [PubMed]
  86. Solomon JP, Linkov I, Rosado A, et al. NTRK fusion detection across multiple assays and 33,997 cases: diagnostic implications and pitfalls. Mod Pathol 2020;33:38-46. [Crossref] [PubMed]
  87. Hechtman JF, Benayed R, Hyman DM, et al. Pan-Trk Immunohistochemistry Is an Efficient and Reliable Screen for the Detection of NTRK Fusions. Am J Surg Pathol 2017;41:1547-51. [Crossref] [PubMed]
  88. Gatalica Z, Xiu J, Swensen J, et al. Molecular characterization of cancers with NTRK gene fusions. Mod Pathol 2019;32:147-53. [Crossref] [PubMed]
  89. Zito Marino F, Pagliuca F, Ronchi A, et al. NTRK Fusions, from the Diagnostic Algorithm to Innovative Treatment in the Era of Precision Medicine. Int J Mol Sci 2020;21:3718. [Crossref] [PubMed]
  90. Reck M, Rodríguez-Abreu D, Robinson AG, et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2016;375:1823-33. [Crossref] [PubMed]
  91. Mok TSK, Wu YL, Kudaba I, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. Lancet 2019;393:1819-30. [Crossref] [PubMed]
  92. Gandhi L, Rodríguez-Abreu D, Gadgeel S, et al. Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer. N Engl J Med 2018;378:2078-92. [Crossref] [PubMed]
  93. Lantuejoul S, Sound-Tsao M, Cooper WA, et al. PD-L1 Testing for Lung Cancer in 2019: Perspective From the IASLC Pathology Committee. J Thorac Oncol 2020;15:499-519. [Crossref] [PubMed]
  94. 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;12:208-22. [Crossref] [PubMed]
  95. Tsao MS, Kerr KM, Kockx M, et al. PD-L1 Immunohistochemistry Comparability Study in Real-Life Clinical Samples: Results of Blueprint Phase 2 Project. J Thorac Oncol 2018;13:1302-11. [Crossref] [PubMed]
  96. Dong Z, Li H, Zhou J, et al. The value of cell block based on fine needle aspiration for lung cancer diagnosis. J Thorac Dis 2017;9:2375-82. [Crossref] [PubMed]
  97. Chae YK, Pan A, Davis AA, et al. Biomarkers for PD-1/PD-L1 Blockade Therapy in Non-Small-cell Lung Cancer: Is PD-L1 Expression a Good Marker for Patient Selection? Clin Lung Cancer 2016;17:350-61. [Crossref] [PubMed]
  98. 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;27:147-53. [Crossref] [PubMed]
  99. Ng Kee Kwong F, Laggner U, McKinney O, et al. Expression of PD-L1 correlates with pleomorphic morphology and histological patterns of non-small-cell lung carcinomas. Histopathology 2018;72:1024-32. [Crossref] [PubMed]
  100. Parra ER, Villalobos P, Mino B, et al. Comparison of Different Antibody Clones for Immunohistochemistry Detection of Programmed Cell Death Ligand 1 (PD-L1) on Non-Small Cell Lung Carcinoma. Appl Immunohistochem Mol Morphol 2018;26:83-93. [Crossref] [PubMed]
  101. Brody R, Zhang Y, Ballas M, et al. PD-L1 expression in advanced NSCLC: Insights into risk stratification and treatment selection from a systematic literature review. Lung Cancer 2017;112:200-15. [Crossref] [PubMed]
  102. Hendry S, Byrne DJ, Wright GM, et al. Comparison of Four PD-L1 Immunohistochemical Assays in Lung Cancer. J Thorac Oncol 2018;13:367-76. [Crossref] [PubMed]
  103. Iaccarino A, Salatiello M, Migliatico I, et al. PD-L1 and beyond: Immuno-oncology in cytopathology. Cytopathology 2021;32:596-603. [Crossref] [PubMed]
  104. Vigliar E, Malapelle U, Iaccarino A, et al. PD-L1 expression on routine samples of non-small cell lung cancer: results and critical issues from a 1-year experience of a centralised laboratory. J Clin Pathol 2019;72:412-7. [Crossref] [PubMed]
doi: 10.21037/jxym-22-7
Cite this article as: Pisapia P, Acanfora G, De Luca C, Iaccarino A, Troncone G, Vigliar E. A narrative review of predictive immuno-histo/cyto chemistry in lung cancer: focus on gene fusions and PD-L1 expression. J Xiangya Med 2022;7:18.

Download Citation