Next Article in Journal
Epigenetic Silencing of Ubiquitin Specific Protease 4 by Snail1 Contributes to Macrophage-Dependent Inflammation and Therapeutic Resistance in Lung Cancer
Previous Article in Journal
Neurocognitive Decline Following Radiotherapy: Mechanisms and Therapeutic Implications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Roles of TrkC Signaling in the Regulation of Tumorigenicity and Metastasis of Cancer

Laboratory of Molecular Disease and Cell Regulation, Department of Biochemistry, School of Medicine, Gachon University, Incheon 21999, Korea
Cancers 2020, 12(1), 147; https://doi.org/10.3390/cancers12010147
Submission received: 14 November 2019 / Revised: 2 January 2020 / Accepted: 7 January 2020 / Published: 8 January 2020

Abstract

:
Tropomyosin receptor kinase (Trk) C contributes to the clinicopathology of a variety of human cancers, and new chimeric oncoproteins containing the tyrosine kinase domain of TrkC occur after fusion to the partner genes. Overexpression of TrkC and TrkC fusion proteins was observed in patients with a variety of cancers, including mesenchymal, hematopoietic, and those of epithelial cell lineage. Both microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) were involved in the regulation of TrkC expression through transcriptional and posttranscriptional alteration. Aberrant activation of TrkC and TrkC fusion proteins markedly induces the epithelial-mesenchymal transition (EMT) program, growth rate, tumorigenic capacity via constitutive activation of Ras-MAP kinase (MAPK), PI3K-AKT, and the JAK2-STAT3 pathway. The clinical trial of TrkC or TrkC fusion-positive cancers with newly developed Trk inhibitors demonstrated that Trk inhibitors were highly effective in inducing tumor regression in patients who do not harbor mutations in the kinase domain. Recently, there has been a progressive accumulation of mutations in TrkC or the TrkC fusion protein detected in the clinic and its related cancer cell lines caused by high-throughput DNA sequencing. Despite given the high overall response rate against Trk or Trk fusion proteins-positive solid tumors, acquired drug resistance was observed in patients with various cancers caused by mutations in the Trk kinase domain. To overcome acquired resistance caused by kinase domain mutation, next-generation Trk inhibitors have been developed, and these inhibitors are currently under investigation in clinical trials.

1. Introduction

Trk proteins (Trk or NTRKs) are identified as members of tropomyosins fused to a tyrosine kinase domain and are single-pass transmembrane receptors. Trk proteins are activated by their neurotrophins, nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and neurotrophin 4/5 (NT4/5). TrkA, TrkB, and TrkC, encoded by NTRK1, NTRK2, and NTRK3, respectively, and neurotrophins, exhibit specificity in interactions with the specific receptors. TrkA preferentially binds NGF, and TrkB binds BDNF and neurotrophin-4/5, and TrkC physiologically binds to neurotrophin-3 as high-affinity transmembrane receptors for neurotrophins [1,2]. Moreover, a small peptide between the second immunoglobin-like C2 type 2 region and the transmembrane domain of Trk proteins affects ligand-binding specificity [3,4,5]. Neurotrophins and their-specific receptors regulate survival, growth, differentiation, and apoptosis in the peripheral and central neuronal systems. Activation of the Ras/MEK/MAPK pathway, PI3K/AKT pathway, and phospholipase C-gamma (PLCγ) signaling by Trk activation is crucial for neuronal survival [2,6,7].
The reduction of TrkC expression has been observed in neurodegenerative diseases, including Alzheimer’s (AD), Parkinson’s (PD), and Huntington’s diseases (HD). The selective degeneration and dysfunction of cholinergic basal forebrain neurons of the nucleus basalis is a feature of AD that mainly correlates with severe cognitive impairment. TrkC (58%) is well expressed in numerous NB of Meynert neurons in control brains, but these expressions were significantly reduced by about two-fold during progression (29.6%) in AD brains [8], and TrkC expression reduced considerably in cholinergic NB neurons during the progress of AD [9,10,11]. Moreover, TrkC expression, as well as NT-3, is remarkably expressed in the adult substantia nigra pars compacta, but reduced expression of TrkC in the SN of PD patients induced abnormal accumulation of α-synuclein as the hallmark of PD [12]. Moreover, TrkC expression restores long-term striatal depression on corticostriatal synaptic plasticity in the 3-NP-treated animal model of HD. TrkC activates the neuronal survival pathways, including the Ras/MEK/MAPK and PI3K/AKT pathways. Hence, TrkC-mediated activation of the Ras/MEK/MAPK and PI3K/AKT pathways promotes cellular functions such as proliferation, growth, and survival in cancer [13], raising the possibility that the role of TrkC protein provided from studies in the sympathetic nervous system may contribute to disease pathology.

2. Incidence of TrkC Expression in Cancer Development

In addition to the functional role of TrkC in the neuronal system, overexpression of TrkC is observed in many human tumors (Table 1). The involvement of TrkC in a variety of human cancers was first reported in studies on TrkC expression in neuroblastoma and glioma. Neuroblastoma is the most common extracranial solid tumor that occurs early childhood, and over 60% of the neuroblastomas are metastatic. It accounts for approximately 15% of pediatric cancer deaths [14]. In neuroblastoma, TrkC is highly expressed in 25% of primary neuroblastomas and is often accompanied by TrkA [15]. Moreover, a subset of stage IV neuroblastomas exhibits high-level NT-3 and TrkC co-expression [16]. In glioma, TrkC was up-regulated in 91.8% of glioma patient samples [17], and high-grade gliomas showed a more positive immunoreactivity than low-grade gliomas in NT-3 and TrkC expression [18]. Furthermore, TrkC was up-regulated in 86% of medulloblastomas and 68% of non-cerebellar primitive neuroectodermal (PNET) tumors (17 glial tumors, three ependymal tumors, and one teratoid tumor) [19].
TrkC is identified in several other types of human cancers as well. In the case of breast cancer, two studies have reported the incidence of TrkC in these tumor types. TrkC was more significantly overexpressed in basal-like breast cancer cells than in luminal cancer cells, and TrkC expression was elevated in 82% of breast cancer patients [20]. Hepatocellular carcinoma (HCC) represents approximately 90% of primary liver cancer and is the second main cause of cancer-related deaths in the world [21]. TrkC is overexpressed significantly in HCC cells, and its elevated expression was found to be correlated with the unmethylated TrkC promoter [22].
Moreover, TrkC expression was observed in 86% of tumors, in which TrkC molecules were present as alternatively spliced isoforms [23]. TrkC expression was also observed in primary and metastatic melanoma cells [24]. TrkC expression caused by immunoreactivity was observed in 62.5% of melanomas of various stages, and its expression significantly increased 58% of melanoma that progressed from in situ lesions, 91% of papillary dermal invasions, 57% of melanomas which invaded the deeper dermis, and 31% of melanomas which metastasized to sites other than the compound nevi [25]. Furthermore, there was an increase by 66% in the observed expression of TrkC in ductal pancreatic tissue compared to in normal adjacent tissue [26,27]. The occurrence of TrkC was observed at similar levels in prostate cancer specimens obtained from patients both with and without neoadjuvant hormonal therapy [28]. Moreover, TrkC expression was identified in lung cancer and leukemia. TrkC receptor was not detected in lung adenocarcinomas and bronchioloalveolar carcinomas, but was observed in 35% of well-differentiated squamous cell carcinomas within large size tumor cells, and in 55% of small cell lung cancers (SCLC) [29]. TrkC was significantly overexpressed in leukemia subtypes such as Lymphoblastic Leukemia, Acute Myeloid Leukemia (AML), and Chronic Lymphocytic Leukemia (CLL) [30]. Additionally, in gastric cancer (GC), high expression of TrkC was significantly correlated with distant metastasis, lymph node metastasis, distant metastasis, or recurrence of the disease [31].
Finally, during progression from typical thyroid C cell hyperplasia to the later stages of medullary thyroid carcinoma (MTC), substantial changes of TrkC were detected by using immunostaining. TrkC expression was not detected in healthy thyroid C cells, but moderate or strong TrkC immunostaining was identified in 87% of MTC tumors. Also, TrkC expression in MTC cell induces tumorigenic ability and primary tumor formation in nude mice [32].

2.1. The Functional Role of Long Noncoding RNA and microRNA in TrkC Expression

Recent reports have demonstrated that miRNAs and lncRNAs are involved in the regulation of TrkC expression. miRNAs are 20–22 nucleotide long non-coding RNA molecules, which regulate gene expression at the post-transcriptional level. The expression of full-length TrkC is markedly reduced by miR-151-3p and miR-185. Also, expression of the truncated TrkC isoform was considerably reduced by miR-128, miR-485-3p, miR-765, and miR-768-5p [33]. Moreover, miR-9, miR-125a, and miR-125b were able to decrease cell growth, and truncated TrkC isoform was the target of miR-8, miR-125, and miR-125b in the repression of human neuroblastoma or medulloblastoma cell proliferation [34,35].
Additionally, lncRNAs are involved in the transcription of proteins directly and indirectly via transcriptional and posttranscriptional alteration, and are a potential therapeutic target of cancer [36]. Long noncoding RNA participates in the regulation of TrkC expression. Elevated expression of PVT1-214 is significantly associated with clinicopathological characteristics and poor survival of GC patients, and induces tumorigenic ability of GC by increasing TrkC expression via inhibition of miR-128 [37]. Another long noncoding RNA LINC00978 was markedly upregulated in GC patients and associated with poor survival outcome of GC patients. In addition, LINC00978 expression induces metastatic potential and inhibits apoptosis of cells. Moreover, LINC00978 promoted tumorigenicity and proliferation of cancer cells through inhibition of reduction of TrkC expression by miR-497 [38]. In contrast, TrkC was one of the target genes of LINC00052, and down-expression of TrkC increases the aggressiveness and proliferation of SMMC7721 cells. Also, LINC00052 suppresses the expression of the truncated isoform of TrkC by forming complementary base pairing with miR128, miR-485-3p, and miR-765 [39].

2.2. Somatic Mutations of TrkC in Cancer

Progressive accumulation of mutations can cause cancer or other diseases, and most tumors carry 1000 to 20,000 somatic point mutations, which has been reported in high-throughput DNA sequencing analyses of cancer cell genomes [40,41], with these mutations leading to constitutive activation of signaling circuits [42]. Thus, we now know about the mutation of TrkC identified in colorectal cancer (CRC) cell lines by mutational analysis of the tyrosine kinome, which was suggested to be a pathogenic mutation (G608S, I695V, R731Q, K732T, L760I) [43]. Also, TrkC has a nonsynonymous somatic mutation in pancreatic (H599Y, G608S, E322K) [44,45,46], breast (R678Q) [47], lung (H677Y, R721F) [48], and gastric (T149R, K746T, E543D) cancers [49]. Moreover, somatic mutations have been identified in 454 sites of TrkC, including the kinase domain, in 36 various cancer types (Table 2), and somatic mutation of the TrkC gene mainly occurs through missense mutations (Figure 1A). Around 25% of human cutaneous squamous cell carcinoma contains missense mutations in TrkC, and these missense mutations were also detected in ~7.8% of human lung adenocarcinomas and ~7% uterine endometrial carcinoma patients (Figure 1B). However, the origin of TrkC mutations remains unknown, but patients of various cancer types have a spectrum of many different TrkC mutations, including modifications of the tyrosine kinase domain.
Figure 1. Somatic mutation frequency of TrkC in patients of 36 various cancer types. (A) Total of 530 mutations in TrkC, including 454 missense mutations, 70 truncating mutations, three inframe mutations, and discovered in patients of various cancers. Specifically, 177 of 530 mutations identified in the tyrosine kinase domain of TrkC. LRRNT: Leucine-rich repeat N-terminal domain, LRR1; Leucine-rich repeat region 1, LRR2: Leucine-rich repeat region 1, LRRCT: Leucine-rich repeat C-terminal domain, Ig-like C2: Immunoglobulin-C2-set domain, TM: transmembrane domain. (B) The overall frequency of mutated TrkC in cancer types. cSCC [50], DM [51], CM: TCGA Dataset, BCC: [52], LA: [53], EC: TCGA Dataset, VV-SCC: [54], NSCLC: [55], UTUC [56], CTCL: [57], AS: TCGA Dataset, CRC: [58], GC: TCGA Dataset, CSCC: TCGA Dataset, EAC: [59], HNC: TCGA Dataset, RMS: [60], PAC: [61], DLBCL: TCGA Dataset, HCC: TCGA Dataset, AC: Ampullary carcinoma [62], UCS: TCGA Dataset, BUC: TCGA Dataset, RCC: TCGA Dataset, PA: [63], CC: [64], GC: [65], PDTC & ATC: [66,67], OSC: TCGA Dataset, BC: [68], CCRCC: TCGA Dataset, CLL: [69], DG: [70].
Figure 1. Somatic mutation frequency of TrkC in patients of 36 various cancer types. (A) Total of 530 mutations in TrkC, including 454 missense mutations, 70 truncating mutations, three inframe mutations, and discovered in patients of various cancers. Specifically, 177 of 530 mutations identified in the tyrosine kinase domain of TrkC. LRRNT: Leucine-rich repeat N-terminal domain, LRR1; Leucine-rich repeat region 1, LRR2: Leucine-rich repeat region 1, LRRCT: Leucine-rich repeat C-terminal domain, Ig-like C2: Immunoglobulin-C2-set domain, TM: transmembrane domain. (B) The overall frequency of mutated TrkC in cancer types. cSCC [50], DM [51], CM: TCGA Dataset, BCC: [52], LA: [53], EC: TCGA Dataset, VV-SCC: [54], NSCLC: [55], UTUC [56], CTCL: [57], AS: TCGA Dataset, CRC: [58], GC: TCGA Dataset, CSCC: TCGA Dataset, EAC: [59], HNC: TCGA Dataset, RMS: [60], PAC: [61], DLBCL: TCGA Dataset, HCC: TCGA Dataset, AC: Ampullary carcinoma [62], UCS: TCGA Dataset, BUC: TCGA Dataset, RCC: TCGA Dataset, PA: [63], CC: [64], GC: [65], PDTC & ATC: [66,67], OSC: TCGA Dataset, BC: [68], CCRCC: TCGA Dataset, CLL: [69], DG: [70].
Cancers 12 00147 g001
Table 1. Detected TrkC in multiple histologies.
Table 1. Detected TrkC in multiple histologies.
Cancer NameFrequencyRef.
Neuroblastoma14/55 (25%)[15]
Glioma215/234 (91.8%)[17]
Medulloblastomas17/26 (85%)[19]
Non-cerebellar primitive neuroectodermal (PNET) tumors21/31 (67.7%)[19]
Breast cancer14/17 (82.4%)[20]
Invasive ductal carcinoma118/236 (49.6%)[71]
Hepatocellular carcinoma 44/51 (86%)[21]
Melanoma40/64 (62.5%)[25]
Pancreatic cancer31/47 (66%)[26,27]
Squamous cell carcinoma3.5/10 (35%)[29]
Small cell lung cancers4.4/8 (55%)[29]
Thyroid cancer21.8/25 (87%)[32]
Table 2. Mutation of TrkC in patients of Cancer types.
Table 2. Mutation of TrkC in patients of Cancer types.
Cancer NameMutation (%)Protein ChangeRef.
Ampullary Carcinoma (AC)1.88S136G, E211A, P796L[62]
Angiosarcoma (AS)4.17P577L, G235ETCGA
Oligodendroglioma (ODG)0.68Q808 *[72]
Bladder Urothelial Carcinoma (BUG)2.94D609N, S477 *, L53F, E398K, G727 *, Q119E, E360KTCGA
Breast Carcinoma (BC)0.75H349R, E810Q, G545A, L574V, E412K, N714K, P210T, A555V, K424T, G547R, V413M, Q177H, L108R, H254Y[68]
Cervical Squamous Cell Carcinoma (CSCC)3.04A122T, F162S, L384M, D476N, L5V, R153Q, F162LTCGA
Chronic Lymphocytic Leukemia (CLL)0.33A647T[69]
Colorectal Adenocarcinoma (CRC)4.17F450L, L653I, S113A, R745Q, L115R, T777M, G608S, T149M, I759M, V217I, S117N, K181N, C782R, Q206H, T777M, T149M, K746R, K125N, R791Q, R745Q, Y456H, E86D, N191D, D624Y, Q159H, R201H, T730S, V97M, R745Q, G487D, R535M, R130H, T149M, A631D, D527G, Q586 *, V97M, R791W, G497R, Q145H, T332M, R130H, R791W, R814Q, A664T, K746T, R89H, Q119H, K346R[58]
Cutaneous Melanoma (CM)20E762K, P509H, E211 *, H370N, P577S, L157F, E778K, D167N, R793 *, E318K, R542Q, E810K, H658Q, D697N, G235E, G356E, G633V, D584N, S359F, G356R, E590K, D576N, Y834N, R735C, G757R, R153L, G623E, R153L, W771 *, L299I, D242N, V217F, V37A, H423N, V726L, R116W, K768E, G178E, G727E, R735C, L152F, G339R, E819K, M667I, E86 *, Q773 *, W243L, Q255 *, P417L, D836H, T831I, P401STCGA
Cutaneous Squamous Cell Carcinoma (cSCC)25.64P577S, H264Y, D576N, Q773 *, E778K, G767E, A548V, D584N, L152F, D61G, G828E, K797E[50]
Cutaneous T-Cell Lymphoma (CTCL)4.65P577S, S741N[57]
Desmoplastic Melanoma (DM)20H128Y, P329L, E351K, S701F[51]
Diffuse Glioma (DG)0.38I488T, L282R, I488V[70]
Diffuse Large B-Cell Lymphoma (DLBCL)2.08D98NTCGA
Endocervical Adenocarcinoma (EA)1.94E546KTCGA
Esophageal Adenocarcinoma (EAC)3.97V687G, G487S, R46W, L115P, A826V[59]
Gallbladder Cancer (GC)3.13I817M[65]
Head and Neck Carcinoma (HNC)2.51H632Y, Q159K, S77 *, T253N, Q531R, A636V, H423N, Y705N, K367N, R326L, C362S, C231F, H729N,TCGA
Hepatocellular Carcinoma (HCC)2.47G233S, Q145R, D527G, S701T, H622N, F395Y, E318DTCGA
Cholangiocarcinoma (CC)2.78V451I[64]
Lung Adenocarcinoma (LA)7.83L282M, P7R, H658N, H370N, L384M, N454S, S39R, Y188H, C320F, G545C, A380D, R121I, R814L, N718Y, P796S, G605L, G666C, G757V, N137K, P417H, R306H, M292I, T420S, H677Q, L639I, L827M, Y376C, P120H, S775 *, G487R, V97L, G67V, H84Y, G828V, K602N, P526Q, V779F, N382I, Y821F, K551N, N218H, E740K, R169L, P120H, T500S, S184R, D428H, S28Y, P330Q, T777K, K181N, A380D, F603I, R814L, E314D, S309I, P526Q, K397N, H394Q, K621N, G652V, G233V, I212T, P383A, R535M, N52K, F147L, V704F, G608C, V221L, W754C, E357D, Y604F, G463 *, E398Q, Y834F, R138L, Y821C, M202L, D240H, Q515H, K461R, V799L, Q773K, M464I, H729Y, E512K, P120H, V324A, Q172H, V273L, R343L, K346N, R121I, R459G, T506A, D495E, R343W, S741I, L364P, S4C, P509T, P612A, A435E, T230S, E314Q, H84N, G642 *, N338Y, T707K, D801N, V241A, G279A, G487S, F123L, S296R, L629F, G649C, A581P, Y744F, R343L, D635N, P738H, R735H, T563N[53,55]
Pancreatic Adenocarcinoma (PA)1.83R153Q, V29M, R306H, K746T, E322K, Q643K, E223Q[61]
Papillary Renal Cell Carcinoma (PRCC)1.41L270V, E179G, T490K, G104 *TCGA
Plasma Cell Myeloma (PCM)0.98R745W, E351D[73]
Prostate Adenocarcinoma (PA)1.13D609N, G497R, V640A, F747S, R793Q, P417L, T93M, P509S, T777M, T332M[63]
Clear Cell Renal Cell Carcinoma (CCRCC)2.86D609N, R735HTCGA
Rhabdomyosarcoma (RMS)2.3Y709F[60]
Ovarian Serous Cancer (OSC)0.69P304L, L827F, D584ETCGA
Skin Cancer, Non-Melanoma
(Basal Cell Carcinoma; BCC)
8.19S741N, S751R, G608I, E475K, R745W, Q673H, E590D, M99I, P467L, S117R, Q255 *, L760F, K381E, E154D, G696E, M202I, Q655R, R735C, K346R[52]
Squamous Cell Carcinoma of the Vulva/Vagina (VV-SCC)6.67G437 *[54]
Gastric cancer (GC)3.18H486N, H521N, K181R, T490M, L115P, R326H, R201H, D277G, W335R, S117T, R787H, R791W, L197F, A435E, L17 *TCGA
Thyroid Cancer (TC)0.85R630W, N294T[66,67]
Upper Tract Urothelial Carcinoma (UTUC)4.71D499N, D527Y, R153Q, R326H[56]
Uterine Endometrioid Carcinosarcoma (UCS)7.18T261S, F617L, C523Y, T390I, M202I, K346N, R459W, P832S, A681T, D537Y, Y456H, R222 *, E598 *, R793Q, K111N, A96T, A580V, R222Q, E357D, H482Y, L187P, P55S, K125N, E556K, R47Q, S117N, A664T, G114R, G699D, I511T, S65N, K397N, I508T, E86D, N816S, E412K, V97M, G233D, G374D, Y352C, Q159H, A387T, E58K, D836A, V217A, D75N, M700I, R518C, E322K, D703N, S151N, F772L, V221I, L712P, E287DTCGA
Esophageal Carcinoma (EC)2.43D277G, W335R, S117T, K181R, R791W, H486N, H521N, L197F, R787H, T490M[59]
*: nonsense mutation; TCGA: TCGA database.

2.3. TrkC Fusion in Cancer

In most NTRK3 fusion proteins, the 3′ region of NTRK3, which contains the tyrosine kinase domain fused with the 5′ region of the partner gene, is expressed in cancer. ETV6-NTRK3 is known as a chimeric oncoprotein, which occurs in various cancer types, including mesenchymal, hematopoietic, and epithelial cell lineages. ETV6-NTRK3 was initially cloned and identified in five (100%) out of five congenital fibrosarcomas (CFS) and a pediatric spindle cell malignancy [74,75] in the case report of AML [76,77]. ETV6-NTRK3 expression was also identified in five of six cellular mesoblastic nephroma (CMN) [75,77] and 10 of 11 CMN [78], an infantile spindle cell tumor of the kidney. However, the expression of ETV6-NTRK3 was not detected in classical CMN and Wilm’s tumor, as there is recurrent chromosomal translocation (t(12;15)(p13;q25)). This rearrangement generates a gene fusion encoding the Helix-Loop-Helix Domain (HLH) of the ETV6 (TEL, ETS family transcription factor) linked to the tyrosine kinase (PTK) domain of TrkC [79].
In breast cancer, ETV6-NTRK3 is expressed in 12 (92%) of 13 patients with human secretory breast carcinoma (SBC), a rare subtype of IDC, and the resulting ETV6-NTRK3 protein functions as a chimeric protein with potent transforming activity in fibroblasts [80]. Moreover, secretory breast carcinomas are triple-negative and express basal markers, while secretory breast carcinomas with ETV6-NTRK3 protein belong to the basal-like breast carcinomas [81,82]. Moreover, ETV6-NTRK3 was identified in CRC [83,84], glioma [85,86], spitz tumor [87], lung adenocarcinoma [88], infantile fibrosarcoma [88,89], gastrointestinal stromal tumor [88,90], thyroid carcinoma [91,92], uterine sarcoma [86], and sinonasal adenocarcinoma [93]. ETV6-NTRK3 expression was detected in 75% of lung adenocarcinomas, 70% of infantile fibrosarcomas, and 49% of gastrointestinal stromal tumors [88]. Moreover, ETV6-NTRK3 was identified in 26% of papillary thyroid carcinomas [91,92], and interestingly, the prevalence in the rearrangement of ETV6-NTRK3 was associated with exposure to radiation based on a case study of patients who suffered from the Chernobyl accident. The rearrangement of ETV-NTRK3 significantly increased to 14.5% in radiation-related papillary thyroid carcinomas (PTCs) from an occurrence of 2% in sporadic PTCs [91]. The results of this study suggest that ETV6-NTRK3 may represent another type of chromosomal rearrangement associated with the robust growth pattern of PTC in patients exposed to radiation [91].
Additional NTRK3 fusion proteins, occurring in small numbers, have been identified in various cancer types. AKAP13-NTRK3 was identified in a rare case of low-grade glioma [94], and BTBD1-NTRK3 was induced in high-grade astrocytoma [85]. Another TrkC fusion, EML4-NTRK3, was identified in rare cases of several types of cancers, including uterine and vaginal sarcomas [95], dermatofibrosarcoma [96], infantile fibrosarcoma and congenital mesoblastic nephroma [97], infantile fibrosarcomas [98], and glioblastoma [84]. Moreover, the expression of EML4-NTRK3 induces the tumorigenic ability of NIH3T3 fibroblast cells in vivo and in vitro [99]. Interestingly, the majority of the TrkC fused to the 3′ region of HOMER2 contains WASP homology region 1 (WH1) and a coiled-coil domain. Moreover, TrkC combined with STRN or STRN3 as paralog of STRN includes a coiled-coil domain. STRN, a calmodulin-binding protein member, is a partner of ALK protein, and its fusion protein, which was identified in thyroid and lung carcinoma [100,101], leads to constitutive activation of ALK tyrosine kinase via dimerization mediated by the coiled-coil domain of STRN [100]. Moreover, most of the TrkC fusion proteins contain a coiled-coil domain, including STRN, STRN3, TFG, TPM4, HOMER2, MYH9, MYO5, and EML4 (Figure 2). There is a possibility that ligand-independent constitutive activation of tyrosine kinase domain of the TrkC fusion protein is mediated by the coiled-coil domain of the partner gene product. TrkC fusion partners have been recently identified in a wide range of cancer types (Figure 2 and Table 3).

2.4. The Biological Function of TrkC in Cancer

Four principal mechanisms mediate aberrant activation of receptor tyrosine kinase (RTK) in human cancers: autocrine activation, chromosomal translocations, overexpression, or gain-of-function mutations [122] and activation of signals by overexpression of oncogenes including RAS, RAF, and MYC would result in correspondingly increased tumorigenicity of cancer cells [123]. For example, the epidermal growth factor receptor (EGFR) was overexpressed in many cancers, and it was leading to overexpression and constitutive activation of EFGR tyrosine kinase activity [124,125]. Moreover, ErbB2, an EFGR family receptor, is highly overexpressed in various cancer types, including breast cancer, and promotes tumor progression via ligand-independent constitutive activation, while the overexpression correlates with poor prognosis [126,127]. As per previous reports, the primary mechanism of action by overexpression of TrkC occurs through increased TrkC activity without NT-3. Overexpression of TrkC in human MTC cells exhibited an increased growth rate, tumorigenic capacity, and primary tumor formation in vivo relative to control cells before ligand addition and after NT-3 addition, consistent with the fact that TrkC is phosphorylated before ligand addition [32]. Moreover, overexpression of TrkC in MCF10A breast cancer cells or RIE-1 normal intestinal epithelial cells increases the metastatic ability without treatment with NT-3 [20,128].
Auto-activation of RTKs recruited and activated a wide range of downstream signaling proteins via interaction with SH2 or PTB (phosphotyrosine binding domain) domain-containing signaling proteins [122,129]. Recent studies have reported that the tyrosine kinase domain of TrkC and ETV6-NTRK3 interacts with the SH2 domain of the Src and PTB domain of IRS-1. Ligand-independent PTK activation of ETV6-NTRK3 and TrkC in breast and colon cancer leads to constitutive activation of the Ras-MAP kinase (MAPK) mitogenic pathway and the phosphatidylinositol 3-kinase (PI3K)-AKT pathway, as well as upregulation of cyclin D1 mediating cell survival [111,113], In addition, its activation by TrkC and ETV6-NTRK3 is mediated via activation of c-Src by using complex formation [130,131]. IRS-1 functions as an adapter protein, linking ETV6-NTRK3 and TrkC for constitutive activation of downstream signals. After ETV6-NTRK3 associates with IRS-1, ETV6-NTRK3 recruits GRB2 and p85 and activates Ras-MAPK and PI3K-AKT pathways. However, mutation of the PTB activation loop tyrosine of ETV6-NTRK3 defects IRS-1-mediated transformation-associated pathways, including Ras-MAPK and PI3K-AKT in NIH3T3 cells by completely blocking phosphorylation of IRS-1 via failure to associated with IRS-1. These results indicate that the C-terminus, including the PTK domain of TrkC, is essential for transforming activity in multiple cell lineages [131,132].
Metastases produced by cancer cells are formed by a complex succession of an invasion-metastasis cascade from invading local epithelial cells in the surrounding extracellular matrix (ECM) to reinitiate neoplastic growths at metastatic sites [42]. In this cascade, the EMT induces a significant loss in junctional E-cadherins, which are essential cell-to-cell adhesion proteins that prevent dissociation of epithelial cells. Through activation of the EMT, the epithelial cells acquire mesenchymal features, which include motility, invasiveness, and heightened resistance to apoptosis [133]. Moreover, constitutive RTK activation and loss of E-cadherin lead to stabilization of the mesenchymal status of cancer cells and maintain continuous EMT-inducing heterotypic signaling from the tumor microenvironment [134]. TrkC enhances the metastatic potential of cancer via the induction of EMT in breast and colon cancers. TrkC induces JAK2 and STAT3 expression, but kinase-dead mutant of TrkC does not induce expression of these proteins. These results have revealed the constitutive activation of the JAK2-STAT3 signaling pathway triggered by the activation of tyrosine kinase of TrkC. Moreover, TrkC induces JAK2 stabilization through the suppression of JAK2 ubiquitination via inhibition of the JAK2-SOCS3 complex formation, activation of the EMT program through Twist-1 expression and the EMT transcription factor (EMT-TFs), and activation of the IL6/JAK2/STAT3 pathway [20,128].
Additionally, analysis has found that ETV6-NTRK3 upregulates genes that are associated with cell motion, membrane invagination, and regulation of cell proliferation, and also downregulates genes involved in cell adhesion [135].
The overexpression and activation of STAT1 were observed in several human cancers [136] and overexpression of STAT1 acquired chemoresistance and radioresistance in breast and lung cancers by suppressing the cytotoxic response and inducting prosurvival genes [137,138,139]. Interestingly, ETV6-NTRK3 attenuated STAT1 acetylation through STAT1 phosphorylation by direct interaction and eventually increased the activity of NF-κB by promoting nuclear translocation of NF-κB, through inhibition of the STAT1-NF-κB complex formation [135].
Transforming growth factor (TGF-β) is a multifunctional cytokine that functions as a potent growth inhibitor of epithelial cells and as a promoter of tumor progression. The property of growth inhibition by TGF-β depended on the signaling through cyclin-dependent kinase (CDK) inhibitors [140]. Recent results have highlighted the importance of PTK activation of TrkC that inhibits the growth inhibitory activity of TGF-β. Both TrkC and ETV6-NTRK3 directly interacted with TGF-β type II receptor and inhibited phosphorylation of TGF-β type II receptor, and eventually blocked both TGF-β-induced Smad2/3 phosphorylation and TGF-β growth inhibitory activity. Moreover, inhibition of the tyrosine kinase activity of TrkC or ETV6-NTRK3 restored TGF-β tumor suppressor activity [141,142].
Bone morphogenetic proteins (BMPs) closely resemble a bifunctional TGF-β in cancer cell regulation. TrkC blocks the BMP tumor suppressor activity through the suppression of bone morphogenetic protein 2 (BMP-2)-induced Smad1 phosphorylation and transcriptional activation by directly interacting with the BMP type II receptor [143]. Additionally, loss of REST function (transcription factor) as a tumor suppressor was identified in colon adenocarcinoma, lung cancer, and breast cancer. Moreover, the loss of REST expression in breast cancer significantly correlated with recurrence and poor survival of breast cancer patients. Additionally, the TSPY2/REST complex induces transcriptional repression of TrkC and restores the tumor inhibitory activity of TGF-β [144,145].

2.5. Targeted Therapies for Trk or Trk Fusion Protein

Trks are specifically intriguing due to the resulting chimeric oncoproteins which occur in various cancer types, and these fusion proteins are driven by ligand-independent constitutive activation, eventually activating downstream signaling pathways involved in proliferation, tumorigenicity, and the EMT in human cancers (Figure 3). This result is the reason why drug companies are developing small molecular inhibitors of TRK to treat cancer, arthritis, or pain-induced disease. Several inhibitors of Trk have been developed for the treatment of Trk or Trk fusion protein-mediated adult and pediatric solid tumors and hematologic malignancies.

2.5.1. Larotrectinib

Larotrectinib (Vitrakvi, LOXO-101, Loxo Oncology Inc., Stamford, CT, USA and Bayer, Leverkusen, Germany) was first approved by the Food and Drug Administration (FDA) in November 2018 for pediatric, adult solid tumors that have Trk and Trk fusion protein without a known acquired resistance mutation, have no satisfactory alternative treatments, or have not progressed following treatment [146]. Larotrectinib is a small molecule which is orally-administered, and the most selective Trk tyrosine kinase inhibitor with half-maximal inhibitory concentration (IC50) levels in a low nanomolar range between 5–11 nmol/L and no activity against other kinase and non-kinase targets. It functions by inhibiting the autophosphorylation of Trk via binding to the ATP-bind site of Trk [146,147].
Figure 3. Diagram of TrkC biology and signaling identified in various cancers. TrkC and TrkC isoform can enhance and activate downstream of TRK signaling, including PLCγ/IP3/DAG [1,148,149], RAS/MAPK [1,79,132,148], PI3K/AKT [1,79,132,148], and JAK/STAT signaling [20,135], resulting in the promotion of survival, aggressiveness, chemoresistance, and generation of CSCs of cancer.
Figure 3. Diagram of TrkC biology and signaling identified in various cancers. TrkC and TrkC isoform can enhance and activate downstream of TRK signaling, including PLCγ/IP3/DAG [1,148,149], RAS/MAPK [1,79,132,148], PI3K/AKT [1,79,132,148], and JAK/STAT signaling [20,135], resulting in the promotion of survival, aggressiveness, chemoresistance, and generation of CSCs of cancer.
Cancers 12 00147 g003
Larotrectinib is highly effective in inhibiting the proliferation of primary cancer cells, derived from patients harboring Trk fusion proteins. The IC50 was less than 100 mmol/L for CUTO-3.29 cells harboring MPRIP–NTRK1 and less than 10 nmol/L for KM12 cell harboring TPM3–NTRK1 or MO-91 cell harboring ETV6–NTRK3. Moreover, Larotrectinib inhibits tumor growth in a xenograft. Moreover, a phase I study of Larotrectinib for a woman with soft-tissue sarcoma metastatic to the lung demonstrated a marked improvement in multiple pulmonary metastases and almost complete tumor disappearance of the most extensive tumors [110]. Several clinical trials have assessed the efficacy and safety of Larotrectinib. The effectiveness of Larotrectinib was evaluated in 55 patients with Trk fusion-positive tumors, including TrkA (45% of the patients), TrkB (2%), and TrkC (53%), and with a recommended phase 2 dose of 100 mg twice daily in adults. The overall response rate (ORR) of patients according to the assessment was 75% (95% CI, 61–85). The complete response rate and the partial response rate was 22% and 53% of the ORR, respectively. At least 15% of the patients showed adverse events during treatment, which led to a dose reduction of Larotrectinib [88]. Moreover, Larotrectinib in a phase 1 trial significantly induced tumor regression in more than 90% of pediatric patients with solid tumors. Moreover, the efficacy of Larotrectinib for pediatric patients with TRK fusion cancers in phase 2 trials showed reductions in cancer occurrence. In total, 14 of 15 patients with Trk fusion-positive tumors in each of the Trk fusion proteins. TrkA (46% of the patients), NTRK2 (6.7%), and NTRK (40%) showed objective responses at a median of 1.7 months. The maximum tolerated dose was estimated to be 100 mg/m2 of Larotrectinib [150]. Furthermore, the clinical trial of children with locally advanced TRK fusion sarcoma demonstrated that Larotrectinib induces a high response rate, including a reduction in the tumor [151]. In the case of a pediatric patient with ETV6-NTRK3 positive secretory breast cancer, treatment with Larotrectinib achieved an almost complete response and induced substantial tumor regression [152]. Additionally, the overall response rate (ORR) of ETV6-NTRK3 positive patients was 85% (95% CI, 64–96) [153].

2.5.2. Entrectinib

The FDA approved Entrectinib (Rozlytek, Gnentech Inc., South San Francisco, CA, USA) as a new Trk inhibitor for pediatric and adult solid tumors that have Trk, ROS proto-oncogene 1 (ROS1), and anaplastic lymphoma kinase (ALK) fusion proteins without a known acquired resistance mutation, and for tumors which are metastatic or for adults with metastatic NSCLC which are ROS1-positive. Entrectinib is a potent oral small-molecule inhibitor of Trk, ROS1, and ALK with IC50 values of 0.1 to 2 nM [154].
Entrectinib has been examined in several clinical trials. ALKA-372-001 phase I Trial showed that Entrectinib has a significant antitumor response in TrkA-positive CRC, ALK-rearranged neuroblastoma, and ROS1-, or ALK-positive NSCLC patients [155]. Moreover, the efficacy was assessed in patients with solid tumors with a Trk gene fusion in other phase trials (STARTRK-1 and STRATRK-2). 96% of the patients received 600 mg orally, once daily, had metastatic disease, and all the patients had Trk fusion proteins detected by Next-Generation Sequencing (NGS) and nucleic acid-based test. The ORR of patients according to the assessment was 57% (95% CI, 43–71), and the ORR by Trk fusion partners showed ENT6-NTRK3 (68%), TPM3-NTRK1 (50%), and TPP-NTRK1 (100%). Moreover, Entrectinib shows a partial response to LMNA-NTRK1 and SQSTM1-NTRK1 fusion proteins [156].

2.5.3. Resistance to Larotrectinib and Entrectinib as Trk Inhibitor

Although there was a high overall response rate to Trk fusion proteins-positive solid tumors, acquired drug resistance was identified as a progressive disease after the administration of Trk inhibitors. The emergence of amino acid substitution in Trk represents a significant resistance mechanism against the Trk inhibitor. In the case of a patient with ETV6-NTRK3-positive mammary analog secretory carcinoma (MASC), treatment with Entrectinib showed a dramatic and durable response with an 89% reduction in tumor burden but revealed further disease progression in the right lower lobe of the lung. The NTRK3 G623R mutation mediated this case of resistance to Entrectinib. This alteration interfered with Entrectinib binding and conferred dramatically reduced sensitivity to Entrectinib inhibition, thereby increasing the IC50 value more than 250-fold. Moreover, NTRK3 G623R mutation conferred an increase in the IC50 value more than 500-fold for Larotrectinib and TSR-011 [157]. Additionally, LMNA-NTRK1-positive patients with metastatic colorectal cancer showed a remarkable response to Entrectinib, followed by resistance to Entrectinib mediated by NTRK1 G595R and G667C mutations and these mutations also showed immense resistance to Larotrectinib and TSR-011 [158].
Additionally, 11% of the patients showed further disease progression during the treatment with Larotrectinib after reported objective response or stable disease. Moreover, in tumor samples from 10 patients, amino acid substitution of the kinase domain was observed involving the solvent front position (TrkA G595R or TrkC G623R), the gatekeeper position (TrkA F589L), and the xDFG position (TrkA G667S or TrkC G696A) which eliminates unfavorable interactions by preventing inhibitor binding. As a result of this, eight out of 10 patients identified acquired resistance to Larotrectinib. Moreover, more than one mutation was identified in three patients [88]. These findings suggest that the biochemical characterization of more than 370 TrkC mutations, including a protein tyrosine kinase, is required to determine the potential mechanism of primary resistance to Trk inhibitors.

2.6. Next-Generation of Trk Inhibitor

Repotrectinib (TPX-0005) and LOXO-195 were developed to overcome acquired drug-resistance mutations, including solvent front and xDFG mutations of the kinase domain.
In terms of cell viability, Repotrectinib had a more potent IC50 value (<0.2 nmol/L) compared to that of Larotrectinib and Entrectinib. Moreover, TrkA G595R, TrkB G639R, TrkC G623R, and TrkC G623E exhibited a dramatic reduction in sensitivity to inhibition by Larotrectinib and Entrectinib, but Repotrectinib has a 42- and 62-fold increase, respectively, in the IC50 value compared to Larotrectinib and Entrectinib. Moreover, treatment with Repotrectinib for ETV6-NTRK3 and TrkC G623E mutation-positive patient with MASC showed a rapid and dramatic response to the drug and achieved regression within a few days of treatment [159].
LOXO-195 is another next-generation Trk kinase inhibitor designed to overcome recurrent resistance mediated by mutation of the gatekeeper position and the kinase domain (solvent front and xDFG). Treatment with LOXO-195 showed low inhibitory activity against TrkA G595R, TrkC G628R, and TrkA G667C, with IC50 values between 2.0 and 9.8 nmol/L. Moreover, LOXO-195 treatment of 2 patients who acquired resistance to Larotrectinib through TRKA G595R, and TRKC G623R, respectively, showed at least 90% inhibition of TRK target coverage at Cmax and 50% inhibition at Cmin of target pharmacokinetic thresholds (Cmax, Cmin). Furthermore, LMNA-NTRK1-positive colon cancer patients with TrkA G595R mutation displayed a rapid clinical response to therapy with LOXO-195. Moreover, ETV6-NTRK3 and TrkC G623R-positive pediatric patients with infantile fibrosarcoma showed visible tumor regression in the head and neck region [160]. Additionally, LOXO-195 is currently being evaluated in a Phase 1/2 trial. Of 29 evaluable patients via Phase I or FDA expanded access single patient protocol, 10 of these patients (34%) had a confirmed complete or partial response, and the ORR of patients with Trk kinase mutation according to the assessment was 45%. Moreover, nine out of 20 patients (45%) with a previous resistance to Larotrectinib treatment showed a complete or partial response to LOXO-195 [161].

3. Conclusions

TrkC and TrkC fusion proteins are associated with a variety of human cancers and play essential roles in the progression and metastasis of human cancers. The development of Larotrectinib and Rozlytek as Trk inhibitors has demonstrated a high overall response rate to Trk or Trk fusion proteins-positive solid tumors. However, acquired drug resistance was identified as a progressive disease by mutation, including mutations in the gatekeeper position and the kinase domain (solvent front and xDFG), still remains challenging, although Repotrectinib (TPX-0005) and LOXO-195 were developed to overcome kinase domain mutations and possesses potent and selective activity against TRKA G595R, TrkA G667C, and TRKC G623R.
In patients of various cancer types, more than 530 different mutations of TrkC are identified, including a mutation in the tyrosine kinase domain (Figure 1A). These findings imply that mutations of TrkC in the kinase domain may lead to constitutive activation of TrkC, and this can lead to acquired resistance to first-generation or next-generation Trk inhibitors. To resolve this possibility, further studies will be required to determine the functional role of the TrkC mutation.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Segal, R.A. Selectivity in neurotrophin signaling: Theme and variations. Annu. Rev. Neurosci. 2003, 26, 299–330. [Google Scholar] [CrossRef] [PubMed]
  2. Reichardt, L.F. Neurotrophin-regulated signalling pathways. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2006, 361, 1545–1564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Urfer, R.; Tsoulfas, P.; O’Connell, L.; Hongo, J.A.; Zhao, W.; Presta, L.G. High resolution mapping of the binding site of TrkA for nerve growth factor and TrkC for neurotrophin-3 on the second immunoglobulin-like domain of the Trk receptors. J. Biol. Chem. 1998, 273, 5829–5840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Lad, S.P.; Neet, K.E.; Mufson, E.J. Nerve growth factor: Structure, function and therapeutic implications for Alzheimer’s disease. Curr. Drug Targets CNS Neurol. Disord. 2003, 2, 315–334. [Google Scholar] [CrossRef]
  5. Wiesmann, C.; de Vos, A.M. Nerve growth factor: Structure and function. Cell. Mol. Life Sci. 2001, 58, 748–759. [Google Scholar] [CrossRef]
  6. Kaplan, D.R.; Miller, F.D. Neurotrophin signal transduction in the nervous system. Curr. Opin. Neurobiol. 2000, 10, 381–391. [Google Scholar] [CrossRef]
  7. Chao, M.V.; Bothwell, M. Neurotrophins: To cleave or not to cleave. Neuron 2002, 33, 9–12. [Google Scholar] [CrossRef] [Green Version]
  8. Salehi, A.; Verhaagen, J.; Dijkhuizen, P.A.; Swaab, D.F. Co-localization of high-affinity neurotrophin receptors in nucleus basalis of Meynert neurons and their differential reduction in Alzheimer’s disease. Neuroscience 1996, 75, 373–387. [Google Scholar] [CrossRef] [Green Version]
  9. Ginsberg, S.D.; Che, S.; Wuu, J.; Counts, S.E.; Mufson, E.J. Down regulation of trk but not p75NTR gene expression in single cholinergic basal forebrain neurons mark the progression of Alzheimer’s disease. J. Neurochem. 2006, 97, 475–487. [Google Scholar] [CrossRef]
  10. Savaskan, E.; Muller-Spahn, F.; Olivieri, G.; Bruttel, S.; Otten, U.; Rosenberg, C.; Hulette, C.; Hock, C. Alterations in trk A, trk B and trk C receptor immunoreactivities in parietal cortex and cerebellum in Alzheimer’s disease. Eur. Neurol. 2000, 44, 172–180. [Google Scholar] [CrossRef]
  11. Ginsberg, S.D.; Alldred, M.J.; Counts, S.E.; Cataldo, A.M.; Neve, R.L.; Jiang, Y.; Wuu, J.; Chao, M.V.; Mufson, E.J.; Nixon, R.A.; et al. Microarray analysis of hippocampal CA1 neurons implicates early endosomal dysfunction during Alzheimer’s disease progression. Biol. Psychiatry 2010, 68, 885–893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Von Bohlen und Halbach, O.; Minichiello, L.; Unsicker, K. Haploinsufficiency for trkB and trkC receptors induces cell loss and accumulation of alpha-synuclein in the substantia Nigra. FASEB J. 2005, 19, 1740–1742. [Google Scholar] [CrossRef] [PubMed]
  13. Pacenta, H.L.; Macy, M.E. Entrectinib and other ALK/TRK inhibitors for the treatment of neuroblastoma. Drug Des. Dev. Ther. 2018, 12, 3549–3561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Cheung, N.K.; Dyer, M.A. Neuroblastoma: Developmental biology, cancer genomics and immunotherapy. Nat. Rev. Cancer 2013, 13, 397–411. [Google Scholar] [CrossRef] [Green Version]
  15. Brodeur, G.M.; Nakagawara, A.; Yamashiro, D.J.; Ikegaki, N.; Liu, X.G.; Azar, C.G.; Lee, C.P.; Evans, A.E. Expression of TrkA, TrkB and TrkC in human neuroblastomas. J. Neuro Oncol. 1997, 31, 49–55. [Google Scholar] [CrossRef]
  16. Bouzas-Rodriguez, J.; Cabrera, J.R.; Delloye-Bourgeois, C.; Ichim, G.; Delcros, J.G.; Raquin, M.A.; Rousseau, R.; Combaret, V.; Benard, J.; Tauszig-Delamasure, S.; et al. Neurotrophin-3 production promotes human neuroblastoma cell survival by inhibiting TrkC-induced apoptosis. J. Clin. Investig. 2010, 120, 850–858. [Google Scholar] [CrossRef]
  17. Lawn, S.; Krishna, N.; Pisklakova, A.; Qu, X.; Fenstermacher, D.A.; Fournier, M.; Vrionis, F.D.; Tran, N.; Chan, J.A.; Kenchappa, R.S.; et al. Neurotrophin signaling via TrkB and TrkC receptors promotes the growth of brain tumor-initiating cells. J. Biol. Chem. 2015, 290, 3814–3824. [Google Scholar] [CrossRef] [Green Version]
  18. Calatozzolo, C.; Salmaggi, A.; Pollo, B.; Sciacca, F.L.; Lorenzetti, M.; Franzini, A.; Boiardi, A.; Broggi, G.; Marras, C. Expression of cannabinoid receptors and neurotrophins in human gliomas. Neurol. Sci. 2007, 28, 304–310. [Google Scholar] [CrossRef]
  19. Tajima, Y.; Molina, R.P., Jr.; Rorke, L.B.; Kaplan, D.R.; Radeke, M.; Feinstein, S.C.; Lee, V.M.; Trojanowski, J.Q. Neurotrophins and neuronal versus glial differentiation in medulloblastomas and other pediatric brain tumors. Acta Neuropathol. 1998, 95, 325–332. [Google Scholar] [CrossRef]
  20. Kim, M.S.; Jeong, J.; Seo, J.; Kim, H.S.; Kim, S.J.; Jin, W. Dysregulated JAK2 expression by TrkC promotes metastasis potential, and EMT program of metastatic breast cancer. Sci. Rep. 2016, 6, 33899. [Google Scholar] [CrossRef]
  21. Llovet, J.M.; Montal, R.; Sia, D.; Finn, R.S. Molecular therapies and precision medicine for hepatocellular carcinoma. Nat. Rev. Clin. Oncol. 2018, 15, 599–616. [Google Scholar] [CrossRef] [PubMed]
  22. Jin, W.; Lee, J.J.; Kim, M.S.; Son, B.H.; Cho, Y.K.; Kim, H.P. DNA methylation-dependent regulation of TrkA, TrkB, and TrkC genes in human hepatocellular carcinoma. Biochem. Biophys. Res. Commun. 2011, 406, 89–95. [Google Scholar] [CrossRef] [PubMed]
  23. Hisaoka, M.; Sheng, W.Q.; Tanaka, A.; Hashimoto, H. Gene expression of TrkC (NTRK3) in human soft tissue tumours. J. Pathol. 2002, 197, 661–667. [Google Scholar] [CrossRef] [PubMed]
  24. Truzzi, F.; Marconi, A.; Lotti, R.; Dallaglio, K.; French, L.E.; Hempstead, B.L.; Pincelli, C. Neurotrophins and their receptors stimulate melanoma cell proliferation and migration. J. Investig. Dermatol. 2008, 128, 2031–2040. [Google Scholar] [CrossRef] [Green Version]
  25. Xu, X.; Tahan, S.R.; Pasha, T.L.; Zhang, P.J. Expression of neurotrophin receptor Trk-C in nevi and melanomas. J. Cutan. Pathol. 2003, 30, 318–322. [Google Scholar] [CrossRef]
  26. Schneider, M.B.; Standop, J.; Ulrich, A.; Wittel, U.; Friess, H.; Andren-Sandberg, A.; Pour, P.M. Expression of nerve growth factors in pancreatic neural tissue and pancreatic cancer. J. Histochem. Cytochem. 2001, 49, 1205–1210. [Google Scholar] [CrossRef] [Green Version]
  27. Miknyoczki, S.J.; Lang, D.; Huang, L.; Klein-Szanto, A.J.; Dionne, C.A.; Ruggeri, B.A. Neurotrophins and Trk receptors in human pancreatic ductal adenocarcinoma: Expression patterns and effects on in vitro invasive behavior. Int. J. Cancer 1999, 81, 417–427. [Google Scholar] [CrossRef]
  28. Satoh, F.; Mimata, H.; Nomura, T.; Fujita, Y.; Shin, T.; Sakamoto, S.; Hamada, Y.; Nomura, Y. Autocrine expression of neurotrophins and their receptors in prostate cancer. Int. J. Urol. 2001, 8, S28–S34. [Google Scholar] [CrossRef]
  29. Ricci, A.; Greco, S.; Mariotta, S.; Felici, L.; Bronzetti, E.; Cavazzana, A.; Cardillo, G.; Amenta, F.; Bisetti, A.; Barbolini, G. Neurotrophins and neurotrophin receptors in human lung cancer. Am. J. Respir. Cell Mol. Biol. 2001, 25, 439–446. [Google Scholar] [CrossRef]
  30. Kim, M.S.; Kim, G.M.; Choi, Y.J.; Kim, H.J.; Kim, Y.J.; Jin, W. TrkC promotes survival and growth of leukemia cells through Akt-mTOR-dependent up-regulation of PLK-1 and Twist-1. Mol. Cells 2013, 36, 177–184. [Google Scholar] [CrossRef] [Green Version]
  31. Kamiya, A.; Inokuchi, M.; Otsuki, S.; Sugita, H.; Kato, K.; Uetake, H.; Sugihara, K.; Takagi, Y.; Kojima, K. Prognostic value of tropomyosin-related kinases A, B, and C in gastric cancer. Clin. Transl. Oncol. 2016, 18, 599–607. [Google Scholar] [CrossRef] [PubMed]
  32. McGregor, L.M.; McCune, B.K.; Graff, J.R.; McDowell, P.R.; Romans, K.E.; Yancopoulos, G.D.; Ball, D.W.; Baylin, S.B.; Nelkin, B.D. Roles of trk family neurotrophin receptors in medullary thyroid carcinoma development and progression. Proc. Natl. Acad. Sci. USA 1999, 96, 4540–4545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Guidi, M.; Muinos-Gimeno, M.; Kagerbauer, B.; Marti, E.; Estivill, X.; Espinosa-Parrilla, Y. Overexpression of miR-128 specifically inhibits the truncated isoform of NTRK3 and upregulates BCL2 in SH-SY5Y neuroblastoma cells. BMC Mol. Biol. 2010, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Laneve, P.; Di Marcotullio, L.; Gioia, U.; Fiori, M.E.; Ferretti, E.; Gulino, A.; Bozzoni, I.; Caffarelli, E. The interplay between microRNAs and the neurotrophin receptor tropomyosin-related kinase C controls proliferation of human neuroblastoma cells. Proc. Natl. Acad. Sci. USA 2007, 104, 7957–7962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Ferretti, E.; De Smaele, E.; Po, A.; Di Marcotullio, L.; Tosi, E.; Espinola, M.S.B.; Di Rocco, C.; Riccardi, R.; Giangaspero, F.; Farcomeni, A.; et al. MicroRNA profiling in human medulloblastoma. Int. J. Cancer 2009, 124, 568–577. [Google Scholar] [CrossRef] [PubMed]
  36. Zhong, Y.; Wu, X.C.; Li, Q.P.; Ge, X.X.; Wang, F.; Wu, P.Y.; Deng, X.T.; Miao, L. Long noncoding RNAs as potential biomarkers and therapeutic targets in gallbladder cancer: A systematic review and meta-analysis. Cancer Cell Int. 2019, 19. [Google Scholar] [CrossRef]
  37. Zhao, S.; Fan, N.F.; Chen, X.H.; Zhuo, C.H.; Xu, C.W.; Lin, R.B. Long noncoding RNA PVT1-214 enhances gastric cancer progression by upregulating TrkC expression in competitively sponging way. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 4173–4184. [Google Scholar] [CrossRef]
  38. Bu, J.Y.; Lv, W.Z.; Liao, Y.F.; Xiao, X.Y.; Lv, B.J. Long non-coding RNA LINC00978 promotes cell proliferation and tumorigenesis via regulating microRNA-497/NTRK3 axis in gastric cancer. Int. J. Biol. Macromol. 2019, 123, 1106–1114. [Google Scholar] [CrossRef]
  39. Xiong, D.M.; Sheng, Y.R.; Ding, S.J.; Chen, J.; Tan, X.X.; Zeng, T.; Qin, D.D.; Zhu, L.Y.; Huang, A.L.; Tang, H. LINC00052 regulates the expression of NTRK3 by miR-128 and miR-485-3p to strengthen HCC cells invasion and migration. Oncotarget 2016, 7, 47593–47608. [Google Scholar] [CrossRef] [Green Version]
  40. Martincorena, I.; Campbell, P.J. Somatic mutation in cancer and normal cells. Science 2015, 349, 1483–1489. [Google Scholar] [CrossRef]
  41. Stratton, M.R. Exploring the genomes of cancer cells: Progress and promise. Science 2011, 331, 1553–1558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Bardelli, A.; Parsons, D.W.; Silliman, N.; Ptak, J.; Szabo, S.; Saha, S.; Markowitz, S.; Willson, J.K.; Parmigiani, G.; Kinzler, K.W.; et al. Mutational analysis of the tyrosine kinome in colorectal cancers. Science 2003, 300, 949. [Google Scholar] [CrossRef] [PubMed]
  44. Wood, L.D.; Calhoun, E.S.; Silliman, N.; Ptak, J.; Szabo, S.; Powell, S.M.; Riggins, G.J.; Wang, T.L.; Yan, H.; Gazdar, A.; et al. Somatic mutations of GUCY2F, EPHA3, and NTRK3 in human cancers. Hum. Mutat. 2006, 27, 1060–1061. [Google Scholar] [CrossRef] [PubMed]
  45. Jones, S.; Zhang, X.; Parsons, D.W.; Lin, J.C.; Leary, R.J.; Angenendt, P.; Mankoo, P.; Carter, H.; Kamiyama, H.; Jimeno, A.; et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 2008, 321, 1801–1806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Kubo, T.; Kuroda, Y.; Kokubu, A.; Hosoda, F.; Arai, Y.; Hiraoka, N.; Hirohashi, S.; Shibata, T. Resequencing analysis of the human tyrosine kinase gene family in pancreatic cancer. Pancreas 2009, 38, e200–e206. [Google Scholar] [CrossRef]
  47. Stephens, P.; Edkins, S.; Davies, H.; Greenman, C.; Cox, C.; Hunter, C.; Bignell, G.; Teague, J.; Smith, R.; Stevens, C.; et al. A screen of the complete protein kinase gene family identifies diverse patterns of somatic mutations in human breast cancer. Nat. Genet. 2005, 37, 590–592. [Google Scholar] [CrossRef]
  48. Davies, H.; Hunter, C.; Smith, R.; Stephens, P.; Greenman, C.; Bignell, G.; Teague, J.; Butler, A.; Edkins, S.; Stevens, C.; et al. Somatic mutations of the protein kinase gene family in human lung cancer. Cancer Res. 2005, 65, 7591–7595. [Google Scholar] [CrossRef] [Green Version]
  49. Kubo, T.; Kuroda, Y.; Shimizu, H.; Kokubu, A.; Okada, N.; Hosoda, F.; Arai, Y.; Nakamura, Y.; Taniguchi, H.; Yanagihara, K.; et al. Resequencing and copy number analysis of the human tyrosine kinase gene family in poorly differentiated gastric cancer. Carcinogenesis 2009, 30, 1857–1864. [Google Scholar] [CrossRef] [Green Version]
  50. Pickering, C.R.; Zhou, J.H.; Lee, J.J.; Drummond, J.A.; Peng, S.A.; Saade, R.E.; Tsai, K.Y.; Curry, J.L.; Tetzlaff, M.T.; Lai, S.Y.; et al. Mutational landscape of aggressive cutaneous squamous cell carcinoma. Clin. Cancer Res. 2014, 20, 6582–6592. [Google Scholar] [CrossRef] [Green Version]
  51. Shain, A.H.; Garrido, M.; Botton, T.; Talevich, E.; Yeh, I.; Sanborn, J.Z.; Chung, J.; Wang, N.J.; Kakavand, H.; Mann, G.J.; et al. Exome sequencing of desmoplastic melanoma identifies recurrent NFKBIE promoter mutations and diverse activating mutations in the MAPK pathway. Nat. Genet. 2015, 47, 1194–1199. [Google Scholar] [CrossRef] [PubMed]
  52. Bonilla, X.; Parmentier, L.; King, B.; Bezrukov, F.; Kaya, G.; Zoete, V.; Seplyarskiy, V.B.; Sharpe, H.J.; McKee, T.; Letourneau, A.; et al. Genomic analysis identifies new drivers and progression pathways in skin basal cell carcinoma. Nat. Genet. 2016, 48, 398–406. [Google Scholar] [CrossRef]
  53. Campbell, J.D.; Alexandrov, A.; Kim, J.; Wala, J.; Berger, A.H.; Pedamallu, C.S.; Shukla, S.A.; Guo, G.; Brooks, A.N.; Murray, B.A.; et al. Distinct patterns of somatic genome alterations in lung adenocarcinomas and squamous cell carcinomas. Nat. Genet. 2016, 48, 607–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Han, M.R.; Shin, S.; Park, H.C.; Kim, M.S.; Lee, S.H.; Jung, S.H.; Song, S.Y.; Lee, S.H.; Chung, Y.J. Mutational signatures and chromosome alteration profiles of squamous cell carcinomas of the vulva. Exp. Mol. Med. 2018, 50, e442. [Google Scholar] [CrossRef] [PubMed]
  55. Jordan, E.J.; Kim, H.R.; Arcila, M.E.; Barron, D.; Chakravarty, D.; Gao, J.; Chang, M.T.; Ni, A.; Kundra, R.; Jonsson, P.; et al. Prospective Comprehensive Molecular Characterization of Lung Adenocarcinomas for Efficient Patient Matching to Approved and Emerging Therapies. Cancer Discov. 2017, 7, 596–609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Sfakianos, J.P.; Cha, E.K.; Iyer, G.; Scott, S.N.; Zabor, E.C.; Shah, R.H.; Ren, Q.; Bagrodia, A.; Kim, P.H.; Hakimi, A.A.; et al. Genomic Characterization of Upper Tract Urothelial Carcinoma. Eur. Urol. 2015, 68, 970–977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Da Silva Almeida, A.C.; Abate, F.; Khiabanian, H.; Martinez-Escala, E.; Guitart, J.; Tensen, C.P.; Vermeer, M.H.; Rabadan, R.; Ferrando, A.; Palomero, T. The mutational landscape of cutaneous T cell lymphoma and Sezary syndrome. Nat. Genet. 2015, 47, 1465–1470. [Google Scholar] [CrossRef] [Green Version]
  58. Giannakis, M.; Mu, X.J.; Shukla, S.A.; Qian, Z.R.; Cohen, O.; Nishihara, R.; Bahl, S.; Cao, Y.; Amin-Mansour, A.; Yamauchi, M.; et al. Genomic Correlates of Immune-Cell Infiltrates in Colorectal Carcinoma. Cell Rep. 2016, 15, 857–865. [Google Scholar] [CrossRef] [Green Version]
  59. Cancer Genome Atlas Research Network; Analysis Working Group: Asan University; BC Cancer Agency; Brigham and Women’s Hospital; Broad Institute; Brown University; Case Western Reserve University; Dana-Farber Cancer Institute; Duke University; Greater Poland Cancer Centre; et al. Integrated genomic characterization of oesophageal carcinoma. Nature 2017, 541, 169–175. [Google Scholar] [CrossRef] [Green Version]
  60. Shern, J.F.; Chen, L.; Chmielecki, J.; Wei, J.S.; Patidar, R.; Rosenberg, M.; Ambrogio, L.; Auclair, D.; Wang, J.; Song, Y.K.; et al. Comprehensive genomic analysis of rhabdomyosarcoma reveals a landscape of alterations affecting a common genetic axis in fusion-positive and fusion-negative tumors. Cancer Discov. 2014, 4, 216–231. [Google Scholar] [CrossRef] [Green Version]
  61. Bailey, P.; Chang, D.K.; Nones, K.; Johns, A.L.; Patch, A.M.; Gingras, M.C.; Miller, D.K.; Christ, A.N.; Bruxner, T.J.; Quinn, M.C.; et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 2016, 531, 47–52. [Google Scholar] [CrossRef] [PubMed]
  62. Gingras, M.C.; Covington, K.R.; Chang, D.K.; Donehower, L.A.; Gill, A.J.; Ittmann, M.M.; Creighton, C.J.; Johns, A.L.; Shinbrot, E.; Dewal, N.; et al. Ampullary Cancers Harbor ELF3 Tumor Suppressor Gene Mutations and Exhibit Frequent WNT Dysregulation. Cell Rep. 2016, 14, 907–919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Armenia, J.; Wankowicz, S.A.M.; Liu, D.; Gao, J.; Kundra, R.; Reznik, E.; Chatila, W.K.; Chakravarty, D.; Han, G.C.; Coleman, I.; et al. Publisher Correction: The long tail of oncogenic drivers in prostate cancer. Nat. Genet. 2019, 51, 1194. [Google Scholar] [CrossRef]
  64. Lowery, M.A.; Ptashkin, R.; Jordan, E.; Berger, M.F.; Zehir, A.; Capanu, M.; Kemeny, N.E.; O’Reilly, E.M.; El-Dika, I.; Jarnagin, W.R.; et al. Comprehensive Molecular Profiling of Intrahepatic and Extrahepatic Cholangiocarcinomas: Potential Targets for Intervention. Clin. Cancer Res. 2018, 24, 4154–4161. [Google Scholar] [CrossRef] [Green Version]
  65. Li, M.L.; Zhang, Z.; Li, X.G.; Ye, J.Y.; Wu, X.S.; Tan, Z.J.; Liu, C.; Shen, B.Y.; Wang, X.A.; Wu, W.G.; et al. Whole-exome and targeted gene sequencing of gallbladder carcinoma identifies recurrent mutations in the ErbB pathway. Nat. Genet. 2014, 46, 872–876. [Google Scholar] [CrossRef]
  66. Cancer Genome Atlas Research Network. Integrated genomic characterization of papillary thyroid carcinoma. Cell 2014, 159, 676–690. [Google Scholar] [CrossRef]
  67. Landa, I.; Ibrahimpasic, T.; Boucai, L.; Sinha, R.; Knauf, J.A.; Shah, R.H.; Dogan, S.; Ricarte-Filho, J.C.; Krishnamoorthy, G.P.; Xu, B.; et al. Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers. J. Clin. Investig. 2016, 126, 1052–1066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Razavi, P.; Chang, M.T.; Xu, G.; Bandlamudi, C.; Ross, D.S.; Vasan, N.; Cai, Y.; Bielski, C.M.; Donoghue, M.T.A.; Jonsson, P.; et al. The Genomic Landscape of Endocrine-Resistant Advanced Breast Cancers. Cancer Cell 2018, 34, 427–438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Landau, D.A.; Tausch, E.; Taylor-Weiner, A.N.; Stewart, C.; Reiter, J.G.; Bahlo, J.; Kluth, S.; Bozic, I.; Lawrence, M.; Bottcher, S.; et al. Mutations driving CLL and their evolution in progression and relapse. Nature 2015, 526, 525–530. [Google Scholar] [CrossRef] [Green Version]
  70. Ceccarelli, M.; Barthel, F.P.; Malta, T.M.; Sabedot, T.S.; Salama, S.R.; Murray, B.A.; Morozova, O.; Newton, Y.; Radenbaugh, A.; Pagnotta, S.M.; et al. Molecular Profiling Reveals Biologically Discrete Subsets and Pathways of Progression in Diffuse Glioma. Cell 2016, 164, 550–563. [Google Scholar] [CrossRef]
  71. Zhang, W.; Lin, Z.C.; Zhang, T.X.; Liu, S.; Liu, X.; Liu, J.J.; Niu, Y. TrkC expression predicts favorable clinical outcome in invasive ductal carcinoma of breast independent of NT-3 expression. Am. J. Cancer Res. 2014, 4, 811–823. [Google Scholar] [CrossRef] [PubMed]
  72. Thomas, A.A.; Abrey, L.E.; Terziev, R.; Raizer, J.; Martinez, N.L.; Forsyth, P.; Paleologos, N.; Matasar, M.; Sauter, C.S.; Moskowitz, C.; et al. Multicenter phase II study of temozolomide and myeloablative chemotherapy with autologous stem cell transplant for newly diagnosed anaplastic oligodendroglioma. Neuro-Oncology 2017, 19, 1380–1390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Lohr, J.G.; Stojanov, P.; Carter, S.L.; Cruz-Gordillo, P.; Lawrence, M.S.; Auclair, D.; Sougnez, C.; Knoechel, B.; Gould, J.; Saksena, G.; et al. Widespread genetic heterogeneity in multiple myeloma: Implications for targeted therapy. Cancer Cell 2014, 25, 91–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. 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] [PubMed]
  75. Rubin, B.P.; Chen, C.J.; Morgan, T.W.; Xiao, S.; Grier, H.E.; Kozakewich, H.P.; Perez-Atayde, A.R.; Fletcher, J.A. Congenital mesoblastic nephroma t(12;15) is associated with ETV6-NTRK3 gene fusion: Cytogenetic and molecular relationship to congenital (infantile) fibrosarcoma. Am. J. Pathol. 1998, 153, 1451–1458. [Google Scholar] [CrossRef]
  76. Eguchi, M.; Eguchi-Ishimae, M.; Tojo, A.; Morishita, K.; Suzuki, K.; Sato, Y.; Kudoh, S.; Tanaka, K.; Setoyama, M.; Nagamura, F.; et al. Fusion of ETV6 to neurotrophin-3 receptor TRKC in acute myeloid leukemia with t(12;15)(p13;q25). Blood 1999, 93, 1355–1363. [Google Scholar] [CrossRef]
  77. Kralik, J.M.; Kranewitter, W.; Boesmueller, H.; Marschon, R.; Tschurtschenthaler, G.; Rumpold, H.; Wiesinger, K.; Erdel, M.; Petzer, A.L.; Webersinke, G. Characterization of a newly identified ETV6-NTRK3 fusion transcript in acute myeloid leukemia. Diagn. Pathol. 2011, 6, 19. [Google Scholar] [CrossRef] [Green Version]
  78. 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]
  79. Tognon, C.; Garnett, M.; Kenward, E.; Kay, R.; Morrison, K.; Sorensen, P.H. The chimeric protein tyrosine kinase ETV6-NTRK3 requires both Ras-Erk1/2 and PI3-kinase-Akt signaling for fibroblast transformation. Cancer Res. 2001, 61, 8909–8916. [Google Scholar]
  80. Wai, D.H.; Knezevich, S.R.; Lucas, T.; Jansen, B.; Kay, R.J.; Sorensen, P.H. The ETV6-NTRK3 gene fusion encodes a chimeric protein tyrosine kinase that transforms NIH3T3 cells. Oncogene 2000, 19, 906–915. [Google Scholar] [CrossRef] [Green Version]
  81. Lae, M.; Freneaux, P.; Sastre-Garau, X.; Chouchane, O.; Sigal-Zafrani, B.; Vincent-Salomon, A. Secretory breast carcinomas with ETV6-NTRK3 fusion gene belong to the basal-like carcinoma spectrum. Mod. Pathol. 2009, 22, 291–298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. 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]
  83. 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]
  84. 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]
  85. 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]
  86. 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]
  87. 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] [Green Version]
  88. 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]
  89. Nagasubramanian, R.; Wei, J.L.; Gordon, P.; Rastatter, J.C.; Cox, M.C.; Pappo, A. Infantile Fibrosarcoma With NTRK3-ETV6 Fusion Successfully Treated with the Tropomyosin-Related Kinase Inhibitor LOXO-101. Pediatr. Blood Cancer 2016, 63, 1468–1470. [Google Scholar] [CrossRef]
  90. Brenca, M.; Rossi, S.; Polano, M.; Gasparotto, D.; Zanatta, L.; Racanelli, D.; Valori, L.; Lamon, S.; Dei Tos, A.P.; Maestro, R. Transcriptome sequencing identifies ETV6-NTRK3 as a gene fusion involved in GIST. J. Pathol. 2016, 238, 543–549. [Google Scholar] [CrossRef]
  91. 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]
  92. Van der Tuin, K.; Ventayol Garcia, M.; Corver, W.E.; Khalifa, M.N.; Ruano Neto, D.; Corssmit, E.P.M.; Hes, F.J.; Links, T.P.; Smit, J.W.A.; Plantinga, T.S.; et al. Targetable gene fusions identified in radioactive iodine refractory advanced thyroid carcinoma. Eur. J. Endocrinol. 2019, 180, 235–241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Xu, B.; Aryeequaye, R.; Wang, L.; Katabi, N. Sinonasal Secretory Carcinoma of Salivary Gland with High Grade Transformation: A Case Report of this Under-Recognized Diagnostic Entity with Prognostic and Therapeutic Implications. Head Neck Pathol. 2018, 12, 274–278. [Google Scholar] [CrossRef] [PubMed]
  94. 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]
  95. Croce, S.; Hostein, I.; Longacre, T.A.; Mills, A.M.; Perot, G.; Devouassoux-Shisheboran, M.; Velasco, V.; Floquet, A.; Guyon, F.; Chakiba, C.; et al. Uterine and vaginal sarcomas resembling fibrosarcoma: A clinicopathological and molecular analysis of 13 cases showing common NTRK-rearrangements and the description of a COL1A1-PDGFB fusion novel to uterine neoplasms. Mod. Pathol. 2019, 32, 1008–1022. [Google Scholar] [CrossRef] [PubMed]
  96. Olson, N.; Rouhi, O.; Zhang, L.; Angeles, C.; Bridge, J.; Lopez-Terrada, D.; Royce, T.; Linos, K. A novel case of an aggressive superficial spindle cell sarcoma in an adult resembling fibrosarcomatous dermatofibrosarcoma protuberans and harboring an EML4-NTRK3 fusion. J. Cutan. Pathol. 2018, 45, 933–939. [Google Scholar] [CrossRef]
  97. Church, A.J.; Calicchio, M.L.; Nardi, V.; Skalova, A.; Pinto, A.; Dillon, D.A.; Gomez-Fernandez, C.R.; Manoj, N.; Haimes, J.D.; Stahl, J.A.; et al. Recurrent EML4-NTRK3 fusions in infantile fibrosarcoma and congenital mesoblastic nephroma suggest a revised testing strategy. Mod. Pathol. 2018, 31, 463–473. [Google Scholar] [CrossRef] [Green Version]
  98. Kao, Y.C.; Fletcher, C.D.M.; Alaggio, R.; Wexler, L.; Zhang, L.; Sung, Y.S.; Orhan, D.; Chang, W.C.; Swanson, D.; Dickson, B.C.; et al. Recurrent BRAF Gene Fusions in a Subset of Pediatric Spindle Cell Sarcomas: Expanding the Genetic Spectrum of Tumors with Overlapping Features with Infantile Fibrosarcoma. Am. J. Surg. Pathol. 2018, 42, 28–38. [Google Scholar] [CrossRef]
  99. Tannenbaum-Dvir, S.; Glade Bender, J.L.; Church, A.J.; Janeway, K.A.; Harris, M.H.; Mansukhani, M.M.; Nagy, P.L.; Andrews, S.J.; Murty, V.V.; Kadenhe-Chiweshe, A.; et al. Characterization of a novel fusion gene EML4-NTRK3 in a case of recurrent congenital fibrosarcoma. Mol. Case Stud. 2015, 1, a000471. [Google Scholar] [CrossRef] [Green Version]
  100. Kelly, L.M.; Barila, G.; Liu, P.Y.; Evdokimova, V.N.; Trivedi, S.; Panebianco, F.; Gandhi, M.; Carty, S.E.; Hodak, S.P.; Luo, J.H.; et al. Identification of the transforming STRN-ALK fusion as a potential therapeutic target in the aggressive forms of thyroid cancer. Proc. Natl. Acad. Sci. USA 2014, 111, 4233–4238. [Google Scholar] [CrossRef] [Green Version]
  101. Yang, Y.; Qin, S.K.; Zhu, J.; Wang, R.; Li, Y.M.; Xie, Z.Y.; Wu, Q. A Rare STRN-ALK Fusion in Lung Adenocarcinoma Identified Using Next-Generation Sequencing-Based Circulating Tumor DNA Profiling Exhibits Excellent Response to Crizotinib. Mayo Clin. Proc. Innov. Qual. Outcomes 2017, 1, 111–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Ling, Q.; Li, B.; Wu, X.; Wang, H.; Shen, Y.; Xiao, M.; Yang, Z.; Ma, R.; Chen, D.; Chen, H.; et al. 75P The landscape of NTRK fusions in Chinese patients with solid tumor. Ann. Oncol. 2018, 29 (Suppl. 8), mdy269.073. [Google Scholar] [CrossRef]
  103. Seethala, R.R.; Chiosea, S.I.; Liu, C.Z.; Nikiforova, M.; Nikiforov, Y.E. Clinical and Morphologic Features of ETV6-NTRK3 Translocated Papillary Thyroid Carcinoma in an Adult Population Without Radiation Exposure. Am. J. Surg. Pathol. 2017, 41, 446–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. 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]
  105. 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]
  106. Taylor, J.; Pavlick, D.; Yoshimi, A.; Marcelus, C.; Chung, S.S.; Hechtman, J.F.; Benayed, R.; Cocco, E.; Durham, B.H.; Bitner, L.; et al. Oncogenic TRK fusions are amenable to inhibition in hematologic malignancies. J. Clin. Investig. 2018, 128, 3819–3825. [Google Scholar] [CrossRef]
  107. Tallegas, M.; Fraitag, S.; Binet, A.; Orbach, D.; Jourdain, A.; Reynaud, S.; Pierron, G.; Machet, M.C.; Maruani, A. Novel KHDRBS1-NTRK3 rearrangement in a congenital pediatric CD34-positive skin tumor: A case report. Virchows Arch. 2019, 474, 111–115. [Google Scholar] [CrossRef]
  108. Yoshihara, K.; Wang, Q.; Torres-Garcia, W.; Zheng, S.; Vegesna, R.; Kim, H.; Verhaak, R.G.W. The landscape and therapeutic relevance of cancer-associated transcript fusions. Oncogene 2015, 34, 4845–4854. [Google Scholar] [CrossRef] [Green Version]
  109. Wang, L.; Busam, K.J.; Benayed, R.; Cimera, R.; Wang, J.; Denley, R.; Rao, M.; Aryeequaye, R.; Mullaney, K.; Cao, L.; et al. Identification of NTRK3 Fusions in Childhood Melanocytic Neoplasms. J. Mol. Diagn. 2017, 19, 387–396. [Google Scholar] [CrossRef]
  110. Doebele, R.C.; Davis, L.E.; Vaishnavi, A.; Le, A.T.; Estrada-Bernal, A.; Keysar, S.; Jimeno, A.; Varella-Garcia, M.; Aisner, D.L.; Li, Y.; et al. An Oncogenic NTRK Fusion in a Patient with Soft-Tissue Sarcoma with Response to the Tropomyosin-Related Kinase Inhibitor LOXO-101. Cancer Discov. 2015, 5, 1049–1057. [Google Scholar] [CrossRef] [Green Version]
  111. Friedman, B.J.; Hernandez, S.; Fidai, C.; Jiang, A.; Shwayder, T.A.; Carskadon, S.; Andea, A.A.; Harms, P.W.; Chitale, D.; Palanisamy, N. A pediatric case of pigmented epithelioid melanocytoma with chromosomal copy number alterations in 15q and 17q and a novel NTRK3-SCAPER gene fusion. J. Cutan. Pathol. 2019. [Google Scholar] [CrossRef] [PubMed]
  112. Torre, M.; Jessop, N.; Hornick, J.L.; Alexandrescu, S. Expanding the spectrum of pediatric NTRK-rearranged fibroblastic tumors to the central nervous system: A case report with RBPMS-NTRK3 fusion. Neuropathology 2018, 38, 624–630. [Google Scholar] [CrossRef] [PubMed]
  113. Chiang, S.; Cotzia, P.; Hyman, D.M.; Drilon, A.; Tap, W.D.; Zhang, L.; Hechtman, J.F.; Frosina, D.; Jungbluth, A.A.; Murali, R.; et al. NTRK Fusions Define a Novel Uterine Sarcoma Subtype with Features of Fibrosarcoma. Am. J. Surg. Pathol. 2018, 42, 791–798. [Google Scholar] [CrossRef] [PubMed]
  114. Iyama, K.; Matsuse, M.; Mitsutake, N.; Rogounovitch, T.; Saenko, V.; Suzuki, K.; Ashizawa, M.; Ookouchi, C.; Suzuki, S.; Mizunuma, H.; et al. Identification of Three Novel Fusion Oncogenes, SQSTM1/NTRK3, AFAP1L2/RET, and PPFIBP2/RET, in Thyroid Cancers of Young Patients in Fukushima. Thyroid 2017, 27, 811–818. [Google Scholar] [CrossRef]
  115. Lu, Z.; Zhang, Y.; Feng, D.; Sheng, J.; Yang, W.; Liu, B. Targeted next generation sequencing identifies somatic mutations and gene fusions in papillary thyroid carcinoma. Oncotarget 2017, 8, 45784–45792. [Google Scholar] [CrossRef] [PubMed]
  116. Farago, A.F.; Taylor, M.S.; Doebele, R.C.; Zhu, V.W.; Kummar, S.; Spira, A.I.; Boyle, T.A.; Haura, E.B.; Arcila, M.E.; Benayed, R.; et al. Clinicopathologic Features of Non-Small-Cell Lung Cancer Harboring an NTRK Gene Fusion. JCO Precis. Oncol. 2018, 2018. [Google Scholar] [CrossRef] [PubMed]
  117. Yamazaki, F.; Nakatani, F.; Asano, N.; Wakai, S.; Sekimizu, M.; Mitani, S.; Kubo, T.; Kawai, A.; Ichikawa, H.; Yoshida, A. Novel NTRK3 Fusions in Fibrosarcomas of Adults. Am. J. Surg. Pathol. 2019, 43, 523–530. [Google Scholar] [CrossRef]
  118. Michal, M.; Hajkova, V.; Skalova, A.; Michal, M. STRN-NTRK3-rearranged Mesenchymal Tumor of the Uterus: Expanding the Morphologic Spectrum of Tumors with NTRK Fusions. Am. J. Surg. Pathol. 2019, 43, 1152–1154. [Google Scholar] [CrossRef]
  119. Wong, D.; Yip, S.; Sorensen, P.H. Methods for Identifying Patients with Tropomyosin Receptor Kinase (TRK) Fusion Cancer. Pathol. Oncol. Res. 2019. [Google Scholar] [CrossRef] [Green Version]
  120. Chmielecki, J.; Bailey, M.; He, J.; Elvin, J.; Vergilio, J.A.; Ramkissoon, S.; Suh, J.; Frampton, G.M.; Sun, J.X.; Morley, S.; et al. Genomic Profiling of a Large Set of Diverse Pediatric Cancers Identifies Known and Novel Mutations across Tumor Spectra. Cancer Res. 2017, 77, 509–519. [Google Scholar] [CrossRef] [Green Version]
  121. Joshi, S.K.; Davare, M.A.; Druker, B.J.; Tognon, C.E. Revisiting NTRKs as an emerging oncogene in hematological malignancies. Leukemia 2019, 33, 2563–2574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Lemmon, M.A.; Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 2010, 141, 1117–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. American Association for the Advancement of Science. Erratum for the Review “Somatic mutation in cancer and normal cells” by I. Martincorena and P. J. Campbell. Science 2016, 351. [Google Scholar] [CrossRef] [Green Version]
  124. Libermann, T.A.; Nusbaum, H.R.; Razon, N.; Kris, R.; Lax, I.; Soreq, H.; Whittle, N.; Waterfield, M.D.; Ullrich, A.; Schlessinger, J. Amplification, enhanced expression and possible rearrangement of EGF receptor gene in primary human brain tumours of glial origin. Nature 1985, 313, 144–147. [Google Scholar] [CrossRef]
  125. Sigismund, S.; Avanzato, D.; Lanzetti, L. Emerging functions of the EGFR in cancer. Mol. Oncol. 2018, 12, 3–20. [Google Scholar] [CrossRef]
  126. Du, Z.; Lovly, C.M. Mechanisms of receptor tyrosine kinase activation in cancer. Mol. Cancer 2018, 17, 58. [Google Scholar] [CrossRef]
  127. Carraway, K.L., 3rd; Sweeney, C. EGF receptor activation by heterologous mechanisms. Cancer Cell 2002, 1, 405–406. [Google Scholar] [CrossRef] [Green Version]
  128. Kim, M.S.; Suh, K.W.; Hong, S.; Jin, W. TrkC promotes colorectal cancer growth and metastasis. Oncotarget 2017, 8, 41319–41333. [Google Scholar] [CrossRef]
  129. Pawson, T.; Gish, G.D.; Nash, P. SH2 domains, interaction modules and cellular wiring. Trends Cell Biol. 2001, 11, 504–511. [Google Scholar] [CrossRef]
  130. Jin, W.; Yun, C.; Hobbie, A.; Martin, M.J.; Sorensen, P.H.B.; Kim, S.J. Cellular transformation and activation of the phosphoinositide-3-kinase-Akt cascade by the ETV6-NTRK3 chimeric tyrosine kinase requires c-Src. Cancer Res. 2007, 67, 3192–3200. [Google Scholar] [CrossRef] [Green Version]
  131. Jin, W.; Yun, C.; Jeong, J.; Park, Y.; Lee, H.D.; Kim, S.J. c-Src is required for tropomyosin receptor kinase C (TrkC)-induced activation of the phosphatidylinositol 3-kinase (PI3K)-AKT pathway. J. Biol. Chem. 2008, 283, 1391–1400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Lannon, C.L.; Martin, M.J.; Tognon, C.E.; Jin, W.; Kim, S.J.; Sorensen, P.H.B. A highly conserved NTRK3 C-terminal sequence in the ETV6-NTRK3 oncoprotein binds the phosphotyrosine binding domain of insulin receptor substrate-1. An essential interaction for transformation. J. Biol. Chem. 2004, 279, 15706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Valastyan, S.; Weinberg, R.A. Tumor Metastasis: Molecular Insights and Evolving Paradigms. Cell 2011, 147, 275–292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Polyak, K.; Weinberg, R.A. Transitions between epithelial and mesenchymal states: Acquisition of malignant and stem cell traits. Nat. Rev. Cancer 2009, 9, 265–273. [Google Scholar] [CrossRef] [PubMed]
  135. Park, J.; Kim, J.; Park, B.; Yang, K.M.; Sun, E.J.; Tognon, C.E.; Sorensen, P.H.; Kim, S.J. Novel identification of STAT1 as a crucial mediator of ETV6-NTRK3-induced tumorigenesis. Oncogene 2018, 37, 2270–2284. [Google Scholar] [CrossRef] [PubMed]
  136. Hix, L.M.; Karavitis, J.; Khan, M.W.; Shi, Y.H.; Khazaie, K.; Zhang, M. Tumor STAT1 transcription factor activity enhances breast tumor growth and immune suppression mediated by myeloid-derived suppressor cells. J. Biol. Chem. 2013, 288, 11676–11688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Khodarev, N.N.; Minn, A.J.; Efimova, E.V.; Darga, T.E.; Labay, E.; Beckett, M.; Mauceri, H.J.; Roizman, B.; Weichselbaum, R.R. Signal transducer and activator of transcription 1 regulates both cytotoxic and prosurvival functions in tumor cells. Cancer Res. 2007, 67, 9214–9220. [Google Scholar] [CrossRef] [Green Version]
  138. Khodarev, N.N.; Beckett, M.; Labay, E.; Darga, T.; Roizman, B.; Weichselbaum, R.R. STAT1 is overexpressed in tumors selected for radioresistance and confers protection from radiation in transduced sensitive cells. Proc. Natl. Acad. Sci. USA 2004, 101, 1714–1719. [Google Scholar] [CrossRef] [Green Version]
  139. Weichselbaum, R.R.; Ishwaran, H.; Yoon, T.; Nuyten, D.S.A.; Baker, S.W.; Khodarev, N.; Su, A.W.; Shaikh, A.Y.; Roach, P.; Kreike, B.; et al. An interferon-related gene signature for DNA damage resistance is a predictive marker for chemotherapy and radiation for breast cancer. Proc. Natl. Acad. Sci. USA 2008, 105, 18490–18495. [Google Scholar] [CrossRef] [Green Version]
  140. Tripathi, V.; Sixt, K.M.; Gao, S.J.; Xu, X.; Huang, J.; Weigert, R.; Zhou, M.; Zhang, Y.E. Direct Regulation of Alternative Splicing by SMAD3 through PCBP1 Is Essential to the Tumor-Promoting Role of TGF-beta. Mol. Cell 2016, 64, 549–564. [Google Scholar] [CrossRef] [Green Version]
  141. Jin, W.; Yun, C.; Kwak, M.K.; Kim, T.A.; Kim, S.J. TrkC binds to the type II TGF-beta receptor to suppress TGF-beta signaling. Oncogene 2007, 26, 7684–7691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Jin, W.; Kim, B.C.; Tognon, C.; Lee, H.J.; Patel, S.; Lannon, C.L.; Maris, J.M.; Triche, T.J.; Sorensen, P.H.B.; Kim, S.J. The ETV6-NTRK3 chimeric tyrosine kinase suppresses TGF-beta signaling by inactivating the TGF-beta type II receptor. Proc. Natl. Acad. Sci. USA 2005, 102, 16239–16244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Jin, W.; Yun, C.; Kim, H.S.; Kim, S.J. TrkC binds to the bone morphogenetic protein type II receptor to suppress bone morphogenetic protein signaling. Cancer Res. 2007, 67, 9869–9877. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Epping, M.T.; Lunardi, A.; Nachmani, D.; Castillo-Martin, M.; Thin, T.H.; Cordon-Cardo, C.; Pandolfi, P.P. TSPYL2 is an essential component of the REST/NRSF transcriptional complex for TGF beta signaling activation. Cell Death Differ. 2015, 22, 1353–1362. [Google Scholar] [CrossRef] [PubMed]
  145. Wagoner, M.P.; Gunsalus, K.T.W.; Schoenike, B.; Richardson, A.L.; Friedl, A.; Roopra, A. The Transcription Factor REST Is Lost in Aggressive Breast Cancer. PLoS Genet. 2010, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Scott, L.J. Larotrectinib: First Global Approval. Drugs 2019, 79, 201–206. [Google Scholar] [CrossRef]
  147. 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]
  148. Liu, Q.; Schwaller, J.; Kutok, J.; Cain, D.; Aster, J.C.; Williams, I.R.; Gilliland, D.G. Signal transduction and transforming properties of the TEL-TRKC fusions associated with t(12;15)(p13;q25) in congenital fibrosarcoma and acute myelogenous leukemia. EMBO J. 2000, 19, 1827–1838. [Google Scholar] [CrossRef] [Green Version]
  149. 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]
  150. Laetsch, T.W.; DuBois, S.G.; Mascarenhas, L. 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]
  151. Geoerger, B.; van Tilburg, C.; DuBois, S.; Albert, C.; Federman, N.; Nagasubramanian, R.; Doz, F.; Orbach, D.; Bielack, S.; Shukla, N.; et al. Larotrectinib Efficacy and Safety in Pediatric Patients with TRK Fusion Cancer. Pediatr. Blood Cancer 2019, 66, S65–S66. [Google Scholar] [CrossRef]
  152. Shukla, N.; Roberts, S.S.; Baki, M.O.; Mushtaq, Q.; Goss, P.E.; Park, B.; Gundem, G.; Tian, K.; Geiger, H.; Redfield, K.; et al. Successful Targeted Therapy of Refractory Pediatric ETV6-NTRK3 Fusion-Positive Secretory Breast Carcinoma. JCO Precis. Oncol. 2017, 1. [Google Scholar] [CrossRef] [PubMed]
  153. 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]
  154. Al-Salama, Z.T.; Keam, S.J. Entrectinib: First Global Approval. Drugs 2019, 79, 1477–1483. [Google Scholar] [CrossRef]
  155. De Braud, F.G.; Niger, M.; Damian, S.; Bardazza, B.; Martinetti, A.; Pelosi, G.; Marrapese, G.; Palmeri, L.; Cerea, G.; Valtorta, E.; et al. Alka-372-001: First-in-human, phase I study of entrectinib—An oral pan-trk, ROS1, and ALK inhibitor—In patients with advanced solid tumors with relevant molecular alterations. J. Clin. Oncol. 2015, 33. [Google Scholar] [CrossRef]
  156. 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. OncoTargets Ther. 2019, 12, 3171–3179. [Google Scholar] [CrossRef] [Green Version]
  157. Drilon, A.; Li, G.; Dogan, S.; Gounder, M.; Shen, R.; Arcila, M.; Wang, L.; Hyman, D.M.; Hechtman, J.; Wei, G.; et al. What hides behind the MASC: Clinical response and acquired resistance to entrectinib after ETV6-NTRK3 identification in a mammary analogue secretory carcinoma (MASC). Ann. Oncol. 2016, 27, 920–926. [Google Scholar] [CrossRef]
  158. Russo, M.; Misale, S.; Wei, G.; Siravegna, G.; Crisafulli, G.; Lazzari, L.; Corti, G.; Rospo, G.; Novara, L.; Mussolin, B.; et al. Acquired Resistance to the TRK Inhibitor Entrectinib in Colorectal Cancer. Cancer Discov. 2016, 6, 36–44. [Google Scholar] [CrossRef] [Green Version]
  159. Drilon, A.; Ou, S.H.I.; Cho, B.C.; Kim, D.W.; Lees, J.; Lin, J.J.; Zhu, V.W.; Ahns, M.J.; Camidge, D.R.; Nguyen, J.; et al. Repotrectinib (TPX-0005) Is a Next-Generation ROS1/TRK/ALK Inhibitor That Potently Inhibit ROS1/TRK/ALK Solvent-Front Mutations. Cancer Discov. 2018, 8, 1227–1236. [Google Scholar] [CrossRef] [Green Version]
  160. 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]
  161. Hyman, D.; Kummar, S.; Farago, A.; Geoerger, B.; Mau-Sorensen, M.; Taylor, M.; Garralda, E.; Nagasubramanian, R.; Natheson, M.; Song, L.; et al. Abstract CT127: Phase I and expanded access experience of LOXO-195 (BAY 2731954), a selective next-generation TRK inhibitor (TRKi). Am. Assoc. Cancer Res. 2019. [Google Scholar] [CrossRef]
Figure 2. Diagram of TrkC fusion proteins identified in various cancers. In most TrkC fusion proteins, the carboxy-terminal of the TrkC protein, including the tyrosine kinase domain, fused with inframe to the amino-terminal binding partner. Nucleotide sequences indicate the breakpoint. CC: Coiled-coil, CC: Coiled-coil, HLH: Helix loop Helix, LRRCT: Leucine-rich repeat C-terminal domain, LRRNT: Leucine-rich repeat N-terminal domain, RRM: RNA recognition motif, BTB: protein-protein interaction, BACK: BTB and C-terminal Kelch, PB1: Phox and Bem1p, SH2: Src Homology 2, SH3: Src Homology 3, MYSc: Myosin motor, UBCs: Ubiquitin-conjugating enzyme E2, WH1: WASP homology region 1, ZnF: Zinc-binding, and TM: Transmembrane.
Figure 2. Diagram of TrkC fusion proteins identified in various cancers. In most TrkC fusion proteins, the carboxy-terminal of the TrkC protein, including the tyrosine kinase domain, fused with inframe to the amino-terminal binding partner. Nucleotide sequences indicate the breakpoint. CC: Coiled-coil, CC: Coiled-coil, HLH: Helix loop Helix, LRRCT: Leucine-rich repeat C-terminal domain, LRRNT: Leucine-rich repeat N-terminal domain, RRM: RNA recognition motif, BTB: protein-protein interaction, BACK: BTB and C-terminal Kelch, PB1: Phox and Bem1p, SH2: Src Homology 2, SH3: Src Homology 3, MYSc: Myosin motor, UBCs: Ubiquitin-conjugating enzyme E2, WH1: WASP homology region 1, ZnF: Zinc-binding, and TM: Transmembrane.
Cancers 12 00147 g002
Table 3. Identification of TrkC fusion protein in cancer.
Table 3. Identification of TrkC fusion protein in cancer.
Fusion ProteinChromosomal LocationRecurrent Chromosomal TranslocationTumor Type
AFAP1-NTRK34p16.1t(4;15)(p16;q25)Glioblastoma [84]
AKAP13-NTRK315q25.3t(15;15)(q25;q25)Lung adenocarcinoma [102], Low-grade glioma [94]
BTBD1-NTRK315q25.2t(15;15)(q25;q25)Glioma [85]
CPEB1-NTRK315q25t(15;15)(q25;q25)Glioma [85]
EML4-NTRK32p21t(2;15)(p21;q25)Uterine and vaginal sarcomas [95], Dermatofibrosarcoma [96], Infantile fibrosarcoma and Congenital mesoblastic nephroma [97,99], Infantile fibrosarcomas [98], Glioblastoma [84].
ETV6-NTRK312p13.2t(12;15)(p13;q25)Congenital fibrosarcomas [74,75], Acute myeloid leukemia [76,77], Cellular mesoblastic nephroma [75,77,78], Secretory breast carcinoma [81,82], Colorectal cancer [83,84], Glioma [85,86], Spitz tumor [87], Lung adenocarcinoma [88], Infantile fibrosarcoma [88,89], Gastrointestinal stromal tumor [88,90], Thyroid carcinoma [103], Uterine sarcoma [86], Sinonasal adenocarcinoma [93], thyroid carcinomas [91,92], Mammary analog secretory carcinoma [104]
FAT1-NTRK34q35.2t(4;15)(q35;q25)Cervical squamous cell carcinoma (TCGA Dataset), [105]
HNRNPA2B1-NTRK37p15.2t(7;15)(p15;q25)Multiple myeloma [106]
KHDRBS1-NTRK31p35.2t(1;15)(p35;q25)Pediatric cutaneous congenital skin cancer [107]
LYN-NTRK38q12t(8;15)(q12;q25)Head and Neck squamous cell carcinoma (TCGA Dataset), [87,108]
MYH9-NTRK322q12.3t(22;15)(q12;q25)Spitz tumor [87]
MYO5A-NTRK315q21.2t(15;15)(q21;q25)Spitzoid tumor [87], Epithelioid melanocytic tumor [109]
NTRK3-HOMER215q25.2t(15;15)(q25;q25)Soft tissue sarcoma [110]
NTRK3-SCAPER15q24.3t(15;15)(q24;q25)Epithelioid melanocytoma [111]
TPM4-NTRK319p13.12t(19;15)(p13;q25)Sarcoma [84]
ZNF710-NTRK315q26t(15;15)(q26;q25)Glioblastoma [84]
RBPMS-NTRK38p12t(8;15)(p12;q25)Glioma [112], Uterine Sarcoma [113], Thyroid carcinoma [66]
SPECC1L-NTRK322q11.23t(22;15)(q11;q25)Uterine sarcoma [86]
SQSTM1-NTRK35q35.3t(5;15)(q35;q25)Thyroid Cancer [114,115], Non-small-cell lung cancer [86,116]
STRN-NTRK32p22.2t(2;15)(p22;q25)Fibrosarcoma [117], Uterine sarcoma [118]
STRN3-NTRK314q12t(14;15)(q12;q25)Fibrosarcoma [117]
TFG-NTKR33q12.2t(3;15)(q12;q25)Solitary fibrous tumor [119,120]
UBE2R2-NTRK39p13.3t(9;15)(p13;q25)Multiple myeloma [106,121]
VIM-NTRK310q13t(10;15)(q13;q25)Thyroid carcinoma [86]
VPS18-NTRK315q15t(15;15)(q15;q25)Colon Adenocarcinoma (TCGA Dataset) [105]

Share and Cite

MDPI and ACS Style

Jin, W. Roles of TrkC Signaling in the Regulation of Tumorigenicity and Metastasis of Cancer. Cancers 2020, 12, 147. https://doi.org/10.3390/cancers12010147

AMA Style

Jin W. Roles of TrkC Signaling in the Regulation of Tumorigenicity and Metastasis of Cancer. Cancers. 2020; 12(1):147. https://doi.org/10.3390/cancers12010147

Chicago/Turabian Style

Jin, Wook. 2020. "Roles of TrkC Signaling in the Regulation of Tumorigenicity and Metastasis of Cancer" Cancers 12, no. 1: 147. https://doi.org/10.3390/cancers12010147

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