Next Article in Journal
Revisiting the Interaction of Melittin with Phospholipid Bilayers: The Effects of Concentration and Ionic Strength
Next Article in Special Issue
The Added Value of Diagnostic and Theranostic PET Imaging for the Treatment of CNS Tumors
Previous Article in Journal
Constitutive Activation of Guanylate Cyclase by the G86R GCAP1 Variant Is Due to “Locking” Cation-π Interactions that Impair the Activator-to-Inhibitor Structural Transition
Previous Article in Special Issue
Dissecting Molecular Features of Gliomas: Genetic Loci and Validated Biomarkers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

NTRK Fusions in Central Nervous System Tumors: A Rare, but Worthy Target

1
Pathology Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy
2
Pathology Unit, Department of Oncology, University of Turin, 10126 Turin, Italy
3
Pediatric Onco-Hematology Unit, Department of Pediatric and Public Health Sciences, University of Turin, 10126 Turin, Italy
4
Neurosurgery Unit, Department of Neurosciences, University of Turin, 10126 Turin, Italy
5
Department of Neuro-Oncology, University and City of Health and Science Hospital, 10126 Turin, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(3), 753; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21030753
Submission received: 30 December 2019 / Revised: 20 January 2020 / Accepted: 21 January 2020 / Published: 23 January 2020
(This article belongs to the Special Issue Molecular Biology of Brain Tumors)

Abstract

:
The neurotrophic tropomyosin receptor kinase (NTRK) genes (NTRK1, NTRK2, and NTRK3) code for three transmembrane high-affinity tyrosine-kinase receptors for nerve growth factors (TRK-A, TRK-B, and TRK-C) which are mainly involved in nervous system development. Loss of function alterations in these genes can lead to nervous system development problems; conversely, activating alterations harbor oncogenic potential, promoting cell proliferation/survival and tumorigenesis. Chromosomal rearrangements are the most clinically relevant alterations of pathological NTRK activation, leading to constitutionally active chimeric receptors. NTRK fusions have been detected with extremely variable frequencies in many pediatric and adult cancer types, including central nervous system (CNS) tumors. These alterations can be detected by different laboratory assays (e.g., immunohistochemistry, FISH, sequencing), but each of these approaches has specific advantages and limitations which must be taken into account for an appropriate use in diagnostics or research. Moreover, therapeutic targeting of this molecular marker recently showed extreme efficacy. Considering the overall lack of effective treatments for brain neoplasms, it is expected that detection of NTRK fusions will soon become a mainstay in the diagnostic assessment of CNS tumors, and thus in-depth knowledge regarding this topic is warranted.

1. Introduction

Traditionally, tumor diagnosis and prognostic evaluation, as well as therapeutic management, were addressed by histological examination alone, which was based on tumor morphology and complementary immunohistochemical profiling. Nowadays this approach is no longer adequate for complete tumor characterization since molecular profiling has become necessary for optimal patient management [1,2,3,4,5]. As a result, diagnostic algorithms are undergoing substantial changes for many tumor types: this molecular revolution has been fully undertaken by the latest 2016 World Health Organization (WHO) classification of central nervous system (CNS) neoplasms, as molecular markers (e.g., IDH1/IDH2 (Isocitrate dehydrogenase 1/2), 1p/19q codeletion, ATRX (transcriptional regulator ATRX), TP53 (tumor protein p53) etc.) have become mandatory for a conclusive diagnosis of many specific tumor entities [6,7,8,9]. Moreover, in the following few years since its publication, the diagnostic/prognostic/predictive importance of many additional molecular traits have been demonstrated and they are now being quickly translated into the routine clinical practice [10,11,12].
Despite the rarity, neurotrophic tropomyosin receptor kinase (NTRK) alterations recently gained attention because of the impressive therapeutic results achieved through their specific targeting. Since NTRK fusions have been found at significant frequencies in CNS tumors, which typically lack effective therapies, their detection is expected to soon become a mainstay in the diagnostic assessment of these tumors, and specific expertise in this topic will become mandatory.
In this Review, we will discuss the biology and physiological role of TRK receptors as well as their role in pathological conditions, focusing on the recently collected knowledge in brain tumors.

2. Biology of TRK Signaling

2.1. Characteristics of NTRK Genes and of TRK Signaling

Tyrosine receptor kinases are a group of cell-membrane high-affinity receptors sharing similar structures and intracellular signaling pathways, but with different mechanisms of activation and regulation. These receptors have specific growth factors as ligands and are involved in several fundamental functions for cell survival and activation, such as growth, differentiation, and apoptosis [13,14,15,16]. The oncogenic role of their alterations is well documented, as well as their possible exploitation as therapeutic targets [17,18,19,20,21,22,23,24,25].
NTRK are part of this group, consisting in a family of genes (NTRK-1, NTRK-2, and NTRK-3) located on chromosomes 1 (1q22), 9 (9q22), and 15 (15q25) and encoding for the TRK-A, TRK-B, and TRK-C proteins, respectively [26]. They were first identified and described as oncogenes in colorectal cancer by Pulciani et al. in 1982 [27], and then recognized as high-affinity neurotrophin receptors in 1989 [28]. They present the canonical structure of tyrosine kinase receptors, consisting of an intracellular domain with tyrosine-dependent kinase activity linked through the transmembrane structure to an extracellular domain made of two immunoglobulin-like high-affinity receptors and three leucine-rich motifs, the latter being specific of the NTRK family [13,14]. Specific neurotrophins, a subset of growth factors, are the main ligands of TRK proteins. TRK-A is probably the most studied and well-characterized receptor of the NTRK family and is preferentially bound by the nerve growth factor (NGF) [29]. Neurotrophin-3 (NT-3) binds TRK-C, while TRK-B has a lower binding specificity since both brain-derived growth factor (BDNF) and neurotrophin-4 (NT-4) can be ligands of this receptor [30,31,32,33,34]. Furthermore, also p75NTR, a membrane receptor, member of the tumor necrosis factor (TNF) receptor family, binds all the spectrum of neurotrophins described above and plays a crucial role in balancing cell survival versus death during CNS development [35]. Indeed, these last ligand-receptor relationships should be considered of low affinity [36,37]. p75NTR can also be considered a sort of “sparring partner” of TRK receptors, since their coexpression can enhance the activity of TRKs by improving the affinity between each TRK receptor and the corresponding ligands [38,39].
TRK receptors activation by their ligands leads to homodimerization of the intracellular domain, followed by phosphorylation of several tyrosine residues and consequent activation of the downstream signaling cascades (Figure 1). So far, TRK-A tyrosine residues have been thoroughly defined (Y496, Y676, Y680, Y681, and Y791) and TRK-B and TRK-C show a similar intracellular domain and activity. The intracellular domain, once phosphorylated, engages at least three different signaling cascades: the Ras-mitogen-activated protein kinase (MAPK), the phospholipase C-γ (PLC-γ), and the phosphatidylinositol 3-kinase (PI3-K) pathways. The final result of these interactions causes the activation of the neural cells, enabling their development and maintenance [40,41].
Another important signal transduction mechanism of TRK signaling is represented by the endocytic pathway. After binding with their respective partners, TRK receptors can be internalized within signaling endosomes which then can be transported back to the cell body where they can exert their function [42,43]. This mechanism, although it has been demonstrated for multiple receptor types, is especially relevant for neurons, since the cell soma can be significantly distant from the axon extremity. In particular, it has been shown that both signaling at the distal axon extremity and the retrograde trafficking of TRK-A bound with NGF are both necessary for neuronal survival and development.
Isoforms have been described for all three TRK, resulting from splicing variants of the NTRK genes and lacking specific subsets of exons [41]. Despite the consequent structural modifications, these isoforms keep the ability to transduce the signal once the ligand is bound [44,45,46]. However, each specific isoform presents peculiar characteristics both in terms of expression (e.g., expression in different tissues or with different timings) and activity [47,48,49,50].

2.2. The Physiological Role of NTRK Signaling and Its Role in Non-Neoplastic Diseases

The role of NTRK in the nervous system has been widely investigated (Figure 2): overall, TRK-B is probably the most represented receptor (mainly located in cortex, cerebellum, striatum, and hippocampus), while TRK-A and TRK-C show more restricted expression profiles, the former being limited to mature forebrain cholinergic neurons, and the latter mainly observed during neuronal development [51,52]. TRKs activation promotes and regulates the growth and elongation of dendrites and axons regardless of the site of origin and of the specific function [52,53].
In the peripheral nervous system (PNS), neutrophins binding and NTRK signaling is required for the survival of sensory and sympathetic ganglia [52,53,54,55,56]. Loss of NTRK expression in the development phase of mice and zebrafish has several consequences on their sensory systems, such as gustatory deficits and hearing and vision impairment [57,58]. Within CNS, NTRK gene expression is fundamental for neuron migration to the cortical layer (in particular for cerebellar granule neurons), and for their growth and maturation. Moreover, the hippocampal long-term maturation is strictly associated with NTRK expression by resident neurons [55,59,60,61,62].
Because of the significant role played in the physiology of nervous system development and maintenance, the level of expression of NTRK genes has been widely studied in pathological non-tumoral CNS conditions [63]. A significant downregulation of these receptors has been observed in the frontal cortex and in cholinergic basal nuclei of patients with Alzheimer’s disease. Moreover, a truncated isoform of TRK-B receptor resulted more expressed than the complete isoform in the cerebral cortex and the hippocampus of these patients. TRK-B truncated isoform lacks the intracellular tyrosine-dependent kinase domain, leading to a non-functional receptor [64].
Also, altered TRK signaling has been suggested in schizophrenia. Although the limited understanding of the pathogenetic mechanisms behind this disorder and the presumptive involvement of multiple genes, the 15q25 region has been identified as a possible culprit. This locus includes the NTRK3 gene: dysfunction of the corresponding TRK-C receptor could impair neural connections, plasticity, and development of the hippocampus and of the prefrontal cortex in these patients, together with a reduction of the overall levels of neurotrophins [65,66,67].
NTRK alterations have been proposed in many other neurological and psychiatric conditions, ranging from epilepsy (where increased levels of BDNF and of TRK-B resulted correlated with seizure induction and severity) to depression and addictive behaviors [67,68]. Additional data regarding the role of NTRK alterations in non-neoplastic diseases are now available, but this topic falls outside the scope of the present review.

3. NTRK in Tumor Development

3.1. The Oncogenic Role of NTRK: Fusions Versus Other Alterations

The oncogenic activation of NTRK can occur in several ways, including structural chromosomal rearrangements leading to gene fusions, splice variants, mutations, copy number alterations and increased expression. Considering their clinical relevance, these alterations can be clustered into two main groups: NTRK gene fusions leading to constitutively activated receptors versus the other mechanisms. Importantly, these other types of alterations are overall more frequent than NTRK fusions, but they cannot be effectively targeted with the currently available drugs (with the important exception of the NTRK mutations developed as a mechanism of resistance to therapeutic inhibition of NTRK fusions) and thus are presently considered non-druggable [69,70].
Regarding fusions, more than fifty NTRK fusion partners have been reported so far, confirming the extremely promiscuous nature of this rearrangement. Nevertheless, the same type of gene structural rearrangement is preserved: the 3′ region of the NTRK gene is fused with the 5′ region of a partner gene. The resulting chimeric protein keeps the NTRK tyrosine kinase domain with the ability to activate the usual intracellular pathway, but it becomes ligand-independent thanks to the partner gene component. The fusion mechanism described above for NTRK oncogenic activation is comparable to those occurring in other oncogenes with a kinase-domain component, such as ALK and ROS1 [71,72,73,74]. Indeed, gene fusions of receptor tyrosine kinases is a common oncogenic mechanism shared by multiple tumor types and leading to oncogene addiction, although the specifically involved genes can vary between the different neoplasms. For instance, if we consider non-small cell lung cancer, fusions involving ALK, ROS1, RET, BRAF, EGFR, and NTRK have been reported [75].
Overall, NTRK fusions seem to be rarely present (<1%) in unselected large series of tumors; conversely, it can be practically considered a pathognomonic marker of specific rare neoplasms including breast secretory carcinomas, mammary analogue secretory carcinoma of the salivary glands, infantile fibrosarcomas and congenital/infantile mesoblastic nephroma, narrowing a 100% prevalence [76,77,78,79,80]. Of interest, tumors harboring NTRK fusions often (>50%) present other genomic co-alterations in genes related to the NTRK intracellular pathways, such as the MAPK and the PI3K signaling cascades, TP53-associated genes, cell-cycle regulatory proteins and other tyrosine kinases, although strong mitogenic/driver alterations are usually mutually exclusive [70,81].
The true oncogenic potential of non-fusion NTRK alterations, such as mutations, gene amplifications and alternative splicing has yet to be confirmed [49,50,82,83,84,85,86]. Moreover, as it will be furtherly discussed later, these alternative types of alterations can play a crucial role in tumor resistance against NTRK-fusions inhibitors and therefore are being increasingly investigated [70,87].

3.2. NTRK Alterations in Non-CNS Tumors

Considered that NTRK was discovered as a potential oncogene in colorectal cancer (CRC) [27,88], and that tumors in which NTRK fusions can be considered pathognomonic belong to non-CNS cell-lineages as well, the oncogenic potential of this signaling pathway is not restricted to tissues with NTRK physiological expression [76,77,78,79,80].
Non-CNS NTRK-altered tumors include neoplasms with high incidence, but low frequency of NTRK fusions and rare tumors with extremely low incidence, but high frequency of this molecular hallmark. In this latter group, assessment of NTRK fusions can be used also as a diagnostic marker.
Among the first group, which represents the sharp majority of cases, NTRK fusions occur in no more than 4% of CRCs and the detected fusions so far are the TPM3-NTRK1 [89,90,91], the LMNA-NTRK1 [92], and the ETV6-NTRK3 [93]. CRC harboring NTRK, ALK, and ROS1 could be distinctively identified as tumors with high frequency of metastasis, poor prognosis, and specific mutational profile, characterized by high microsatellite instability (MSI) and RAS and BRAF wild-type status [70,94]. These observations can help aim NTRK assessment in this setting. The second main tumor type within this group is lung adenocarcinoma, in which NTRK rearrangements occur in about 3% of lesions. The main observed fusions are CD74-NTRK1, MPRIP-NTRK1, and TRIM24-NTRK2 [72,95]. Another carcinoma harboring NTRK as a potential oncogene is papillary thyroid carcinoma (PTC), with at least two different fusion products, TPM3-NTRK1 [96,97] and ETV6-NTRK3 [98].
The second group includes three entities, namely secretory carcinoma (either arising from breast or from salivary glands), congenital/infantile fibrosarcoma and congenital mesoblastic nephroma [76,99,100,101]. All these tumors present an NTRK fusion in more than 95% of cases, usually the ETV6-NTRK3. This fusion derives from a chromosomal translocation, t(12;15) (p13;q25), which combines exon 4, 5, or 6 of ETV6 and the kinase domain of NTRK3 [102].
Considering that the potential therapeutic efficacy has been demonstrated across all tumor types, NTRK fusions have been evaluated in several other neoplastic entities, ranging from Spitz tumor and melanoma to sarcomas (especially in the pediatric population), pancreatic cancer and cholangiocarcinoma, and neuroendocrine tumors [103,104,105,106,107,108,109]. Variable frequencies of NTRK rearrangements have been observed, but they usually are <10%.

3.3. NTRK Fusions in Pediatric CNS Tumors

In Europe and North America, the outcome landscape of pediatric tumors has recently changed: CNS neoplasms overtook hematological neoplasms as the leading cause of death within this population mainly because of the limited efficacy of the available treatments [110,111]. For this reason, pediatric CNS tumors represent an unmet need in oncology, requiring novel approaches for management and treatment.
Pediatric diffuse low and high-grade gliomas are undergoing significant changes in terms of diagnostic assessment, due to the increasing importance of molecular markers for classification and stratification [10,112,113]. These tumors also harbor peculiar molecular profiles which vary significantly from adult tumors, even in cases with similar histological features. Whenever a definitive diagnosis is achieved, the clinical behavior is still very heterogeneous and tumor recurrences are frequent even after multi-modal integrated treatments [112,114,115,116,117]. In particular, high-grade pediatric gliomas are associated with very limited outcomes and the possible treatments, which include radiotherapy, can lead to severe toxicities in children. A druggable target, like NTRK or other fusions, can thus actually have a major impact in this setting, improving disease control and allowing to delay other treatments with less favorable risk/benefit profiles [118].
NTRK alterations have been widely described in pediatric gliomas, both in low-grade and high-grade lesions (Table 1). Pilocytic astrocytoma (PA) is the most common pediatric glioma and usually shows a good outcome, especially after complete surgical resection; however, since recurrences do occur, it has been thoroughly investigated to look for new potential therapeutic targets including druggable fusions. MAPK pathway is commonly altered in PA and BRAF is the most frequently involved gene (e.g., KIAA1549: BRAF fusion, BRAF V600E mutation), while KRAS, and NF1 mutations can be observed in rare cases. Recently, NTRK fusions have also been observed in rare supratentorial PA with involvement of the NTRK2 gene [112,114,119].
Pediatric high-grade gliomas (pHGGs) are rare lesions with a dismal survival rate: the two-year survival rate for patients with supratentorial pHGGs range from 10 to 30 percent, and it is even lower (<10%) for diffuse intrinsic pontine gliomas (DIPGs). In this unsatisfactory scenario, molecular profiling of pHGGs seems imperative to improve the outcome of these patients by exploitation of specific therapeutic targets. As expected by their heterogeneity in terms of morphological features and clinical behavior, their molecular analysis showed a wide and challenging landscape, with several aberrant pathways and multiple mechanisms of tumor initiation/promotion being concurrently present. Although these findings are usually associated with intrinsic resistance to targeting of single alterations, a particular subset of non-brainstem high-grade gliomas has been identified in younger children (less than three years old) with high frequencies (up to 40%) of NTRK fusions (TPM3-NTRK1 and ETV6-NTRK3) without significant additional alterations, opening up new treatment scenarios for these selected cases [117,120,121,122,123]. Nevertheless, the overall NTRK-fusion rate of almost 4% observed in unselected cohorts of pediatric gliomas suggests its routine diagnostic assessment [70].
So far, NTRK fusions have not been detected in ependymoma, another frequent pediatric tumor. Although chromosomal rearrangements are key drivers of this neoplasm (e.g., RELA-fusion), NTRK signaling is not likely to be specifically involved based on the available data [120].
NTRK rearrangements have been investigated and discovered with a notable frequency in mixed glioneuronal tumors, a rare group of pediatric epileptogenic CNS neoplasms. Once again, although rare, NTRK (particularly NTRK1) fusions have been identified in both low-grade and high-grade glioneuronal tumors, ranging from ganglioglioma to diffuse leptomeningeal glioneuronal tumors [124,125,126,127].
Medulloblastoma, one of the most common and highly aggressive CNS non-glial pediatric tumors, showed no NTRK fusions. However, increased expression of non-mutated receptors, TRK-C in particular, has been found to be associated with a better clinical outcome and prognosis, suggesting the potential exploitation of NTRK signaling as a prognostic rather than predictive marker [128,129,130,131].

3.4. NTRK Fusions in Adult CNS Tumors

CNS tumors represent a challenging context also among adults with discouraging outcomes. Comprehensive molecular analyses of large cohorts of these tumors have been conducted, focusing on high grade gliomas (HGG) and glioblastoma (GBM), the latter being the most common glioma in adults with an extremely severe prognosis. IDH-wildtype GBM (the so-called primary glioblastoma) shows a broad spectrum of potentially targetable alterations, including a significant rate of fusions: chimeric fusion genes are often present, and involvement of all of the three NTRK genes has been demonstrated (Table 1), although with significant differences among the series [132,133]. Up to date, NTRK2 appears to be the most frequently involved gene (up to 11% of GBM), while NTRK1 fusions are definitely rarer (about 1%) and NTRK3 fusions seem to be extremely rare (one single case reported) [133,134,135,136,137]. Among low grade gliomas (LGG), a NTRK1 fusion was reported in an adult pilocytic astrocytoma [133].
Expression and methylation of wild-type NTRK genes has been investigated in different types of gliomas as well, revealing that LGGs present higher expression of TRK receptors compared to HGGs. Although these findings need to be further confirmed, lower expression levels in tumoral cells seem to be associated with increased malignant potential and poorer prognosis [138,139]. Accordingly, higher expression of NTRK receptors in neuroblastomas was found to be associated with a better outcome. This finding is possibly due to immunoregulatory mechanisms, thus widening the potential range of modulatory effects associated with this signaling pathway [140].

4. NTRK as a Novel Therapeutic Target

4.1. NTRK-Fusions Targeting: A Novel, Effective, Histology-Independent Anti-Neoplastic Treatment

Drug development in oncology has significantly changed since the discovery of targetable molecular alterations. Since these alterations are shared among completely independent tumor sites and types, basket trials were initiated, testing cohorts of patients with common molecular targets, despite the different tumor entity [141]. However, in some cases (e.g., mutated BRAF-inhibitors) response to treatments was still histology or tissue-dependent and thus drug approval was limited to specific indications. More recently, pembrolizumab, an anti-PD1 immunomodulatory drug, received tissue-agnostic approval considered the efficacy in a wide range of advanced tumor types sharing mismatch repair deficiency or high microsatellite instability. Similarly, NTRK-inhibitors are receiving tissue-agnostic (FDA) or histology-independent (EMA) approval based on high efficacy in pediatric and adult tumors harboring NTRK fusion regardless of the tumor site or specific fusion partner. So far, two first-generation molecules (entrectinib and larotrectinib) received FDA therapeutic approval for the treatment of NTRK fusion-positive tumors and the latter recently gained EMA approval as well [41].
Entrectinib (RXDX-101) was the first drug developed against NTRK fusions, targeting also ALK and ROS1 fusion proteins and harboring a good delivery rate through the blood-brain barrier [142]. In phase-I and II trials (ALKA-372-001, STARTRK-1, STARTRK-2, and STARTRK-NG), it showed significant results in pediatric and adult solid tumors, with efficacy in both primary and secondary CNS tumors [69,143]. In a recent series of pediatric high-grade gliomas reported at ASCO 2019, all 4 patients achieved a radiological response, including a complete response (2019 ASCO Annual Meeting, Abstract #: 10009).
Larotrectinib (LOXO-101) is highly specific for NTRK fusions only, and its efficacy has been tested in several trials (registered on ClinicalTrials.gov: NCT02637687, NCT02122913, NCT02637687, and NCT02576431) [144,145], with a well-documented efficacy against CNS tumors [145], as recently confirmed [146].
These results are important for two main reasons: (i) the overall rate of clinical and radiological responses is high (even close to 80%); (ii) response is usually durable, with patients achieving disease control for many months or even years.
Ongoing clinical trials with entrectinib and larotrectinib are now focused on elucidating their activity profile (e.g., to assess possible correlations with the specific fusion partners) and safety data (Table 2). Moreover, development and clinical testing of second-generation NTRK inhibitors is already ongoing (e.g., repotrectinib-TPX-0005 and LOXO-195-BAY2731954) [147,148], in order to compare their efficacy with first-generation drugs and, more importantly, to tackle tumor resistance to them.

4.2. Resistance Mechanisms to First-Generation NTRK Inhibitors

Acquired resistance during long-term treatment with targeted therapies is a major concern, as experienced with EGFR, ALK, and ROS1 inhibitors [149,150,151,152,153,154]. NTRK inhibitors make no exception to this statement, and disease progression has been now observed within the ongoing clinical trials.
Notably, excluding sporadic cases whose failure was related to non-appropriate patient recruitment [155], at least two broad mechanisms of resistance have been detected. The first one is related to off-target alterations, which reactivate one of the cellular pathways associated with NTRK fusions, usually the MAPK. As a matter of fact, MAPK signaling cascade may get activated by several signal transducers not related to NTRK at all. Examples of this resistance mechanism are the acquisition of the BRAFV600E or KRASG12D mutations or MET amplification. Of note, in these cases, prompt treatment with drugs targeting the new resistance-related alterations enabled new tumor responses [156].
The second tumor escape strategy (the so-called on-target resistance) is related to point mutations (i.e., solvent front, gatekeeper and xDFG mutations) of the NTRK fusion proteins, blocking drug binding. In this regard, next-generation NTRK inhibitors (e.g., repotrectnib—TPX-0005, LOXO-195-BAY2731954) have been developed, showing promising efficacy in targeting these mutated fusion proteins [87,155,157].
These data open several questions that will be answered by the upcoming trials: can resistance to first-generation inhibitors be avoided by modulating the treatment over time? Should patients directly receive second-generation inhibitors? How should patients be monitored during treatment to promptly detect resistance?

5. Testing for NTRK Fusions. Where Is Waldo?

Based on the previous considerations, NTRK fusions must now be considered an important molecular marker in CNS tumors, which can enable significant improvement of patients’ outcome by specific targeting. So, how can we efficiently test for these alterations taken into consideration their rarity?
NTRK oncogenic activation is a process that, starting from the chromosomal rearrangement, requires translation of the fusion gene and expression of the chimeric TRK protein. In light of these consequential steps, different laboratory assays can be used to find out whether a tumor is harboring a NTRK fusion (Table 3). Firstly, to investigate the DNA status, fluorescence in-situ hybridization (FISH) and DNA-based next-generation sequencing (NGS) can be used, while reverse transcription-polymerase chain reaction (RT-PCR), real time-PCR and RNA-based NGS analyses can evaluate the transcribed RNA. Finally, immunohistochemical staining (IHC) can directly assess the protein product.

5.1. Immunohistochemistry

IHC is a common, well-known and validated assay, with limited costs, and quick turnaround time (TAT), allowing histological correlations and also capable of intrinsically confirming the protein expression. The main limitation is that available antibodies are for the wild-type epitopes of TRK receptors, thus not specific for fusions and obviously they do not provide any information regarding the fusion partner. Different TRK antibodies are available, staining either single receptors (so far, antibodies for TRK-A and TRK-B are available as well as a cocktail of anti-TRK-A and anti-TRK-B) or all TRK proteins (pan-TRK antibody, clone EPR17341), which is also available for in vitro diagnostics (Ventana Medical Systems Inc., Tucson, AZ, USA). Of note, different staining patterns can be expected based on the involved genes: for example, NTRK3 fusions more often lead to a nuclear staining and a higher rate of false negatives (up to 45%) [77,78,79,80,106,158,159]. IHC can thus be used as an effective screening tool for most tumor types, but unfortunately specificity in CNS neoplasms seems to be low due to the physiological expression of NTRK in neural tissues. For instance, Solomon et al., reported an unsatisfactory specificity value of 20.8% in gliomas [160], thus screening by IHC should be avoided in this setting or used with extreme caution and confirmation by other techniques is warranted. A significant rate of false positive IHC results has also been observed in cases with smooth muscle or neuroendocrine differentiation and in small round cell tumors. Of note, in false positive samples, staining was limited to cytoplasm and/or cell membrane without nuclear staining.

5.2. Fluorescence In-Situ Hybridization

FISH-based assays are well-established to investigate chromosomal alterations, such as translocations, deletions, or amplifications, thus they could also be applied for evaluating NTRK fusions. Considered their promiscuous nature, break apart probes must be used which do not provide information on the fusion partner. Although, as it is true for IHC, FISH requires minimal formalin-fixed paraffin-embedded material and enables a low TAT, a specific expertise for a correct interpretation is required. Moreover, since investigation of all three NTRK genes requires three independent assays, a FISH-based approach cannot be envisaged for screening [161]. On the other hand, FISH has been suggested as a confirmatory assay with high sensitivity and specificity, although evaluations of larger series of NTRK-fusion tumors are warranted to assess potential limitations. In particular, if the fusion breakpoint is non-canonical, a false negative result can be observed.

5.3. DNA and RNA Molecular Testing

The second group of assays that can be used to look for NTRK fusions is based on extraction and analysis of nucleic acids. These techniques vary significantly in terms of complexity, costs, TAT (which is usually longer than IHC or FISH), required material, information provided, and thus optimal indications. DNA or RNA can be successfully tested by different assays, but with some important caveats: (i) RNA is more prone to be damaged, especially in FFPE material, thus special attention must be payed to pre-analytics; (ii) RNA-based assays usually require simpler analyses as intronic regions have been already removed; (iii) DNA-based assays can detect rearrangements which are not even transcribed and thus lack any relevance, while, conversely, they can miss fusions involving large intronic regions [162].
Considering the specific techniques, RT-PCR can be used for orthogonal validation of a specific fusion, but fusion partners must be already known, and specific primers must be designed, thus, it cannot be used for screening despite the low costs. Real-time PCR-based assays are now progressively becoming available, allowing assessment of a wide range of combinations of specific rearrangements/partners through an overall inexpensive analysis. The main limitations of this approach are that it usually does not provide information regarding the specific fusion partners and, since it evaluates a pre-determined set of rearrangements, rare or novel fusions will not be detected.
Regarding NGS, DNA assays are becoming routinely used in diagnostics as they can assess a wide range of clinically relevant alterations (mutations, copy number variations, tumor mutation burden), but reporting time and costs are significant, and specific facilities and expertise are required. Chromosomal rearrangements can also be detected, but sensitivity depends on the probe coverage of the involved genes. For example, the breakpoint of NTRK3 fusions often occurs within a highly repetitive, intronic region, leading to high false negative rates. Indeed, Solomon et al. found a 76.9% sensitivity when evaluating NTRK3 fusions using the MSK-IMPACT DNA-based NGS assay [160]. Conversely, RNA-NGS, including total RNA analysis, represent the optimal tools to investigate the whole fusion landscape of a tumor sample with high sensitivity and specificity. Integrated DNA/RNA-NGS assays can thus be used to achieve complete molecular profiling of a tumor and they will probably enter the diagnostic routine practice in the coming years considered the demands posed by precision medicine, of which targeting of NTRK fusions is an example.
Since every technique presents specific advantages and disadvantages, it is difficult to designate gold standard technique. Indeed, several algorithms have been already suggested, tailored to the different settings or tumor types. Most of them combine IHC staining as a screening tool, followed by confirmation through other techniques: following these algorithms, tumors are first evaluated by a rapid and cost-effective (but less specific) assay, allowing to focus more expensive, but highly specific tests on a smaller subset of cases [123,160,161,163,164,165,166,167]. For CNS tumors, considering the ever-increasing importance of extensive molecular profiling to achieve a correct diagnosis/classification, integration of NTRK fusion assessment in a dedicated NGS workflow seems desirable in the medium-term. However, as NGS availability is still limited for routine diagnostics in many centers and that IHC-screening efficacy is limited in this setting (because of a low specificity), real time-PCR assays could represent a good compromise in terms of cost-efficacy.
Finally, the optimal strategy for molecular profiling at disease progression after NTRK fusions-targeting should now be investigated. Present data suggest efficacy of liquid-based assessment, but given the wide range of both on-target and off-target resistance mechanisms, comprehensive assays seem to be necessary [156].

6. Conclusions

Management of CNS tumors represents a challenging therapeutic issue as curative surgical resection is often not feasible, and radiotherapy may have significant negative long-term consequences on neurocognitive functions (especially in children). The efficacy of chemotherapy drugs is limited, also due to the fact that blood-brain barrier considerably limits the chance of drugs to reach the tumor. Although the recognition of NTRK as a potential oncogene is now dated, the proper understanding of the specific mechanisms involved and their appreciation as a potential therapeutic target is far more recent. Despite the rarity of NTRK fusions, the potential clinical benefit for the small group of patients harboring these alterations appears to be extremely significant, thus fully awareness by physicians caring for brain tumors is now mandatory.

Author Contributions

Conceptualization, P.C. and L.B.; data collection and curation, A.G., R.S. (Rebecca Senetta), G.C., S.G.V., M.M., F.C., P.Z., D.G., A.P., R.R., R.S. (Riccardo Soffietti), F.F., M.P., P.C., and L.B., writing—original draft preparation, A.G. and L.B.; writing—review and editing, A.G., R.S. (Rebecca Senetta), G.C., S.G.V., M.M., F.C., P.Z., D.G., A.P., R.R., R.S. (Riccardo Soffietti), F.F., M.P., P.C., and L.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Rete Oncologica del Piemonte e della Valle d’Aosta and received funding specifically dedicated to the Department of Medical Sciences, University of Turin from Italian Ministry for Education, University and Research (Ministero dell’Istruzione, dell’Università e della Ricerca-MIUR) under the programme “Dipartimenti di Eccellenza 2018 – 2022”, Project n° D15D18000410001.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Chen, D.S.; Mellman, I. Elements of cancer immunity and the cancer-immune set point. Nature 2017, 541, 321–330. [Google Scholar] [CrossRef] [PubMed]
  2. Herbst, R.S.; Morgensztern, D.; Boshoff, C. The biology and management of non-small cell lung cancer. Nature 2018, 553, 446–454. [Google Scholar] [CrossRef] [PubMed]
  3. Ducreux, M.; Chamseddine, A.; Laurent-Puig, P.; Smolenschi, C.; Hollebecque, A.; Dartigues, P.; Samallin, E.; Boige, V.; Malka, D.; Gelli, M. Molecular targeted therapy of BRAF-mutant colorectal cancer. Ther. Adv. Med. Oncol. 2019, 11, 1758835919856494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Fussey, J.M.; Vaidya, B.; Kim, D.; Clark, J.; Ellard, S.; Smith, J.A. The role of molecular genetics in the clinical management of sporadic medullary thyroid carcinoma: A systematic review. Clin. Endocrinol. (Oxf.) 2019, 91, 697–707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Jorgensen, J.T. A paradigm shift in biomarker guided oncology drug development. Ann. Transl. Med. 2019, 7, 148. [Google Scholar] [CrossRef]
  6. Louis, D.N.; Ohgaki, H.; Wiestler, O.D.; Cavenee, W.K.; Ellison, D.W.; Figarella-Branger, D.; Perry, A.; Reifenberger, G.; von Deimling, A. International Agency for Research on Cancer. In WHO Classification of Tumours of the Central Nervous System, Revised 4th ed.; International Agency for Research on Cancer: Lyon, France, 2016; 408p. [Google Scholar]
  7. Scheie, D.; Kufaishi, H.H.A.; Broholm, H.; Lund, E.L.; de Stricker, K.; Melchior, L.C.; Grauslund, M. Biomarkers in tumors of the central nervous system—a review. APMIS 2019, 127, 265–287. [Google Scholar] [CrossRef] [Green Version]
  8. Burford, C.; Laxton, R.; Sidhu, Z.; Aizpurua, M.; King, A.; Bodi, I.; Ashkan, K.; Al-Sarraj, S. ATRX immunohistochemistry can help refine ‘not elsewhere classified’ categorisation for grade II/III gliomas. Br. J. Neurosurg. 2019, 33, 536–540. [Google Scholar] [CrossRef]
  9. Capper, D.; Jones, D.T.W.; Sill, M.; Hovestadt, V.; Schrimpf, D.; Sturm, D.; Koelsche, C.; Sahm, F.; Chavez, L.; Reuss, D.E.; et al. DNA methylation-based classification of central nervous system tumours. Nature 2018, 555, 469–474. [Google Scholar] [CrossRef]
  10. Ellison, D.W.; Hawkins, C.; Jones, D.T.W.; Onar-Thomas, A.; Pfister, S.M.; Reifenberger, G.; Louis, D.N. cIMPACT-NOW update 4: Diffuse gliomas characterized by MYB, MYBL1, or FGFR1 alterations or BRAF(V600E) mutation. Acta Neuropathol. 2019, 137, 683–687. [Google Scholar] [CrossRef]
  11. Ghasemi, D.R.; Sill, M.; Okonechnikov, K.; Korshunov, A.; Yip, S.; Schutz, P.W.; Scheie, D.; Kruse, A.; Harter, P.N.; Kastelan, M.; et al. MYCN amplification drives an aggressive form of spinal ependymoma. Acta Neuropathol. 2019, 138, 1075–1089. [Google Scholar] [CrossRef] [Green Version]
  12. Wefers, A.K.; Stichel, D.; Schrimpf, D.; Coras, R.; Pages, M.; Tauziede-Espariat, A.; Varlet, P.; Schwarz, D.; Soylemezoglu, F.; Pohl, U.; et al. Isomorphic diffuse glioma is a morphologically and molecularly distinct tumour entity with recurrent gene fusions of MYBL1 or MYB and a benign disease course. Acta Neuropathol. 2019. [Google Scholar] [CrossRef] [PubMed]
  13. Zwick, E.; Bange, J.; Ullrich, A. Receptor tyrosine kinase signalling as a target for cancer intervention strategies. Endocr. Relat. Cancer 2001, 8, 161–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Lemmon, M.A.; Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 2010, 141, 1117–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Schlessinger, J. Receptor tyrosine kinases: Legacy of the first two decades. Cold Spring Harb. Perspect. Biol. 2014, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Du, Z.; Lovly, C.M. Mechanisms of receptor tyrosine kinase activation in cancer. Mol. Cancer 2018, 17, 58. [Google Scholar] [CrossRef]
  17. Miles, J.; White, Y. Neratinib for the Treatment of Early-Stage HER2-Positive Breast Cancer. J. Adv. Pract. Oncol. 2018, 9, 750–754. [Google Scholar]
  18. Russo, A.; Franchina, T.; Ricciardi, G.; Battaglia, A.; Picciotto, M.; Adamo, V. Heterogeneous Responses to Epidermal Growth Factor Receptor (EGFR) Tyrosine Kinase Inhibitors (TKIs) in Patients with Uncommon EGFR Mutations: New Insights and Future Perspectives in this Complex Clinical Scenario. Int. J. Mol. Sci. 2019, 20, 1431. [Google Scholar] [CrossRef] [Green Version]
  19. Teishima, J.; Hayashi, T.; Nagamatsu, H.; Shoji, K.; Shikuma, H.; Yamanaka, R.; Sekino, Y.; Goto, K.; Inoue, S.; Matsubara, A. Fibroblast Growth Factor Family in the Progression of Prostate Cancer. J. Clin. Med. 2019, 8, 183. [Google Scholar] [CrossRef] [Green Version]
  20. Carlisle, J.W.; Ramalingam, S.S. Role of osimertinib in the treatment of EGFR-mutation positive non-small-cell lung cancer. Future Oncol. 2019, 15, 805–816. [Google Scholar] [CrossRef]
  21. Roskoski, R., Jr. Small molecule inhibitors targeting the EGFR/ErbB family of protein-tyrosine kinases in human cancers. Pharmacol. Res. 2019, 139, 395–411. [Google Scholar] [CrossRef]
  22. Goetze, T.O.; Al-Batran, S.E.; Berlth, F.; Hoelscher, A.H. Multimodal Treatment Strategies in Esophagogastric Junction Cancer: A Western Perspective. J. Gastric Cancer 2019, 19, 148–156. [Google Scholar] [CrossRef] [PubMed]
  23. Katoh, M. Fibroblast growth factor receptors as treatment targets in clinical oncology. Nat. Rev. Clin. Oncol. 2019, 16, 105–122. [Google Scholar] [CrossRef] [PubMed]
  24. Garcia-Gutierrez, V.; Hernandez-Boluda, J.C. Tyrosine Kinase Inhibitors Available for Chronic Myeloid Leukemia: Efficacy and Safety. Front. Oncol. 2019, 9, 603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Pathak, M.; Dwivedi, S.N.; Deo, S.V.S.; Thakur, B.; Sreenivas, V.; Rath, G.K. Effectiveness of Added Targeted Therapies to Neoadjuvant Chemotherapy for Breast Cancer: A Systematic Review and Meta-analysis. Clin. Breast Cancer 2019. [Google Scholar] [CrossRef] [PubMed]
  26. Valent, A.; Danglot, G.; Bernheim, A. Mapping of the tyrosine kinase receptors trkA (NTRK1), trkB (NTRK2) and trkC(NTRK3) to human chromosomes 1q22, 9q22 and 15q25 by fluorescence in situ hybridization. Eur. J. Hum. Genet. 1997, 5, 102–104. [Google Scholar] [CrossRef] [PubMed]
  27. Pulciani, S.; Santos, E.; Lauver, A.V.; Long, L.K.; Aaronson, S.A.; Barbacid, M. Oncogenes in solid human tumours. Nature 1982, 300, 539–542. [Google Scholar] [CrossRef] [PubMed]
  28. Martin-Zanca, D.; Oskam, R.; Mitra, G.; Copeland, T.; Barbacid, M. Molecular and biochemical characterization of the human trk proto-oncogene. Mol. Cell Biol. 1989, 9, 24–33. [Google Scholar] [CrossRef] [Green Version]
  29. Klein, R.; Jing, S.Q.; Nanduri, V.; O’Rourke, E.; Barbacid, M. The trk proto-oncogene encodes a receptor for nerve growth factor. Cell 1991, 65, 189–197. [Google Scholar] [CrossRef]
  30. Soppet, D.; Escandon, E.; Maragos, J.; Middlemas, D.S.; Reid, S.W.; Blair, J.; Burton, L.E.; Stanton, B.R.; Kaplan, D.R.; Hunter, T.; et al. The neurotrophic factors brain-derived neurotrophic factor and neurotrophin-3 are ligands for the trkB tyrosine kinase receptor. Cell 1991, 65, 895–903. [Google Scholar] [CrossRef]
  31. Davies, A.M.; Horton, A.; Burton, L.E.; Schmelzer, C.; Vandlen, R.; Rosenthal, A. Neurotrophin-4/5 is a mammalian-specific survival factor for distinct populations of sensory neurons. J. Neurosci. 1993, 13, 4961–4967. [Google Scholar] [CrossRef]
  32. Dechant, G.; Biffo, S.; Okazawa, H.; Kolbeck, R.; Pottgiesser, J.; Barde, Y.A. Expression and binding characteristics of the BDNF receptor chick trkB. Development 1993, 119, 545–558. [Google Scholar] [PubMed]
  33. Chao, M.V. Neurotrophins and their receptors: A convergence point for many signalling pathways. Nat. Rev. Neurosci. 2003, 4, 299–309. [Google Scholar] [CrossRef] [PubMed]
  34. Deinhardt, K.; Chao, M.V. Trk receptors. Handb. Exp. Pharmacol. 2014, 220, 103–119. [Google Scholar] [CrossRef] [PubMed]
  35. Marchetti, L.; Bonsignore, F.; Gobbo, F.; Amodeo, R.; Calvello, M.; Jacob, A.; Signore, G.; Schirripa Spagnolo, C.; Porciani, D.; Mainardi, M.; et al. Fast-diffusing p75(NTR) monomers support apoptosis and growth cone collapse by neurotrophin ligands. Proc. Natl. Acad. Sci. USA 2019, 116, 21563–21572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Mahadeo, D.; Kaplan, L.; Chao, M.V.; Hempstead, B.L. High affinity nerve growth factor binding displays a faster rate of association than p140trk binding. Implications for multi-subunit polypeptide receptors. J. Biol. Chem. 1994, 269, 6884–6891. [Google Scholar]
  37. Saadipour, K.; MacLean, M.; Pirkle, S.; Ali, S.; Lopez-Redondo, M.L.; Stokes, D.L.; Chao, M.V. The transmembrane domain of the p75 neurotrophin receptor stimulates phosphorylation of the TrkB tyrosine kinase receptor. J. Biol. Chem. 2017, 292, 16594–16604. [Google Scholar] [CrossRef] [Green Version]
  38. Vilar, M. Structural Characterization of the p75 Neurotrophin Receptor: A Stranger in the TNFR Superfamily. Vitam. Horm. 2017, 104, 57–87. [Google Scholar] [CrossRef]
  39. Alshehri, M.M.; Robbins, S.M.; Senger, D.L. The Role of Neurotrophin Signaling in Gliomagenesis: A Focus on the p75 Neurotrophin Receptor (p75(NTR)/CD271). Vitam. Horm. 2017, 104, 367–404. [Google Scholar] [CrossRef]
  40. Vaishnavi, A.; Le, A.T.; Doebele, R.C. TRKing down an old oncogene in a new era of targeted therapy. Cancer Discov. 2015, 5, 25–34. [Google Scholar] [CrossRef] [Green Version]
  41. Cocco, E.; Scaltriti, M.; Drilon, A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat. Rev. Clin. Oncol. 2018, 15, 731–747. [Google Scholar] [CrossRef]
  42. Yamashita, N.; Kuruvilla, R. Neurotrophin signaling endosomes: Biogenesis, regulation, and functions. Curr. Opin. Neurobiol. 2016, 39, 139–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Barford, K.; Deppmann, C.; Winckler, B. The neurotrophin receptor signaling endosome: Where trafficking meets signaling. Dev. Neurobiol. 2017, 77, 405–418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Clary, D.O.; Reichardt, L.F. An alternatively spliced form of the nerve growth factor receptor TrkA confers an enhanced response to neurotrophin 3. Proc. Natl. Acad. Sci. USA 1994, 91, 11133–11137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Huang, E.J.; Reichardt, L.F. Trk receptors: Roles in neuronal signal transduction. Annu. Rev. Biochem. 2003, 72, 609–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Luberg, K.; Wong, J.; Weickert, C.S.; Timmusk, T. Human TrkB gene: Novel alternative transcripts, protein isoforms and expression pattern in the prefrontal cerebral cortex during postnatal development. J. Neurochem. 2010, 113, 952–964. [Google Scholar] [CrossRef] [PubMed]
  47. Stoilov, P.; Castren, E.; Stamm, S. Analysis of the human TrkB gene genomic organization reveals novel TrkB isoforms, unusual gene length, and splicing mechanism. Biochem. Biophys. Res. Commun. 2002, 290, 1054–1065. [Google Scholar] [CrossRef] [Green Version]
  48. Brodeur, G.M.; Minturn, J.E.; Ho, R.; Simpson, A.M.; Iyer, R.; Varela, C.R.; Light, J.E.; Kolla, V.; Evans, A.E. Trk receptor expression and inhibition in neuroblastomas. Clin. Cancer Res. 2009, 15, 3244–3250. [Google Scholar] [CrossRef] [Green Version]
  49. Tacconelli, A.; Farina, A.R.; Cappabianca, L.; Desantis, G.; Tessitore, A.; Vetuschi, A.; Sferra, R.; Rucci, N.; Argenti, B.; Screpanti, I.; et al. TrkA alternative splicing: A regulated tumor-promoting switch in human neuroblastoma. Cancer Cell 2004, 6, 347–360. [Google Scholar] [CrossRef] [Green Version]
  50. Tacconelli, A.; Farina, A.R.; Cappabianca, L.; Gulino, A.; Mackay, A.R. Alternative TrkAIII splicing: A potential regulated tumor-promoting switch and therapeutic target in neuroblastoma. Future Oncol. 2005, 1, 689–698. [Google Scholar] [CrossRef]
  51. Snider, W.D. Functions of the neurotrophins during nervous system development: What the knockouts are teaching us. Cell 1994, 77, 627–638. [Google Scholar] [CrossRef]
  52. Bibel, M.; Barde, Y.A. Neurotrophins: Key regulators of cell fate and cell shape in the vertebrate nervous system. Genes Dev. 2000, 14, 2919–2937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Minichiello, L.; Korte, M.; Wolfer, D.; Kuhn, R.; Unsicker, K.; Cestari, V.; Rossi-Arnaud, C.; Lipp, H.P.; Bonhoeffer, T.; Klein, R. Essential role for TrkB receptors in hippocampus-mediated learning. Neuron 1999, 24, 401–414. [Google Scholar] [CrossRef] [Green Version]
  54. Schropel, A.; von Schack, D.; Dechant, G.; Barde, Y.A. Early expression of the nerve growth factor receptor ctrkA in chick sympathetic and sensory ganglia. Mol. Cell Neurosci. 1995, 6, 544–566. [Google Scholar] [CrossRef] [PubMed]
  55. Minichiello, L.; Klein, R. TrkB and TrkC neurotrophin receptors cooperate in promoting survival of hippocampal and cerebellar granule neurons. Genes Dev. 1996, 10, 2849–2858. [Google Scholar] [CrossRef] [Green Version]
  56. Tchetchelnitski, V.; van den Eijnden, M.; Schmidt, F.; Stoker, A.W. Developmental co-expression and functional redundancy of tyrosine phosphatases with neurotrophin receptors in developing sensory neurons. Int. J. Dev. Neurosci. 2014, 34, 48–59. [Google Scholar] [CrossRef]
  57. Ito, A.; Nosrat, C.A. Gustatory papillae and taste bud development and maintenance in the absence of TrkB ligands BDNF and NT-4. Cell Tissue Res. 2009, 337, 349–359. [Google Scholar] [CrossRef]
  58. Nittoli, V.; Sepe, R.M.; Coppola, U.; D’Agostino, Y.; De Felice, E.; Palladino, A.; Vassalli, Q.A.; Locascio, A.; Ristoratore, F.; Spagnuolo, A.; et al. A comprehensive analysis of neurotrophins and neurotrophin tyrosine kinase receptors expression during development of zebrafish. J. Comp. Neurol. 2018, 526, 1057–1072. [Google Scholar] [CrossRef]
  59. Minichiello, L.; Calella, A.M.; Medina, D.L.; Bonhoeffer, T.; Klein, R.; Korte, M. Mechanism of TrkB-mediated hippocampal long-term potentiation. Neuron 2002, 36, 121–137. [Google Scholar] [CrossRef] [Green Version]
  60. Gorski, J.A.; Zeiler, S.R.; Tamowski, S.; Jones, K.R. Brain-derived neurotrophic factor is required for the maintenance of cortical dendrites. J. Neurosci. 2003, 23, 6856–6865. [Google Scholar] [CrossRef]
  61. Medina, D.L.; Sciarretta, C.; Calella, A.M.; Von Bohlen Und Halbach, O.; Unsicker, K.; Minichiello, L. TrkB regulates neocortex formation through the Shc/PLCgamma-mediated control of neuronal migration. EMBO J. 2004, 23, 3803–3814. [Google Scholar] [CrossRef] [Green Version]
  62. Calella, A.M.; Nerlov, C.; Lopez, R.G.; Sciarretta, C.; von Bohlen und Halbach, O.; Bereshchenko, O.; Minichiello, L. Neurotrophin/Trk receptor signaling mediates C/EBPalpha, -beta and NeuroD recruitment to immediate-early gene promoters in neuronal cells and requires C/EBPs to induce immediate-early gene transcription. Neural Dev. 2007, 2, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Farhang, S.; Barar, J.; Fakhari, A.; Mesgariabbasi, M.; Khani, S.; Omidi, Y.; Farnam, A. Asymmetrical expression of BDNF and NTRK3 genes in frontoparietal cortex of stress-resilient rats in an animal model of depression. Synapse 2014, 68, 387–393. [Google Scholar] [CrossRef] [PubMed]
  64. Chen, Z.; Simmons, M.S.; Perry, R.T.; Wiener, H.W.; Harrell, L.E.; Go, R.C. Genetic association of neurotrophic tyrosine kinase receptor type 2 (NTRK2) With Alzheimer’s disease. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2008, 147, 363–369. [Google Scholar] [CrossRef] [PubMed]
  65. Weickert, C.S.; Ligons, D.L.; Romanczyk, T.; Ungaro, G.; Hyde, T.M.; Herman, M.M.; Weinberger, D.R.; Kleinman, J.E. Reductions in neurotrophin receptor mRNAs in the prefrontal cortex of patients with schizophrenia. Mol. Psychiatry 2005, 10, 637–650. [Google Scholar] [CrossRef]
  66. Otnaess, M.K.; Djurovic, S.; Rimol, L.M.; Kulle, B.; Kahler, A.K.; Jonsson, E.G.; Agartz, I.; Sundet, K.; Hall, H.; Timm, S.; et al. Evidence for a possible association of neurotrophin receptor (NTRK-3) gene polymorphisms with hippocampal function and schizophrenia. Neurobiol. Dis. 2009, 34, 518–524. [Google Scholar] [CrossRef]
  67. Autry, A.E.; Monteggia, L.M. Brain-derived neurotrophic factor and neuropsychiatric disorders. Pharmacol. Rev. 2012, 64, 238–258. [Google Scholar] [CrossRef] [Green Version]
  68. Boulle, F.; Kenis, G.; Cazorla, M.; Hamon, M.; Steinbusch, H.W.; Lanfumey, L.; van den Hove, D.L. TrkB inhibition as a therapeutic target for CNS-related disorders. Prog. Neurobiol. 2012, 98, 197–206. [Google Scholar] [CrossRef]
  69. Drilon, A.; Siena, S.; Ou, S.I.; Patel, M.; Ahn, M.J.; Lee, J.; Bauer, T.M.; Farago, A.F.; Wheler, J.J.; Liu, S.V.; et al. Safety and Antitumor Activity of the Multitargeted Pan-TRK, ROS1, and ALK Inhibitor Entrectinib: Combined Results from Two Phase I Trials (ALKA-372-001 and STARTRK-1). Cancer Discov. 2017, 7, 400–409. [Google Scholar] [CrossRef] [Green Version]
  70. Okamura, R.; Boichard, A.; Kato, S.; Sicklick, J.K.; Bazhenova, L.; Kurzrock, R. Analysis of NTRK Alterations in Pan-Cancer Adult and Pediatric Malignancies: Implications for NTRK-Targeted Therapeutics. JCO Precis. Oncol. 2018, 2018. [Google Scholar] [CrossRef]
  71. Shaw, A.T.; Hsu, P.P.; Awad, M.M.; Engelman, J.A. Tyrosine kinase gene rearrangements in epithelial malignancies. Nat. Rev. Cancer 2013, 13, 772–787. [Google Scholar] [CrossRef]
  72. Stransky, N.; Cerami, E.; Schalm, S.; Kim, J.L.; Lengauer, C. The landscape of kinase fusions in cancer. Nat. Commun. 2014, 5, 4846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Gross, S.; Rahal, R.; Stransky, N.; Lengauer, C.; Hoeflich, K.P. Targeting cancer with kinase inhibitors. J. Clin. Invest. 2015, 125, 1780–1789. [Google Scholar] [CrossRef] [PubMed]
  74. Khotskaya, Y.B.; Holla, V.R.; Farago, A.F.; Mills Shaw, K.R.; Meric-Bernstam, F.; Hong, D.S. Targeting TRK family proteins in cancer. Pharmacol. Ther. 2017, 173, 58–66. [Google Scholar] [CrossRef] [PubMed]
  75. Farago, A.F.; Azzoli, C.G. Beyond ALK and ROS1: RET, NTRK, EGFR and BRAF gene rearrangements in non-small cell lung cancer. Transl. Lung Cancer Res. 2017, 6, 550–559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Tognon, C.; Knezevich, S.R.; Huntsman, D.; Roskelley, C.D.; Melnyk, N.; Mathers, J.A.; Becker, L.; Carneiro, F.; MacPherson, N.; Horsman, D.; et al. Expression of the ETV6-NTRK3 gene fusion as a primary event in human secretory breast carcinoma. Cancer Cell 2002, 2, 367–376. [Google Scholar] [CrossRef] [Green Version]
  77. Hung, Y.P.; Fletcher, C.D.M.; Hornick, J.L. Evaluation of pan-TRK immunohistochemistry in infantile fibrosarcoma, lipofibromatosis-like neural tumour and histological mimics. Histopathology 2018, 73, 634–644. [Google Scholar] [CrossRef] [PubMed]
  78. Hung, Y.P.; Jo, V.Y.; Hornick, J.L. Immunohistochemistry with a pan-TRK antibody distinguishes secretory carcinoma of the salivary gland from acinic cell carcinoma. Histopathology 2019, 75, 54–62. [Google Scholar] [CrossRef] [PubMed]
  79. Xu, B.; Haroon Al Rasheed, M.R.; Antonescu, C.R.; Alex, D.; Frosina, D.; Ghossein, R.; Jungbluth, A.A.; Katabi, N. Pan-Trk immunohistochemistry is a sensitive and specific ancillary tool in diagnosing secretory carcinoma of salivary gland and detecting ETV6-NTRK3 fusion. Histopathology 2019. [Google Scholar] [CrossRef]
  80. Harrison, B.T.; Fowler, E.; Krings, G.; Chen, Y.Y.; Bean, G.R.; Vincent-Salomon, A.; Fuhrmann, L.; Barnick, S.E.; Chen, B.; Hosfield, E.M.; et al. Pan-TRK Immunohistochemistry: A Useful Diagnostic Adjunct for Secretory Carcinoma of the Breast. Am. J. Surg. Pathol. 2019. [Google Scholar] [CrossRef]
  81. Gao, Q.; Liang, W.W.; Foltz, S.M.; Mutharasu, G.; Jayasinghe, R.G.; Cao, S.; Liao, W.W.; Reynolds, S.M.; Wyczalkowski, M.A.; Yao, L.; et al. Driver Fusions and Their Implications in the Development and Treatment of Human Cancers. Cell Rep. 2018, 23, 227–238 e3. [Google Scholar] [CrossRef] [Green Version]
  82. Reuther, G.W.; Lambert, Q.T.; Caligiuri, M.A.; Der, C.J. Identification and characterization of an activating TrkA deletion mutation in acute myeloid leukemia. Mol. Cell. Biol. 2000, 20, 8655–8666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Thiele, C.J.; Li, Z.; McKee, A.E. On Trk--the TrkB signal transduction pathway is an increasingly important target in cancer biology. Clin. Cancer Res. 2009, 15, 5962–5967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Light, J.E.; Koyama, H.; Minturn, J.E.; Ho, R.; Simpson, A.M.; Iyer, R.; Mangino, J.L.; Kolla, V.; London, W.B.; Brodeur, G.M. Clinical significance of NTRK family gene expression in neuroblastomas. Pediatr. Blood Cancer 2012, 59, 226–232. [Google Scholar] [CrossRef] [Green Version]
  85. Lee, S.J.; Kim, N.K.D.; Lee, S.-H.; Kim, S.T.; Park, S.H.; Park, J.O.; Park, Y.S.; Lim, H.Y.; Kang, W.K.; Park, W.Y.; et al. NTRK gene amplification in patients with metastatic cancer. Precis Future Med. 2017, 1, 129–137. [Google Scholar] [CrossRef] [Green Version]
  86. Farina, A.R.; Cappabianca, L.; Ruggeri, P.; Gneo, L.; Pellegrini, C.; Fargnoli, M.C.; Mackay, A.R. The oncogenic neurotrophin receptor tropomyosin-related kinase variant, TrkAIII. J. Exp. Clin. Cancer Res. 2018, 37, 119. [Google Scholar] [CrossRef] [PubMed]
  87. Fuse, M.J.; Okada, K.; Oh-Hara, T.; Ogura, H.; Fujita, N.; Katayama, R. Mechanisms of Resistance to NTRK Inhibitors and Therapeutic Strategies in NTRK1-Rearranged Cancers. Mol. Cancer Ther. 2017, 16, 2130–2143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Martin-Zanca, D.; Hughes, S.H.; Barbacid, M. A human oncogene formed by the fusion of truncated tropomyosin and protein tyrosine kinase sequences. Nature 1986, 319, 743–748. [Google Scholar] [CrossRef]
  89. Ardini, E.; Bosotti, R.; Borgia, A.L.; De Ponti, C.; Somaschini, A.; Cammarota, R.; Amboldi, N.; Raddrizzani, L.; Milani, A.; Magnaghi, P.; et al. The TPM3-NTRK1 rearrangement is a recurring event in colorectal carcinoma and is associated with tumor sensitivity to TRKA kinase inhibition. Mol. Oncol. 2014, 8, 1495–1507. [Google Scholar] [CrossRef]
  90. Creancier, L.; Vandenberghe, I.; Gomes, B.; Dejean, C.; Blanchet, J.C.; Meilleroux, J.; Guimbaud, R.; Selves, J.; Kruczynski, A. Chromosomal rearrangements involving the NTRK1 gene in colorectal carcinoma. Cancer Lett. 2015, 365, 107–111. [Google Scholar] [CrossRef]
  91. Lee, S.J.; Li, G.G.; Kim, S.T.; Hong, M.E.; Jang, J.; Yoon, N.; Ahn, S.M.; Murphy, D.; Christiansen, J.; Wei, G.; et al. NTRK1 rearrangement in colorectal cancer patients: Evidence for actionable target using patient-derived tumor cell line. Oncotarget 2015, 6, 39028–39035. [Google Scholar] [CrossRef] [Green Version]
  92. Sartore-Bianchi, A.; Ardini, E.; Bosotti, R.; Amatu, A.; Valtorta, E.; Somaschini, A.; Raddrizzani, L.; Palmeri, L.; Banfi, P.; Bonazzina, E.; et al. Sensitivity to Entrectinib Associated With a Novel LMNA-NTRK1 Gene Fusion in Metastatic Colorectal Cancer. J. Natl. Cancer Inst. 2016, 108. [Google Scholar] [CrossRef]
  93. Hechtman, J.F.; Zehir, A.; Yaeger, R.; Wang, L.; Middha, S.; Zheng, T.; Hyman, D.M.; Solit, D.; Arcila, M.E.; Borsu, L.; et al. Identification of Targetable Kinase Alterations in Patients with Colorectal Carcinoma That are Preferentially Associated with Wild-Type RAS/RAF. Mol. Cancer Res. 2016, 14, 296–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Pietrantonio, F.; Di Nicolantonio, F.; Schrock, A.B.; Lee, J.; Tejpar, S.; Sartore-Bianchi, A.; Hechtman, J.F.; Christiansen, J.; Novara, L.; Tebbutt, N.; et al. ALK, ROS1, and NTRK Rearrangements in Metastatic Colorectal Cancer. J. Natl. Cancer Inst. 2017, 109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Vaishnavi, A.; Capelletti, M.; Le, A.T.; Kako, S.; Butaney, M.; Ercan, D.; Mahale, S.; Davies, K.D.; Aisner, D.L.; Pilling, A.B.; et al. Oncogenic and drug-sensitive NTRK1 rearrangements in lung cancer. Nat. Med. 2013, 19, 1469–1472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Bongarzone, I.; Pierotti, M.A.; Monzini, N.; Mondellini, P.; Manenti, G.; Donghi, R.; Pilotti, S.; Grieco, M.; Santoro, M.; Fusco, A.; et al. High frequency of activation of tyrosine kinase oncogenes in human papillary thyroid carcinoma. Oncogene 1989, 4, 1457–1462. [Google Scholar] [PubMed]
  97. Butti, M.G.; Bongarzone, I.; Ferraresi, G.; Mondellini, P.; Borrello, M.G.; Pierotti, M.A. A sequence analysis of the genomic regions involved in the rearrangements between TPM3 and NTRK1 genes producing TRK oncogenes in papillary thyroid carcinomas. Genomics 1995, 28, 15–24. [Google Scholar] [CrossRef] [PubMed]
  98. Leeman-Neill, R.J.; Kelly, L.M.; Liu, P.; Brenner, A.V.; Little, M.P.; Bogdanova, T.I.; Evdokimova, V.N.; Hatch, M.; Zurnadzy, L.Y.; Nikiforova, M.N.; et al. ETV6-NTRK3 is a common chromosomal rearrangement in radiation-associated thyroid cancer. Cancer 2014, 120, 799–807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Knezevich, S.R.; Garnett, M.J.; Pysher, T.J.; Beckwith, J.B.; Grundy, P.E.; Sorensen, P.H. ETV6-NTRK3 gene fusions and trisomy 11 establish a histogenetic link between mesoblastic nephroma and congenital fibrosarcoma. Cancer Res. 1998, 58, 5046–5048. [Google Scholar]
  100. Knezevich, S.R.; McFadden, D.E.; Tao, W.; Lim, J.F.; Sorensen, P.H. A novel ETV6-NTRK3 gene fusion in congenital fibrosarcoma. Nat. Genet. 1998, 18, 184–187. [Google Scholar] [CrossRef]
  101. Skalova, A.; Vanecek, T.; Sima, R.; Laco, J.; Weinreb, I.; Perez-Ordonez, B.; Starek, I.; Geierova, M.; Simpson, R.H.; Passador-Santos, F.; 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, 599–608. [Google Scholar] [CrossRef]
  102. Lannon, C.L.; Sorensen, P.H. ETV6-NTRK3: A chimeric protein tyrosine kinase with transformation activity in multiple cell lineages. Semin. Cancer Biol. 2005, 15, 215–223. [Google Scholar] [CrossRef] [PubMed]
  103. Yeh, I.; Tee, M.K.; Botton, T.; Shain, A.H.; Sparatta, A.J.; Gagnon, A.; Vemula, S.S.; Garrido, M.C.; Nakamaru, K.; Isoyama, T.; et al. NTRK3 kinase fusions in Spitz tumours. J. Pathol. 2016, 240, 282–290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Johnson, M.D.; Stone, B.; Thibodeau, B.J.; Baschnagel, A.M.; Galoforo, S.; Fortier, L.E.; Ketelsen, B.; Ahmed, S.; Kelley, Z.; Hana, A.; et al. The significance of Trk receptors in pancreatic cancer. Tumour. Biol. 2017, 39, 1010428317692256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Sigal, D.; Tartar, M.; Xavier, M.; Bao, F.; Foley, P.; Luo, D.; Christiansen, J.; Hornby, Z.; Maneval, E.C.; Multani, P. Activity of Entrectinib in a Patient With the First Reported NTRK Fusion in Neuroendocrine Cancer. J. Natl. Compr. Canc. Netw. 2017, 15, 1317–1322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Rudzinski, E.R.; Lockwood, C.M.; Stohr, B.A.; Vargas, S.O.; Sheridan, R.; Black, J.O.; Rajaram, V.; Laetsch, T.W.; Davis, J.L. Pan-Trk Immunohistochemistry Identifies NTRK Rearrangements in Pediatric Mesenchymal Tumors. Am. J. Surg. Pathol. 2018, 42, 927–935. [Google Scholar] [CrossRef] [PubMed]
  107. Sigal, D.S.; Bhangoo, M.S.; Hermel, J.A.; Pavlick, D.C.; Frampton, G.; Miller, V.A.; Ross, J.S.; Ali, S.M. Comprehensive genomic profiling identifies novel NTRK fusions in neuroendocrine tumors. Oncotarget 2018, 9, 35809–35812. [Google Scholar] [CrossRef] [Green Version]
  108. Lezcano, C.; Shoushtari, A.N.; Ariyan, C.; Hollmann, T.J.; Busam, K.J. Primary and Metastatic Melanoma With NTRK Fusions. Am. J. Surg. Pathol. 2018, 42, 1052–1058. [Google Scholar] [CrossRef]
  109. Miettinen, M.; Felisiak-Golabek, A.; Luina Contreras, A.; Glod, J.; Kaplan, R.N.; Killian, J.K.; Lasota, J. New fusion sarcomas: Histopathology and clinical significance of selected entities. Hum. Pathol. 2019, 86, 57–65. [Google Scholar] [CrossRef]
  110. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  111. Ostrom, Q.T.; Cioffi, G.; Gittleman, H.; Patil, N.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2012-2016. Neuro. Oncol. 2019, 21, v1–v100. [Google Scholar] [CrossRef]
  112. Zhang, J.; Wu, G.; Miller, C.P.; Tatevossian, R.G.; Dalton, J.D.; Tang, B.; Orisme, W.; Punchihewa, C.; Parker, M.; Qaddoumi, I.; et al. Whole-genome sequencing identifies genetic alterations in pediatric low-grade gliomas. Nat. Genet. 2013, 45, 602–612. [Google Scholar] [CrossRef] [PubMed]
  113. Lake, J.A.; Donson, A.M.; Prince, E.; Davies, K.D.; Nellan, A.; Green, A.L.; Mulcahy Levy, J.; Dorris, K.; Vibhakar, R.; Hankinson, T.C.; et al. Targeted fusion analysis can aid in the classification and treatment of pediatric glioma, ependymoma, and glioneuronal tumors. Pediatr. Blood Cancer 2020, 67, e28028. [Google Scholar] [CrossRef] [PubMed]
  114. Jones, D.T.; Hutter, B.; Jager, N.; Korshunov, A.; Kool, M.; Warnatz, H.J.; Zichner, T.; Lambert, S.R.; Ryzhova, M.; Quang, D.A.; et al. Recurrent somatic alterations of FGFR1 and NTRK2 in pilocytic astrocytoma. Nat. Genet. 2013, 45, 927–932. [Google Scholar] [CrossRef] [Green Version]
  115. Chen, C.; Han, S.; Meng, L.; Li, Z.; Zhang, X.; Wu, A. TERT promoter mutations lead to high transcriptional activity under hypoxia and temozolomide treatment and predict poor prognosis in gliomas. PLoS One 2014, 9, e100297. [Google Scholar] [CrossRef] [Green Version]
  116. Lassaletta, A.; Zapotocky, M.; Bouffet, E.; Hawkins, C.; Tabori, U. An integrative molecular and genomic analysis of pediatric hemispheric low-grade gliomas: An update. Childs Nerv. Syst. 2016, 32, 1789–1797. [Google Scholar] [CrossRef] [PubMed]
  117. Vanan, M.I.; Underhill, D.A.; Eisenstat, D.D. Targeting Epigenetic Pathways in the Treatment of Pediatric Diffuse (High Grade) Gliomas. Neurotherapeutics 2017, 14, 274–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Bornhorst, M.; Hwang, E.I. Molecularly Targeted Agents in the Therapy of Pediatric Brain Tumors. Paediatr. Drugs 2019. [Google Scholar] [CrossRef]
  119. Collins, V.P.; Jones, D.T.; Giannini, C. Pilocytic astrocytoma: Pathology, molecular mechanisms and markers. Acta Neuropathol. 2015, 129, 775–788. [Google Scholar] [CrossRef] [Green Version]
  120. Nobusawa, S.; Hirato, J.; Yokoo, H. Molecular genetics of ependymomas and pediatric diffuse gliomas: A short review. Brain Tumor Pathol. 2014, 31, 229–233. [Google Scholar] [CrossRef]
  121. Wu, G.; Diaz, A.K.; Paugh, B.S.; Rankin, S.L.; Ju, B.; Li, Y.; Zhu, X.; Qu, C.; Chen, X.; Zhang, J.; et al. The genomic landscape of diffuse intrinsic pontine glioma and pediatric non-brainstem high-grade glioma. Nat. Genet. 2014, 46, 444–450. [Google Scholar] [CrossRef]
  122. Chamdine, O.; Gajjar, A. Molecular characteristics of pediatric high-grade gliomas. CNS Oncol. 2014, 3, 433–443. [Google Scholar] [CrossRef] [PubMed]
  123. Albert, C.M.; Davis, J.L.; Federman, N.; Casanova, M.; Laetsch, T.W. TRK Fusion Cancers in Children: A Clinical Review and Recommendations for Screening. J. Clin. Oncol. 2019, 37, 513–524. [Google Scholar] [CrossRef] [PubMed]
  124. Zheng, Z.; Liebers, M.; Zhelyazkova, B.; Cao, Y.; Panditi, D.; Lynch, K.D.; Chen, J.; Robinson, H.E.; Shim, H.S.; Chmielecki, J.; et al. Anchored multiplex PCR for targeted next-generation sequencing. Nat. Med. 2014, 20, 1479–1484. [Google Scholar] [CrossRef] [PubMed]
  125. Deng, M.Y.; Sill, M.; Chiang, J.; Schittenhelm, J.; Ebinger, M.; Schuhmann, M.U.; Monoranu, C.M.; Milde, T.; Wittmann, A.; Hartmann, C.; et al. Molecularly defined diffuse leptomeningeal glioneuronal tumor (DLGNT) comprises two subgroups with distinct clinical and genetic features. Acta Neuropathol. 2018, 136, 239–253. [Google Scholar] [CrossRef] [Green Version]
  126. Prabhakaran, N.; Guzman, M.A.; Navalkele, P.; Chow-Maneval, E.; Batanian, J.R. Novel TLE4-NTRK2 fusion in a ganglioglioma identified by array-CGH and confirmed by NGS: Potential for a gene targeted therapy. Neuropathology 2018. [Google Scholar] [CrossRef]
  127. Kurozumi, K.; Nakano, Y.; Ishida, J.; Tanaka, T.; Doi, M.; Hirato, J.; Yoshida, A.; Washio, K.; Shimada, A.; Kohno, T.; et al. High-grade glioneuronal tumor with an ARHGEF2-NTRK1 fusion gene. Brain Tumor Pathol. 2019, 36, 121–128. [Google Scholar] [CrossRef]
  128. Segal, R.A.; Goumnerova, L.C.; Kwon, Y.K.; Stiles, C.D.; Pomeroy, S.L. Expression of the neurotrophin receptor TrkC is linked to a favorable outcome in medulloblastoma. Proc. Natl. Acad. Sci. USA 1994, 91, 12867–12871. [Google Scholar] [CrossRef] [Green Version]
  129. Kim, J.Y.; Sutton, M.E.; Lu, D.J.; Cho, T.A.; Goumnerova, L.C.; Goritchenko, L.; Kaufman, J.R.; Lam, K.K.; Billet, A.L.; Tarbell, N.J.; et al. Activation of neurotrophin-3 receptor TrkC induces apoptosis in medulloblastomas. Cancer Res. 1999, 59, 711–719. [Google Scholar]
  130. Grotzer, M.A.; Janss, A.J.; Fung, K.; Biegel, J.A.; Sutton, L.N.; Rorke, L.B.; Zhao, H.; Cnaan, A.; Phillips, P.C.; Lee, V.M.; et al. TrkC expression predicts good clinical outcome in primitive neuroectodermal brain tumors. J. Clin. Oncol. 2000, 18, 1027–1035. [Google Scholar] [CrossRef]
  131. Brandes, A.A.; Franceschi, E. Shedding light on adult medulloblastoma: Current management and opportunities for advances. Am. Soc. Clin. Oncol. Educ. Book 2014, e82–e87. [Google Scholar] [CrossRef]
  132. Amatu, A.; Sartore-Bianchi, A.; Siena, S. NTRK gene fusions as novel targets of cancer therapy across multiple tumour types. ESMO Open 2016, 1, e000023. [Google Scholar] [CrossRef] [PubMed]
  133. Ferguson, S.D.; Zhou, S.; Huse, J.T.; de Groot, J.F.; Xiu, J.; Subramaniam, D.S.; Mehta, S.; Gatalica, Z.; Swensen, J.; Sanai, N.; et al. Targetable Gene Fusions Associate With the IDH Wild-Type Astrocytic Lineage in Adult Gliomas. J. Neuropathol. Exp. Neurol. 2018, 77, 437–442. [Google Scholar] [CrossRef] [Green Version]
  134. Frattini, V.; Trifonov, V.; Chan, J.M.; Castano, A.; Lia, M.; Abate, F.; Keir, S.T.; Ji, A.X.; Zoppoli, P.; Niola, F.; et al. The integrated landscape of driver genomic alterations in glioblastoma. Nat. Genet. 2013, 45, 1141–1149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Shah, N.; Lankerovich, M.; Lee, H.; Yoon, J.G.; Schroeder, B.; Foltz, G. Exploration of the gene fusion landscape of glioblastoma using transcriptome sequencing and copy number data. BMC Genomics 2013, 14, 818. [Google Scholar] [CrossRef] [Green Version]
  136. Kim, J.; Lee, Y.; Cho, H.J.; Lee, Y.E.; An, J.; Cho, G.H.; Ko, Y.H.; Joo, K.M.; Nam, D.H. NTRK1 fusion in glioblastoma multiforme. PLoS One 2014, 9, e91940. [Google Scholar] [CrossRef] [Green Version]
  137. Cook, P.J.; Thomas, R.; Kannan, R.; de Leon, E.S.; Drilon, A.; Rosenblum, M.K.; Scaltriti, M.; Benezra, R.; Ventura, A. Somatic chromosomal engineering identifies BCAN-NTRK1 as a potent glioma driver and therapeutic target. Nat. Commun. 2017, 8, 15987. [Google Scholar] [CrossRef] [PubMed]
  138. Assimakopoulou, M.; Kondyli, M.; Gatzounis, G.; Maraziotis, T.; Varakis, J. Neurotrophin receptors expression and JNK pathway activation in human astrocytomas. BMC Cancer 2007, 7, 202. [Google Scholar] [CrossRef] [Green Version]
  139. Palani, M.; Arunkumar, R.; Vanisree, A.J. Methylation and expression patterns of tropomyosin-related kinase genes in different grades of glioma. Neuromolecular Med. 2014, 16, 529–539. [Google Scholar] [CrossRef]
  140. Pajtler, K.W.; Rebmann, V.; Lindemann, M.; Schulte, J.H.; Schulte, S.; Stauder, M.; Leuschner, I.; Schmid, K.W.; Kohl, U.; Schramm, A.; et al. Expression of NTRK1/TrkA affects immunogenicity of neuroblastoma cells. Int. J. Cancer 2013, 133, 908–919. [Google Scholar] [CrossRef]
  141. Offin, M.; Liu, D.; Drilon, A. Tumor-Agnostic Drug Development. Am. Soc. Clin. Oncol. Educ. Book 2018, 38, 184–187. [Google Scholar] [CrossRef]
  142. Liu, D.; Offin, M.; Harnicar, S.; Li, B.T.; Drilon, A. Entrectinib: An orally available, selective tyrosine kinase inhibitor for the treatment of NTRK, ROS1, and ALK fusion-positive solid tumors. Ther. Clin. Risk Manag. 2018, 14, 1247–1252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Ardini, E.; Menichincheri, M.; Banfi, P.; Bosotti, R.; De Ponti, C.; Pulci, R.; Ballinari, D.; Ciomei, M.; Texido, G.; Degrassi, A.; et al. Entrectinib, a Pan-TRK, ROS1, and ALK Inhibitor with Activity in Multiple Molecularly Defined Cancer Indications. Mol. Cancer Ther. 2016, 15, 628–639. [Google Scholar] [CrossRef] [Green Version]
  144. Drilon, A.; Laetsch, T.W.; Kummar, S.; DuBois, S.G.; Lassen, U.N.; Demetri, G.D.; Nathenson, M.; Doebele, R.C.; Farago, A.F.; Pappo, A.S.; et al. Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children. N. Engl. J. Med. 2018, 378, 731–739. [Google Scholar] [CrossRef] [PubMed]
  145. Laetsch, T.W.; DuBois, S.G.; Mascarenhas, L.; Turpin, B.; Federman, N.; Albert, C.M.; Nagasubramanian, R.; Davis, J.L.; Rudzinski, E.; Feraco, A.M.; et al. Larotrectinib for paediatric solid tumours harbouring NTRK gene fusions: Phase 1 results from a multicentre, open-label, phase 1/2 study. Lancet. Oncol. 2018, 19, 705–714. [Google Scholar] [CrossRef]
  146. Drilon, A.E.; DuBois, S.G.; Farago, A.F.; Geoerger, B.; Grilley-Olson, J.E.; Hong, D.S.; Sohal, D.; van Tilburg, C.M.; Ziegler, D.S.; Ku, N.; et al. Activity of larotrectinib in TRK fusion cancer patients with brain metastases or primary central nervous system tumors. J. Clin. Oncol. 2019, 37, 2006. [Google Scholar] [CrossRef]
  147. Drilon, A.; Nagasubramanian, R.; Blake, J.F.; Ku, N.; Tuch, B.B.; Ebata, K.; Smith, S.; Lauriault, V.; Kolakowski, G.R.; Brandhuber, B.J.; et al. A Next-Generation TRK Kinase Inhibitor Overcomes Acquired Resistance to Prior TRK Kinase Inhibition in Patients with TRK Fusion-Positive Solid Tumors. Cancer Discov. 2017, 7, 963–972. [Google Scholar] [CrossRef] [Green Version]
  148. Drilon, A.; Ou, S.I.; Cho, B.C.; Kim, D.W.; Lee, J.; Lin, J.J.; Zhu, V.W.; Ahn, M.J.; Camidge, D.R.; Nguyen, J.; et al. Repotrectinib (TPX-0005) Is a Next-Generation ROS1/TRK/ALK Inhibitor That Potently Inhibits ROS1/TRK/ALK Solvent- Front Mutations. Cancer Discov. 2018, 8, 1227–1236. [Google Scholar] [CrossRef] [Green Version]
  149. Ercan, D.; Choi, H.G.; Yun, C.H.; Capelletti, M.; Xie, T.; Eck, M.J.; Gray, N.S.; Janne, P.A. EGFR Mutations and Resistance to Irreversible Pyrimidine-Based EGFR Inhibitors. Clin. Cancer Res. 2015, 21, 3913–3923. [Google Scholar] [CrossRef] [Green Version]
  150. Gainor, J.F.; Dardaei, L.; Yoda, S.; Friboulet, L.; Leshchiner, I.; Katayama, R.; Dagogo-Jack, I.; Gadgeel, S.; Schultz, K.; Singh, M.; et al. Molecular Mechanisms of Resistance to First- and Second-Generation ALK Inhibitors in ALK-Rearranged Lung Cancer. Cancer Discov. 2016, 6, 1118–1133. [Google Scholar] [CrossRef] [Green Version]
  151. Bennouna, J.; Girard, N.; Audigier-Valette, C.; le Thuaut, A.; Gervais, R.; Masson, P.; Marcq, M.; Molinier, O.; Cortot, A.; Debieuvre, D.; et al. Phase II Study Evaluating the Mechanisms of Resistance on Tumor Tissue and Liquid Biopsy in Patients With EGFR-mutated Non-pretreated Advanced Lung Cancer Receiving Osimertinib Until and Beyond Radiologic Progression: The MELROSE Trial. Clin. Lung Cancer 2019. [Google Scholar] [CrossRef] [Green Version]
  152. Recondo, G.; Mezquita, L.; Facchinetti, F.; Planchard, D.; Gazzah, A.; Bigot, L.; Rizvi, A.Z.; Frias, R.L.; Thiery, J.P.; Scoazec, J.Y.; et al. Diverse resistance mechanisms to the third-generation ALK inhibitor lorlatinib in ALK-rearranged lung cancer. Clin. Cancer Res. 2019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Morris, T.A.; Khoo, C.; Solomon, B.J. Targeting ROS1 Rearrangements in Non-small Cell Lung Cancer: Crizotinib and Newer Generation Tyrosine Kinase Inhibitors. Drugs 2019, 79, 1277–1286. [Google Scholar] [CrossRef]
  154. Roys, A.; Chang, X.; Liu, Y.; Xu, X.; Wu, Y.; Zuo, D. Resistance mechanisms and potent-targeted therapies of ROS1-positive lung cancer. Cancer Chemother Pharmacol. 2019, 84, 679–688. [Google Scholar] [CrossRef]
  155. Ricciuti, B.; Genova, C.; Crino, L.; Libra, M.; Leonardi, G.C. Antitumor activity of larotrectinib in tumors harboring NTRK gene fusions: A short review on the current evidence. Onco. Targets Ther. 2019, 12, 3171–3179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Cocco, E.; Schram, A.M.; Kulick, A.; Misale, S.; Won, H.H.; Yaeger, R.; Razavi, P.; Ptashkin, R.; Hechtman, J.F.; Toska, E.; et al. Resistance to TRK inhibition mediated by convergent MAPK pathway activation. Nat. Med. 2019, 25, 1422–1427. [Google Scholar] [CrossRef] [PubMed]
  157. Kummar, S.; Lassen, U.N. TRK Inhibition: A New Tumor-Agnostic Treatment Strategy. Target. Oncol. 2018, 13, 545–556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Hechtman, J.F.; Benayed, R.; Hyman, D.M.; Drilon, A.; Zehir, A.; Frosina, D.; Arcila, M.E.; Dogan, S.; Klimstra, D.S.; Ladanyi, M.; et al. Pan-Trk Immunohistochemistry Is an Efficient and Reliable Screen for the Detection of NTRK Fusions. Am. J. Surg. Pathol. 2017, 41, 1547–1551. [Google Scholar] [CrossRef]
  159. Bourhis, A.; Redoulez, G.; Quintin-Roue, I.; Marcorelles, P.; Uguen, A. Screening for NTRK-rearranged Tumors Using Immunohistochemistry: Comparison of 2 Different pan-TRK Clones in Melanoma Samples. Appl. Immunohistochem. Mol. Morphol. 2019. [Google Scholar] [CrossRef]
  160. Solomon, J.P.; Linkov, I.; Rosado, A.; Mullaney, K.; Rosen, E.Y.; Frosina, D.; Jungbluth, A.A.; Zehir, A.; Benayed, R.; Drilon, A.; et al. NTRK fusion detection across multiple assays and 33,997 cases: Diagnostic implications and pitfalls. Mod. Pathol. 2019. [Google Scholar] [CrossRef]
  161. Solomon, J.P.; Hechtman, J.F. Detection of NTRK Fusions: Merits and Limitations of Current Diagnostic Platforms. Cancer Res. 2019, 79, 3163–3168. [Google Scholar] [CrossRef]
  162. Gatalica, Z.; Xiu, J.; Swensen, J.; Vranic, S. Molecular characterization of cancers with NTRK gene fusions. Mod. Pathol. 2019, 32, 147–153. [Google Scholar] [CrossRef] [PubMed]
  163. Murphy, D.A.; Ely, H.A.; Shoemaker, R.; Boomer, A.; Culver, B.P.; Hoskins, I.; Haimes, J.D.; Walters, R.D.; Fernandez, D.; Stahl, J.A.; et al. Detecting Gene Rearrangements in Patient Populations Through a 2-Step Diagnostic Test Comprised of Rapid IHC Enrichment Followed by Sensitive Next-Generation Sequencing. Appl. Immunohistochem. Mol. Morphol. 2017, 25, 513–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Marchio, C.; Scaltriti, M.; Ladanyi, M.; Iafrate, A.J.; Bibeau, F.; Dietel, M.; Hechtman, J.F.; Troiani, T.; Lopez-Rios, F.; Douillard, J.Y.; et al. ESMO recommendations on the standard methods to detect NTRK fusions in daily practice and clinical research. Ann. Oncol. 2019, 30, 1417–1427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Penault-Llorca, F.; Rudzinski, E.R.; Sepulveda, A.R. Testing algorithm for identification of patients with TRK fusion cancer. J. Clin. Pathol. 2019, 72, 460–467. [Google Scholar] [CrossRef] [PubMed]
  166. Hsiao, S.J.; Zehir, A.; Sireci, A.N.; Aisner, D.L. Detection of Tumor NTRK Gene Fusions to Identify Patients Who May Benefit from Tyrosine Kinase (TRK) Inhibitor Therapy. J. Mol. Diagn. 2019, 21, 553–571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Cocco, E.; Benhamida, J.; Middha, S.; Zehir, A.; Mullaney, K.; Shia, J.; Yaeger, R.; Zhang, L.; Wong, D.; Villafania, L.; et al. Colorectal Carcinomas Containing Hypermethylated MLH1 Promoter and Wild-Type BRAF/KRAS Are Enriched for Targetable Kinase Fusions. Cancer Res. 2019, 79, 1047–1053. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Physiological and rearranged NTRK genes/TRK receptors and intracellular signaling. The PLC-γ, MAPK, and PI3-K intracellular pathways (here represented by the DAG/IP3, RAS/MEK/ERK, and PI3-K/AKT components, respectively) are activated either from the wild-type form of NTRK, and the chimeric fusion receptors (e.g., BCAN-NTRK1 and ETV6-NTRK3). However, the latter happens in a ligand-free constitutively activated fashion, leading to oncogenic activation. The NTRK inhibitors (TKI, here represented by entrectinib and larotrectinib) achieve their antitumor activity by interacting with the intracellular domain of the chimeric receptors, inhibiting the recruitment of the signaling pathway.
Figure 1. Physiological and rearranged NTRK genes/TRK receptors and intracellular signaling. The PLC-γ, MAPK, and PI3-K intracellular pathways (here represented by the DAG/IP3, RAS/MEK/ERK, and PI3-K/AKT components, respectively) are activated either from the wild-type form of NTRK, and the chimeric fusion receptors (e.g., BCAN-NTRK1 and ETV6-NTRK3). However, the latter happens in a ligand-free constitutively activated fashion, leading to oncogenic activation. The NTRK inhibitors (TKI, here represented by entrectinib and larotrectinib) achieve their antitumor activity by interacting with the intracellular domain of the chimeric receptors, inhibiting the recruitment of the signaling pathway.
Ijms 21 00753 g001
Figure 2. Neurophysiological functions of TRK signaling and possible consequences of its alterations.
Figure 2. Neurophysiological functions of TRK signaling and possible consequences of its alterations.
Ijms 21 00753 g002
Table 1. NTRK fusions in CNS tumors.
Table 1. NTRK fusions in CNS tumors.
Tumor EntityNTRK Fusions FrequencyMost Frequently Reported NTRK Fusions
Glioblastoma1.1% (Frattini et al.) [134]
1.1% (Shah et al.) [135]
2.6% (Zheng et al.) [124]
1.2% (Kim et al.) [136]
1.7% (Ferguson et al.) [133]
BCAN-NTRK1
NFASC-NTRK1
ARHGEF2-NTRK1
CHTOP-NTRK1
GKAP-NTRK2
KCTD8-NTRK2
TBC1D2-NTRK2
EML4-NTRK3
Non-brainstem high-grade glioma10%–40% (Wu et al.)# [121]ETV6-NTRK3
TPM3-NTRK1
BTBD1-NTRK3
VCL-NTRK2
AGBL4-NTRK2
DIPG°4% (Wu et al.) [121]
Pilocytic astrocytoma 16.6% (Ferguson et al.) [133]
3.1% (Jones et al.) [114]
BCAN-NTRK1
NACC2-NTRK2
QKI-NTRK2
Anaplastic astrocytoma2.3% (Ferguson et al.) [133]NOS1AP-NTRK2
Glioma NOS4.1% (Ferguson et al.) [133]SQSTM1-NTRK2
Low-grade glioma0.7% (Zhang et al.) [112]
0.43% (Stransky et al.) [72]
4.3% (Ferguson et al.) [133]
ETV6-NTRK3
AFAP1-NTRK2
VCAN-NTRK2
High-grade glioneuronal tumorCase report (Kurozumi et al.) [127]ARHGEF2-NTRK1
GangliogliomaCase report (Prabhakaran et al.) [124]TLE4-NTRK2
# Age-dependent frequency (highest rate was observed in <3yy patients). ° Diffuse Intrinsic Pontine Glioma.
Table 2. Main clinical trials evaluating NTRK-fusion inhibitors.
Table 2. Main clinical trials evaluating NTRK-fusion inhibitors.
MoleculePopulation and EnrollmentAllocation and Intervention ModelPhasePrimary OutcomesStart Date and Current StatusIdentifier
Entrectinib
(RXDX-101)
Adult (minimum age: 18 Years)—84 participantsNon-Randomized—Single Group AssignmentIDose limiting toxicity
Maximum tolerated dose
Recommended Phase II dose
Overall response rate
2014—Active, Not RecruitingNCT02097810
RXDX-101-01
(STARTRK-1)
Entrectinib
(RXDX-101)
Adult (minimum age: 18 Years)—300 participants (estimated)Non-Randomized—Parallel AssignmentIIObjective response rate2015—RecruitingNCT02568267
RXDX-101-02
(STARTRK-2)
Entrectinib
(RXDX-101)
Pediatric and Adult
(maximum age: 22 Years)—65 participants
Non-Randomized—Single Group AssignmentIMaximum tolerated dose
Recommended Phase II dose
Objective response rate
2016—RecruitingNCT02650401
RXDX-101-03
(STARTRK-NG)
Larotrectinib
(LOXO-101)
Pediatric and Adult
(minimum age: 18 Years)—6452 participants
Non-Randomized—Parallel AssignmentIIProportion of patients with objective response2015—RecruitingNCT02465060
EAY131
NCI-2015-00054
Larotrectinib
(LOXO-101)
Pediatric and Adult (minimum age: 12 Years)—320 participantsNon-Randomized—Parallel AssignmentIIBest overall response rate2015—RecruitingNCT02576431
LOXO-TRK-15002
(NAVIGATE)
Larotrectinib
(LOXO-101)
Pediatric and Adult
(maximum age: 21 Years)
—174 participants
Non-Randomized—Parallel AssignmentI/IINumber and severity of adverse events (Phase I)
Overall response rate (Phase II)
2015—RecruitingNCT02637687
LOXO-TRK-15003
(SCOUT)
Larotrectinib
(LOXO-101)
Pediatric and Adult
(from 12 Months to 21 Years)—1000 participants (estimated)
Non-Randomized—Parallel AssignmentIIObjective response rate2017—RecruitingNCT03155620
APEC1621SC
NCI-2017-01251
Larotrectinib
(LOXO-101)
Pediatric and Adult
(from 12 Months to 21 Years)—49 participants
Non-Randomized—Single Group AssignmentIIObjective response rate2017—RecruitingNCT03213704
APEC1621A
NCI-2017-01264
Larotrectinib
(LOXO-101)
Pediatric and Adult
(maximum age: 30 Years)—70 participants
Non-Randomized—Single Group AssignmentIIObjective response rate2019—RecruitingNCT03834961
ADVL1823
NCI-2019-00015
Repotrectinib
(TPX-0005)
Pediatric and Adult (12 Years and older)—450 (estimated)Non-Randomized—Single Group AssignmentI/IIDose limiting toxicities
Recommended Phase II dose
Overall response rate
2019—RecruitingNCT03093116
TPX-0005-01
(TRIDENT-1)
Repotrectinib
(TPX-0005)
Pediatric (4 Years to 12 Years)—12 participantsNon-Randomized—Single Group AssignmentIDose limiting toxicities
Pediatric recommended Phase II dose
2019—RecruitingNCT04094610
TPX-0005-07
Selitrectinib
(LOXO-195)
Pediatric and Adult (minimum age: 1 Month)Expanded Access (Individual Patients)NANA2017—Available (Expanded Access)NCT03206931
Selitrectinib
(LOXO-195)
Pediatric and Adult (minimum age: 1 Month)—93 participantsNon-Randomized—Sequential AssignmentI/IIMaximum tolerated dose
Recommended dose
Overall response rate
2017—RecruitingNCT03215511
LOXO-EXT-17005
NA: Not applicable.
Table 3. Available diagnostic assays for detecting NTRK fusions.
Table 3. Available diagnostic assays for detecting NTRK fusions.
Assay TypeAdvantagesLimitationsTurnaround TimeMain Role in Potential Diagnostic Algorithms
IHC
  • Commonly available
  • Limited cost
  • Minimal tissue required
  • Allows correlation with histology
  • Confirms protein expression
  • panTRK antibody available
  • Low sensitivity or specificity in specific settings
  • No information about the fusion partner
1–2 daysScreening
FISH
  • Minimal tissue required
  • High sensitivity and specificity although false negative results are possible
  • Specific lab facilities required and expertise for interpretation
  • No information about the fusion partner
  • One probe-one gene evaluation, thus time-consuming and higher costs
3–5 daysConfirmatory
RT-PCR
  • Limited cost
  • High sensitivity and specificity
  • Requires knowledge about the fusion partners before testing and specific primers must be prepared
  • Good pre-analytics required to preserve RNA
5–7 daysConfirmatory
Real time-PCR
  • Limited cost
  • High sensitivity
  • High specificity
  • Good pre-analytics required to preserve RNA
  • It does not provide information regarding the specific fusion partners and it evaluates a pre-determined set of rearrangements, thus novel or rare fusions will be missed
5–7 daysScreening/Confirmatory*
RNA-NGS
  • Evaluation of all potential fusions in a sample if Total RNA is analyzed
  • Provides characterization of fusion partners
  • High sensitivity
  • High specificity
  • Specific lab facilities required and expertise for interpretation
  • High costs
  • Good pre-analytics required to preserve RNA
  • Longer TAT
1–3 weeksScreening/Confirmatory*
DNA-NGS
  • It can provide an overall characterization of tumor molecular profile (mutations, CNV, tumor mutation burden…)
  • Provides characterization of fusion partners
  • High sensitivity with some caveats
  • High specificity
  • Chance of detecting non-significant chromosomal rearrangements
  • Potential low sensitivity for specific fusions
  • Specific lab facilities required and expertise for interpretation
  • High costs
  • Longer TAT
1–3 weeksScreening/Confirmatory*
DNA/RNA-NGS
  • It provides the most complete characterization of tumor molecular profile (mutations, CNV, tumor mutation burden, fusions…)
  • Provides characterization of fusion partners
  • High sensitivity
  • High specificity
  • Specific lab facilities required and expertise for interpretation
  • High costs
  • Longer TAT
1–3 weeksScreening/Confirmatory*
* depending on each laboratory diagnostic routine workup of a sample (for instance based on tumor type) and available resources/facilities.

Share and Cite

MDPI and ACS Style

Gambella, A.; Senetta, R.; Collemi, G.; Vallero, S.G.; Monticelli, M.; Cofano, F.; Zeppa, P.; Garbossa, D.; Pellerino, A.; Rudà, R.; et al. NTRK Fusions in Central Nervous System Tumors: A Rare, but Worthy Target. Int. J. Mol. Sci. 2020, 21, 753. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21030753

AMA Style

Gambella A, Senetta R, Collemi G, Vallero SG, Monticelli M, Cofano F, Zeppa P, Garbossa D, Pellerino A, Rudà R, et al. NTRK Fusions in Central Nervous System Tumors: A Rare, but Worthy Target. International Journal of Molecular Sciences. 2020; 21(3):753. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21030753

Chicago/Turabian Style

Gambella, Alessandro, Rebecca Senetta, Giammarco Collemi, Stefano Gabriele Vallero, Matteo Monticelli, Fabio Cofano, Pietro Zeppa, Diego Garbossa, Alessia Pellerino, Roberta Rudà, and et al. 2020. "NTRK Fusions in Central Nervous System Tumors: A Rare, but Worthy Target" International Journal of Molecular Sciences 21, no. 3: 753. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21030753

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop