Vol 20, No 1 (2024)
Review paper
Published online: 2023-05-29

open access

Page views 417
Article views/downloads 358
Get Citation

Connect on Social Media

Connect on Social Media

Pan-TRK immunohistochemistry as a tool in the screening for NTRK gene fusions in cancer patients

Monika Durzyńska1, Irmina M. Michałek1
Oncol Clin Pract 2024;20(1):15-21.

Abstract

Therapy with TRK inhibitors is a tumor-agnostic treatment directed against specific molecular changes rather than cancer type. NTRK fusions are rare in most prevalent cancers, accounting for less than 0.5% of cases. However, there is a group of rare cancers in which NTRK fusion is more prevalent. These include secretory carcinoma of the breast and salivary gland, childhood sarcomas, such as infantile fibrosarcoma, and cellular and mixed congenital mesoblastic nephroblastoma. The most common rearrangement pertains to NTRK3 and the most common fusion gene is ETV6. Identifying patients with NTRK gene fusions who would likely benefit from targeted therapy with TRK inhibitors requires practical diagnostic tools and an appropriate management strategy of diagnostic trajectory. The fusions can be detected by molecular biology techniques or pan-TRK immunohistochemistry. The latter detects NTRK1/2/3 gene fusions independently of the resulting fusion gene but does not determine which of them has been rearranged or what the fusion partner is. The sensitivity and specificity of the method reach 97% and 100%, respectively. Other advantages include the relatively low cost, short duration of examination, and broad accessibility of immunohistochemistry laboratories. These characteristics make this method a useful screening tool for detecting patients with NTRK gene fusions.

REVIEW ARTICLE

Oncology in Clinical Practice

Copyright © 2024 Via Medica

ISSN 2450–1654

e-ISSN 2450–6478

Pan-TRK immunohistochemistry as a tool in the screening for NTRK gene fusions in cancer patients

Monika DurzyńskaIrmina M. Michałek
Department of Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland

Address for correspondence:

Monika Durzyńska MD PhD

Department of Pathology,

Maria Sklodowska-Curie National Research

Institute of Oncology

ul. Roentgena 5, 02–781 Warsaw, Poland

e-mail: monika.durzynska@nio-pib.pl

DOI: 10.5603/OCP.2023.0024

Received: 19.04.2023 Accepted: 24.04.2023 Early publication date: 29.05.2023

ABSTRACT

Therapy with TRK inhibitors is a tumor-agnostic treatment directed against specific molecular changes rather than cancer type. NTRK fusions are rare in most prevalent cancers, accounting for less than 0.5% of cases. However, there is a group of rare cancers in which NTRK fusion is more prevalent. These include secretory carcinoma of the breast and salivary gland, childhood sarcomas, such as infantile fibrosarcoma, and cellular and mixed congenital mesoblastic nephroblastoma. The most common rearrangement pertains to NTRK3 and the most common fusion gene is ETV6. Identifying patients with NTRK gene fusions who would likely benefit from targeted therapy with TRK inhibitors requires practical diagnostic tools and an appropriate management strategy of diagnostic trajectory. The fusions can be detected by molecular biology techniques or pan-TRK immunohistochemistry. The latter detects NTRK1/2/3 gene fusions independently of the resulting fusion gene but does not determine which of them has been rearranged or what the fusion partner is. The sensitivity and specificity of the method reach 97% and 100%, respectively. Other advantages include the relatively low cost, short duration of examination, and broad accessibility of immunohistochemistry laboratories. These characteristics make this method a useful screening tool for detecting patients with NTRK gene fusions.

Keywords: NTRK genes, TRK inhibitors, diagnostic methods; immunohistochemistry

Oncol Clin Pract 2024; 20, 1: 1521

Cancers with NTRK gene fusions as a therapeutic target for TRK inhibitors

In recent years, apart from the methods used so far in the treatment of oncological patients, such as surgical treatment or radio- and chemotherapy, an increasing role is played by targeted therapy, including “tumor-agnostic” therapy, directed at specific molecular changes and not cancer type [1, 2]. Tropomyosin receptor kinase (TRK) inhibitors are examples of such therapies [3, 4].

Neurotrophic TRKs are transmembrane tyrosine kinases that are essential for regulating nerve cell growth, proliferation, and differentiation. These include three groups of proteins: TRKA, TRKB, and TRKC, encoded by NTRK1, NTRK2, and NTRK3, respectively [5]. The NTRK genes can be rearranged during carcinogenesis. The NTRK fusion combines sequences coding for TRK proteins with sequences of other genes, leading to new active protein production [6]. In tumors with NTRK gene fusion, constitutive (ligand-independent) activation of intracellular biological pathways leads to a signaling cascade that controls cell cycle progression, proliferation, apoptosis, and/or survival of cancer cells [7, 8].

Tropomyosin receptor kinase inhibitors can be used in patients with confirmed NTRK gene rearrangement, regardless of cancer type [3, 5]. Clinical trials with a TRK inhibitor, entrectinib, have shown effectiveness in treating diverse types of cancer, both locally advanced and generalized [9].

Cancers with NTRK gene fusions are rare, regardless of age group, and account for up to 0.3% of all malignancies [10]. NTRK gene fusions have been described in over 40 types of solid tumors [11], including pulmonary, colorectal, breast, and thyroid cancers; melanoma; glioblastoma; and several sarcomas [7, 12]. In addition, some rare tumors have a remarkably high incidence of NTRK fusions (> 90%). In adults, these tumors include secretory breast and salivary gland cancer, whereas, in children, they include infantile fibrosarcoma, secretory cancer of the salivary gland, and cellular and mixed congenital mesoblastic nephroblastoma [13, 14].

NTRK fusion detection methods

The infrequent occurrence of tumors with NTRK gene fusion requires practical diagnostic tools and appropriate diagnostic strategies [6, 7]. These fusions can be detected by immunohistochemistry (IHC), fluorescence in situ hybridization (FISH), reverse transcriptase polymerase chain reaction (RT-PCR), and next-generation sequencing (NGS) [7, 14]. These methods have different sensitivities, specificities, strengths, and limitations (Tab. 1).

Table 1. Methods for detecting NTRK gene fusions in tumors

Sensitivity

Specificity

Detection of all fusions

Detection of fusion partners

Detection of protein expression

IHC

Relatively high*

Relatively high*

Yes

No

Yes

FISH

High

High

One per probe

One per probe

No

RNA NGS

High

High

Yes

Yes

Yes

DNA NGS

Moderate

High

Yes

Yes

No

Using pan-TRK IHC, NTRK1/2/3 gene fusions are detected independently of the resulting fusion genes. However, it is not possible to determine the fusion partner or the rearranged NTRK gene. The sensitivity of the method varies between 75% and 97%, and specificity ranges from 92% to 100%. The advantages of the pan-TRK IHC technique include the relatively low cost of the test, short execution time, and availability of IHC laboratories. Due to the above qualities, this technique can be used to screen patients for NTRK fusion [6, 15].

Fluorescence in situ hybridization is a widely used diagnostic method that allows for the detection of chromosomal rearrangements. Fusion probes detect a specific type of fusion gene, such as ETV6-NTRK3, or break-apart probes that detect breaks such as those in NTRK3. However, FISH cannot determine whether the resulting fusion gene encodes a productive in-frame chimeric transcript or not. The recommendations for detecting the ETV6-NTRK3 fusion gene are the same as the general principles of the FISH method for detecting fusion genes. They include counting the fluorescent signals in at least 50 randomly selected, non-overlapping tumor cell nuclei by at least two experienced specialists. The usefulness of FISH in cancer screening for NTRK fusions is limited because of the variety of fusion partners and the ability to evaluate only one gene rearrangement at a time. This method may help detect the ETV6-NTRK3 fusion gene in tumors where this gene is present in most cases, such as secretory breast and salivary gland cancers [6, 7].

RNA NGS allows the detection of fusion genes that are transcribed. The main limitation of this method is the instability of the RNA material, especially in archival paraffin blocks. Evaluating the quality of RNA is critical for distinguishing possible false-negative results. According to the literature, only approximately 55% of archival samples meet the quality control requirements before sequencing, and the probability of quality control failure increases with the age of the analyzed material [7, 16].

Targeted DNA NGS tests consisting of panels of selected genes are increasingly being used, including those detecting NTRK1, NTRK2, and NTRK3 fusions. Although the DNA NGS method successfully detects gene rearrangements, not all NTRK fusions can be detected using targeted assays. NTRK2 and NTRK3 are particularly problematic, as they have large intronic regions [6]. Moreover, many NTRK fusions detected by DNA-based sequencing are of unknown functional significance and require confirmation by other assays such as RNA sequencing or IHC [6, 7].

Performance and interpretation of the pan-TRK IHC test

The IHC test aims to detect tumors with NTRK fusions, which will be subjected to further molecular analysis, usually using the DNA NGS technique. Therefore, special attention should be paid to pre-analytical factors and the assay process to minimize false-negative rates [17]. Proper conduct of the respective phases of the study affects the credibility of the results.

The first step is to select the optimal material for testing. IHC should be performed using histopathological samples that were fixed in 10% buffered formalin. The fixation time depends on the size of the tested sample and is 648 hours for small materials and 2472 hours for larger materials.

Among the available antibodies, the most frequently used and best-characterized clone is EPR17341 [7, 15]. This antibody detects the C-terminal region of TRK proteins A, B, and C, which are conserved in both the wild-type and fusion proteins. Although the expression of the wild-type TRK protein in most solid tumors is minimal and rare, the pan-TRK IHC assay does not distinguish between wild-type and fusion proteins. IHC determination should be performed following the staining protocol provided by the manufacturer [18]. In addition, negative and positive control stains should be performed each time to minimize the incidence of false positive and false negative results. A negative control is performed using rabbit monoclonal antibodies. A positive control is performed using a normal human appendix. The nerves and ganglion cells in the wall show a positive reaction in the pan-TRK IHC test, whereas other structures are not stained. Performing an external positive control allows for verification of the correctness of the IHC staining process, but it does not constitute control of the pre-analytical stage. Therefore, during the assessment of pan-TRK IHC preparations, attention should be paid to whether any neural structures would constitute an internal positive control [6, 14].

The pan-TRK IHC color reaction is highly variable and can be nuclear, perinuclear/nuclear membrane, cytoplasmic, cellular membrane, or a combination of these. In addition, the staining intensity varies from weak to strong. Any of the above types of staining, stronger than that in the background and present in at least 1% of tumor cells, is interpreted as a positive reaction [14, 15, 19]. The percentage of stained cells and the intensity of staining is higher at the periphery of the specimen and lower in the central part. This type of staining is related to pre-analytical factors such as material fixation. Therefore, pan-TRK IHC tests are best performed with a small amount of material, such as a core needle biopsy, rather than with postoperative material [14]. The most common type of staining observed is the cytoplasmic reaction, which is the most common source of false-positive results compared to other types of expression. Moreover, false-positive pan-TRK IHC results are more common in tumors with muscular and nervous differentiation (leiomyosarcoma, glioma, and neuroblastoma) [7, 14]. In addition, there is a link between the type of color reaction and the occurrence of a specific fusion gene. Positive nuclear staining is often associated with ETV6-NTRK3 and EML4-NTRK3 fusions, nuclear membrane staining with LMNA-NTRK1 fusions, and cell membrane staining with TPM3-NTRK1 and TRAF- -NTRK2 fusions [15].

As mentioned above, the sensitivity of the pan-TRK IHC test has been reported to be between 75% and 97%. Discrepancies in the obtained results may result from different study populations (cancer types and fusion genes present in them) and pre-analytical procedures. The false-negative rate was higher for NTRK3 gene fusions (2127%) than for NTRK1 and NTRK2 fusions (< 10%) [13].

NTRK gene fusion tumors in the context of pan-TRK IHC results

Common neoplasms with the rare occurrence of NTRK gene fusions

This group of cancers includes colorectal, pulmonary, and breast cancers, where NTRK gene fusions occur in fewer than 1% of cases [14]. Within the gastrointestinal tract, NTRK fusions have also been detected in cancers of the pancreas, biliary tract, liver, appendix, and gallbladder (20). The most commonly described fusion genes include TPM3-NTRK1, LMNA-NTRK1, TPR-NTRK1, and ETV6-NTRK3 [15, 20]. In wild-type BRAF/RAS and high-grade microsatellite instability (MSI), an increase in NTRK fusions to approximately 5% has been observed [14]. In pan-TRK IHC, these tumors are usually characterized by strong cytoplasmic staining, which may be accompanied by perinuclear staining (LMNA fusion partner) or membrane staining (TPM3 fusion partner) [15].

In non-small cell lung cancer (NSCLC), mainly glandular NSCLC, NTRK gene rearrangements have been detected (most commonly NTRK1). The prevalence of such detected fusions is less than 1% [8]. In pan-TRK IHC, strong nuclear and cytoplasmic staining is usually observed [14].

In adult thyroid cancers, NTRK gene fusions occur in 24% of cases, both in well-differentiated, poorly differentiated, and undifferentiated cancers. In the pediatric group, NTRK fusions are more common in papillary thyroid carcinoma (815%) [21, 22]. The most common fusion gene is ETV6-NTRK3. A positive granular cytoplasmic reaction is observed in pan-TRK IHC (Fig. 1A, B). The sensitivity of this method in thyroid cancers is low, and the rate of false-negative results varies between 25% and 50% and is more common in the case of NTRK3 fusions [13].

Figure 1. A. Papillary thyroid carcinoma, follicular variant with a confirmed VIM-NTRK3 fusion gene, HE 200×; B. Papillary thyroid carcinoma, follicular variant with a confirmed VIM-NTRK3 fusion gene; Pan-TRK IHC 200×, with perinuclear and cytoplasmic staining of weak and medium intensity; C. Secretory carcinoma of the salivary gland with the detected ETV6-NTRK3 fusion gene, HE 200×; D. Secretory carcinoma of the salivary gland with the detected ETV6-NTRK3 fusion gene; Pan-TRK IHC 200×, with a strong nuclear and cytoplasmic staining with a weak staining intensity; E. Spindle cell sarcoma of the cervix with a confirmed EML4-NTRK3 fusion gene, HE 200×; F. Spindle cell sarcoma of the cervix with a confirmed EML4-NTRK3 fusion gene; Pan-TRK IHC 200×, a strong cytoplasmic reaction is visible in the tumor cells, no color reaction in the overlying epithelium and the subepithelial layer

Rare tumors with a low prevalence of NTRK gene rearrangements include glioblastoma multiforme [15, 23], a malignant brain tumor with poor prognosis. In the case of this cancer, an effective anti-TRK-targeted therapy would be beneficial. The identification of rare glioblastomas with NTRK rearrangements requires reliable diagnostic tests. However, the pan-TRK IHC test is of limited use as a screening method in this group of cancers because of the high rate of false-positive results [23].

Rare tumors with a very high prevalence of NTRK gene fusions

This group of tumors includes cancers such as breast secretory carcinoma and salivary gland secretory carcinoma, as well as sarcomas, including infantile fibrosarcoma, cellular and mixed congenital mesoblastic nephroblastoma [14], and the recently described group of low-grade spindle cell sarcomas with NTRK gene rearrangements [24, 25].

Secretory carcinoma accounts for fewer than 0.05% of all infiltrating breast cancers and occurs mainly in adult women. In most cases, it is a triple-negative tumor or a tumor with low estrogen and progesterone receptor expression [26]. ETV6-NTRK3 fusion occurs in over 90% of cases [27, 28]. Pan-TRK IHC is positive in 96% of cases. It is usually characterized by a strong nuclear reaction, and rarely by a nuclear-cytoplasmic reaction of varying intensity. NTRK gene rearrangements may also occur in approximately 10% of non-secretory breast cancers, most often NTRK1 with various fusion partners [29]. The occurrence of NTRK fusions in both secretory and non-secretory breast cancers supports the rationale for performing the pan-TRK IHC test as a screening method to detect patients for treatment with TRK inhibitors [30].

Secretory carcinoma with a morphology similar to that of the breast may develop in the salivary glands, most often in the parotid gland, usually in adults [31, 32]. In nearly 100% of cases, it is characterized by ETV6 gene rearrangements, with NTRK3 being the fusion partner in 90% of the cases [31]. On pan-TRK IHC, strong nuclear expression is seen, usually accompanied by low-intensity positive cytoplasmic staining (Fig. 1C, D). The pan-TRK IHC method is characterized by high sensitivity (91%) and specificity (nearly 100%) for the detection of secretory carcinoma of the salivary gland with ETV6--NTRK3 fusion [33, 34]. In some cases, the nuclear reaction may be weakly intense or occur only focally, making the IHC test challenging. In addition, only cytoplasmic or membrane expression may be present in non-secretory salivary carcinomas [35]. A particular group is adenoid cystic carcinoma, in which a positive pan-TRK IHC test result is found in nearly 40% of cases (strong cytoplasmic staining), which does not correlate with the presence of NTRK gene fusions [36].

Sarcomas with widespread occurrence of NTRK gene fusions primarily include childhood cancer. Infantile fibrosarcoma is a fibroblastic tumor that typically affects superficial and deep soft tissues of the limbs, trunk, head, and neck. Analogous tumors in the kidney are termed cellular and mixed congenital mesoblastic nephromas. These cancers usually develop during the first year of life [37]. Approximately 90% of cases are characterized by the ETV6-NTRK3 gene fusion [38]. Other less common molecular changes include EML4-NTRK3 fusions or NTRK1 and NTRK2 gene rearrangements [16, 38]. Another group of spindle cell sarcomas with NTRK gene rearrangement is a newly described group of rare sarcomas with immunohistochemical co-expression of S100 and CD34 in the absence of SOX10 expression. This new category includes tumors previously described as lipofibromatosis-like neural and peripheral nerve sheath tumors. Most of these tumors develop superficially or deep within the extremities or trunk during the first two decades of life [38, 39]. In this group of sarcomas, NTRK1 fusions with various partners such as TPR and TPM3 are the most common [25]. In the described sarcomas, pan-TRK IHC reaction is positive in most cases (> 90%). In infantile fibrosarcomas, it is a strong nuclear reaction, whereas in neural tumors, similar to lipofibromatosis, it is usually a perinuclear and/or cytoplasmic reaction. In spindle cell sarcomas without NTRK fusion, pan-TRK IHC may only be positive in approximately 8% of the cases. The pan-TRK IHC test is characterized by high sensitivity in detecting childhood sarcomas with NTRK gene fusions and can be used as a screening method to qualify patients for therapy with TRK inhibitors [24].

A newly described adult sarcoma with an NTRK rearrangement is a cervical spindle cell sarcoma. It usually occurs in pre-menopausal women. The co-expression of S100 and CD34 characterizes the tumor cells. Desmin, estrogen receptor (ER), and progesterone receptor (PGR) are not expressed [40]. NTRK1 and NTRK3 rearrangements with different fusion partners occur in this group of sarcomas. Fusion genes described so far include but are not limited to TPM3-NTRK1, LMNA-NTRK1, TPR-NTRK1, SPECC1L-NTRK3, and RBPMS-NTRK3 [40–42]. In pan-TRK IHC, TRK expression was observed in tumor cells in all cases (100%). The type of staining (cytoplasmic, perinuclear, or nuclear) may be associated with the formation of the fusion gene (Fig. 1E, F). It should be emphasized that in a low percentage of leiomyosarcomas (approximately 5%), in which there is no NTRK gene fusion, a positive pan-TRK IHC test is observed [40].

Tumors expressing pan-TRK IHC without NTRK gene fusions

A group of cancers is pan-TRK-positive IHC without NTRK gene fusion. Other specific molecular changes may characterize these tumors. Within the head and neck, this group of tumors includes bi-phenotypic sarcomas of the nose and paranasal sinuses (BSNS). The tumor comprises spindle-shaped cells that co-express S100 and SMA but do not express SOX10 [43]. Bi-phenotypic sarcomas of the nose and paranasal sinuses with non-specific pan-TRK IHC expression have been reported [44]. A characteristic feature of BSNS is rearrangement of the PAX3 gene with the MAML3 fusion gene [45]. Because of the microscopic image, S100 expression, and the possibility of a positive pan-TRK IHC result, it is necessary to differentiate this tumor from spindle-cell sarcomas with NTRK fusion. Other tumors in this area with frequent positive pan-TRK IHC without NTRK rearrangements are olfactory neuroblastoma, childhood small-round-cell tumors, such as Ewing’s sarcoma [14, 46], adenoid cystic carcinoma of the salivary gland, and leiomyosarcoma.

Conclusions

Identifying cancer patients with NTRK gene fusions who could benefit from targeted therapy using TRK inhibitors requires adequate diagnostic tools. These tumors are diverse and rare. On the one hand, there is a group of rare cancers with widespread occurrence of NTRK gene fusions, and on the other hand, there is a group of common cancers in which such molecular changes occur very rarely.

The pan-TRK method is characterized by high sensitivity and specificity, which may vary depending on the type of cancer. The ability to correctly interpret the results of the pan-TRK IHC test in correlation with the type of cancer is crucial in detecting cancer patients with NTRK gene fusions.

The pan-TRK IHC test can be used as a screening method because of its low cost, short execution time, and widespread use of IHC techniques. Pan-TRK IHC-positive tumors should be further investigated by molecular biology techniques to confirm the existence of NTRK fusions definitively.

Article Information and Declaration

Author contributions

M.D.: concept and design, analysis and interpretation of data, drafting of the manuscript and critical revision of the manuscript for intellectual content.

I.M.M.: critical revision of the manuscript for intellectual content.

Funding

None to declared.

Acknowledgments

None to declared.

Conflict of interest

M.D.: fees for lectures from Roche and Via Medica. Did not affect the content of this article.

I.M.M.: declares no conflict of interests.

References

  1. Tsimberidou AM, Fountzilas E, Nikanjam M, et al. Review of precision cancer medicine: Evolution of the treatment paradigm. Cancer Treat Rev. 2020; 86: 102019, doi: 10.1016/j.ctrv.2020.102019, indexed in Pubmed: 32251926.
  2. Huang FW, Feng FY. A Tumor-Agnostic NTRK (TRK) Inhibitor. Cell. 2019; 177(1): 8, doi: 10.1016/j.cell.2019.02.049, indexed in Pubmed: 30901551.
  3. Drilon AT. inhibitors in TRK fusion-positive cancers. Ann Oncol. 2019; 30(Suppl 8): viii23viii30.
  4. Cocco E, Scaltriti M, Drilon A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat Rev Clin Oncol. 2018; 15(12): 731747, doi: 10.1038/s41571-018-0113-0, indexed in Pubmed: 30333516.
  5. Jiang T, Wang G, Liu Y, et al. Development of small-molecule tropomyosin receptor kinase (TRK) inhibitors for fusion cancers. Acta Pharm Sin B. 2021; 11(2): 355372, doi: 10.1016/j.apsb.2020.05.004, indexed in Pubmed: 33643817.
  6. 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(9): 14171427, doi: 10.1093/annonc/mdz204, indexed in Pubmed: 31268127.
  7. Solomon JP, Benayed R, Hechtman JF, et al. Identifying patients with NTRK fusion cancer. Ann Oncol. 2019; 30(Suppl_8): viii16viii22, doi: 10.1093/annonc/mdz384, indexed in Pubmed: 31738428.
  8. Gatalica Z, Xiu J, Swensen J, et al. Molecular characterization of cancers with NTRK gene fusions. Mod Pathol. 2019; 32(1): 147153, doi: 10.1038/s41379-018-0118-3, indexed in Pubmed: 30171197.
  9. Drilon A, Chiu CH, Fan Y, et al. trial investigators, trial investigators. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol. 2020; 21(2): 271282, doi: 10.1016/S1470-2045(19)30691-6, indexed in Pubmed: 31838007.
  10. Okamura R, Boichard A, Kato S, et al. Analysis of Alterations in Pan-Cancer Adult and Pediatric Malignancies: Implications for NTRK-Targeted Therapeutics. JCO Precis Oncol. 2018; 2018, doi: 10.1200/PO.18.00183, indexed in Pubmed: 30637364.
  11. Frampton JE, Frampton JE. Entrectinib: A Review in NTRK+ Solid Tumours and ROS1+ NSCLC. Drugs. 2021; 81(6): 697708, doi: 10.1007/s40265-021-01503-3, indexed in Pubmed: 33871816.
  12. Sbaraglia M, Bellan E, Dei Tos AP. The 2020 WHO Classification of Soft Tissue Tumours: news and perspectives. Pathologica. 2021; 113(2): 7084, doi: 10.32074/1591-951X-213, indexed in Pubmed: 33179614.
  13. Hondelink LM, Schrader AMR, Asri Aghmuni G, et al. The sensitivity of pan-TRK immunohistochemistry in solid tumours: A meta-analysis. Eur J Cancer. 2022; 173: 229237, doi: 10.1016/j.ejca.2022.06.030, indexed in Pubmed: 35933886.
  14. Conde E, Hernandez S, Sanchez E, et al. Pan-TRK Immunohistochemistry: An Example-Based Practical Approach to Efficiently Identify Patients With NTRK Fusion Cancer. Arch Pathol Lab Med. 2021; 145(8): 10311040, doi: 10.5858/arpa.2020-0400-RA, indexed in Pubmed: 33112951.
  15. Chiang S, Cotzia P, 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(11): 15471551, doi: 10.1097/PAS.0000000000000911, indexed in Pubmed: 28719467.
  16. Church AJ, Calicchio ML, Nardi V, et al. Recurrent EML4-NTRK3 fusions in infantile fibrosarcoma and congenital mesoblastic nephroma suggest a revised testing strategy. Mod Pathol. 2018; 31(3): 463473, doi: 10.1038/modpathol.2017.127, indexed in Pubmed: 29099503.
  17. Marchiò C, Dowsett M, Reis-Filho JS. Revisiting the technical validation of tumour biomarker assays: how to open a Pandora’s box. BMC Med. 2011; 9: 41, doi: 10.1186/1741-7015-9-41, indexed in Pubmed: 21504565.
  18. Roche. VENTANA pan-TRK (EPR17341) Assay 2022. https://www.rochebiomarkers.be/content/media/Files/Bijsluiter_790-70261017533EN.pdf (05.04.2023).
  19. Brčić I, Godschachner TM, Bergovec M, et al. Broadening the spectrum of NTRK rearranged mesenchymal tumors and usefulness of pan-TRK immunohistochemistry for identification of NTRK fusions. Mod Pathol. 2021; 34(2): 396407, doi: 10.1038/s41379-020-00657-x, indexed in Pubmed: 32860002.
  20. Lasota J, Chłopek M, Lamoureux J, et al. Colonic Adenocarcinomas Harboring NTRK Fusion Genes: A Clinicopathologic and Molecular Genetic Study of 16 Cases and Review of the Literature. Am J Surg Pathol. 2020; 44(2): 16273.
  21. Macerola E, Proietti A, Poma AM, et al. Limited Accuracy of Pan-Trk Immunohistochemistry Screening for Rearrangements in Follicular-Derived Thyroid Carcinoma. Int J Mol Sci. 2022; 23(13), doi: 10.3390/ijms23137470, indexed in Pubmed: 35806472.
  22. Ricarte-Filho J, Halada S, O’Neill A, et al. The clinical aspect of NTRK-fusions in pediatric papillary thyroid cancer. Cancer Genetics. 2022; 262-263: 5763, doi: 10.1016/j.cancergen.2022.01.002.
  23. Bourhis A, Caumont C, Quintin-Roué I, et al. Detection of NTRK fusions in glioblastoma: fluorescent in situ hybridisation is more useful than pan-TRK immunohistochemistry as a screening tool prior to RNA sequencing. Pathology. 2022; 54(1): 5562, doi: 10.1016/j.pathol.2021.05.100, indexed in Pubmed: 34518039.
  24. Hung YP, Fletcher CDM, Hornick JL. Evaluation of pan-TRK immunohistochemistry in infantile fibrosarcoma, lipofibromatosis-like neural tumour and histological mimics. Histopathology. 2018; 73(4): 634644, doi: 10.1111/his.13666, indexed in Pubmed: 29863809.
  25. Agaram NP, Zhang L, Sung YS, et al. Recurrent NTRK1 Gene Fusions Define a Novel Subset of Locally Aggressive Lipofibromatosis-like Neural Tumors. Am J Surg Pathol. 2016; 40(10): 14071416, doi: 10.1097/PAS.0000000000000675, indexed in Pubmed: 27259011.
  26. Horowitz DP, Sharma CS, Connolly E, et al. Secretory carcinoma of the breast: results from the survival, epidemiology and end results database. Breast. 2012; 21(3): 350353, doi: 10.1016/j.breast.2012.02.013, indexed in Pubmed: 22494666.
  27. Del Castillo M, Chibon F, Arnould L, et al. Secretory Breast Carcinoma: A Histopathologic and Genomic Spectrum Characterized by a Joint Specific ETV6-NTRK3 Gene Fusion. Am J Surg Pathol. 2015; 39(11): 14581467, doi: 10.1097/PAS.0000000000000487, indexed in Pubmed: 26291510.
  28. Krings G, Joseph NM, Bean GR, et al. Genomic profiling of breast secretory carcinomas reveals distinct genetics from other breast cancers and similarity to mammary analog secretory carcinomas. Mod Pathol. 2017; 30(8): 10861099, doi: 10.1038/modpathol.2017.32, indexed in Pubmed: 28548128.
  29. Maund SL, Sokol ES, Ang Houle A, et al. NTRK gene fusions are detected in both secretory and non-secretory breast cancers. Pathol Int. 2022; 72(3): 187192, doi: 10.1111/pin.13204, indexed in Pubmed: 35102630.
  30. Shukla N, Roberts SS, Baki MO, et al. Successful Targeted Therapy of Refractory Pediatric Fusion-Positive Secretory Breast Carcinoma. JCO Precis Oncol. 2017; 2017, doi: 10.1200/PO.17.00034, indexed in Pubmed: 29623306.
  31. Skálová A, Vanecek T, Sima R, et al. Mammary analogue secretory carcinoma of salivary glands, containing the ETV6-NTRK3 fusion gene: a hitherto undescribed salivary gland tumor entity. Am J Surg Pathol. 2010; 34(5): 599608, doi: 10.1097/PAS.0b013e3181d9efcc, indexed in Pubmed: 20410810.
  32. Bishop JA, Yonescu R, Batista D, et al. Most nonparotid „acinic cell carcinomas” represent mammary analog secretory carcinomas. Am J Surg Pathol. 2013; 37(7): 10531057, doi: 10.1097/PAS.0b013e3182841554, indexed in Pubmed: 23681074.
  33. Sharma P, Sivakumar N, Pandiar D. Diagnostic accuracy of pan-TRK immunohistochemistry in differentiating secretory carcinoma from acinic cell carcinoma of salivary gland-A systematic review. J Oral Pathol Med. 2023; 52(3): 255262, doi: 10.1111/jop.13373, indexed in Pubmed: 36207812.
  34. Su YJ, Lee YH, Jin YT, et al. Using pan-TRK and RET Immunohistochemistry for the Detection of Fusion Types of Salivary Gland Secretory Carcinoma. Appl Immunohistochem Mol Morphol. 2022; 30(4): 264272, doi: 10.1097/PAI.0000000000001003, indexed in Pubmed: 35384876.
  35. Hung YP, Jo VY, Hornick JL. Immunohistochemistry with a pan-TRK antibody distinguishes secretory carcinoma of the salivary gland from acinic cell carcinoma. Histopathology. 2019; 75(1): 5462, doi: 10.1111/his.13845, indexed in Pubmed: 30801752.
  36. Guibourg B, Cloarec E, Conan-Charlet V, et al. EPR17341 and A7H6R pan-TRK Immunohistochemistry Result in Highly Different Staining Patterns in a Series of Salivary Gland Tumors. Appl Immunohistochem Mol Morphol. 2020; 28(9): 719724, doi: 10.1097/PAI.0000000000000825, indexed in Pubmed: 32187023.
  37. Orbach D, Rey A, Cecchetto G, et al. Infantile fibrosarcoma: management based on the European experience. J Clin Oncol. 2010; 28(2): 318323, doi: 10.1200/JCO.2009.21.9972, indexed in Pubmed: 19917847.
  38. Davis JL, Lockwood CM, Stohr B, et al. Expanding the Spectrum of Pediatric NTRK-rearranged Mesenchymal Tumors. Am J Surg Pathol. 2019; 43(4): 435445, doi: 10.1097/PAS.0000000000001203, indexed in Pubmed: 30585824.
  39. Antonescu CR. Emerging soft tissue tumors with kinase fusions: An overview of the recent literature with an emphasis on diagnostic criteria. Genes Chromosomes Cancer. 2020; 59(8): 437444, doi: 10.1002/gcc.22846, indexed in Pubmed: 32243019.
  40. Chiang S, Cotzia P, Hyman DM, et al. NTRK Fusions Define a Novel Uterine Sarcoma Subtype With Features of Fibrosarcoma. Am J Surg Pathol. 2018; 42(6): 791798, doi: 10.1097/PAS.0000000000001055, indexed in Pubmed: 29553955.
  41. Nilforoushan N, Wethington SL, Nonogaki H, et al. NTRK-Fusion Sarcoma of the Uterine Cervix: Report of 2 Cases With Comparative Clinicopathologic Features. Int J Gynecol Pathol. 2022; 41(6): 642648, doi: 10.1097/PGP.0000000000000834, indexed in Pubmed: 34723848.
  42. Hodgson A, Pun C, Djordjevic B, et al. NTRK-rearranged Cervical Sarcoma: Expanding the Clinicopathologic Spectrum. Int J Gynecol Pathol. 2021; 40(1): 7377, doi: 10.1097/PGP.0000000000000669, indexed in Pubmed: 32044823.
  43. Kuczkiewicz-Siemion O, Prochorec-Sobieszek M, Rysz M, et al. Small Biopsy Samples: Are They Representative for Biphenotypic Sinonasal Sarcoma? Diagnostics (Basel). 2022; 12(10), doi: 10.3390/diagnostics12102528, indexed in Pubmed: 36292216.
  44. Nichols MM, Alruwaii F, Chaaban M, et al. Biphenotypic Sinonasal Sarcoma with a Novel PAX3::FOXO6 Fusion: A Case Report and Review of the Literature. Head Neck Pathol. 2023; 17(1): 259264, doi: 10.1007/s12105-022-01479-w, indexed in Pubmed: 36169791.
  45. Fritchie KJ, Jin L, Wang X, et al. Fusion gene profile of biphenotypic sinonasal sarcoma: an analysis of 44 cases. Histopathology. 2016; 69(6): 930936, doi: 10.1111/his.13045, indexed in Pubmed: 27454570.
  46. Wong DD, Vargas AC, Bonar F, et al. NTRK-rearranged mesenchymal tumours: diagnostic challenges, morphological patterns and proposed testing algorithm. Pathology. 2020; 52(4): 401409, doi: 10.1016/j.pathol.2020.02.004, indexed in Pubmed: 32278476.