Next Article in Journal
Can Nitric Oxide-Based Therapy Be Improved for the Treatment of Cancers? A Perspective
Previous Article in Journal
Antibody Cross-Reactivity in Auto-Immune Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

ALK, ROS1, RET and NTRK1–3 Gene Fusions in Colorectal and Non-Colorectal Microsatellite-Unstable Cancers

by
Rimma S. Mulkidjan
1,
Evgeniya S. Saitova
1,
Elena V. Preobrazhenskaya
1,2,
Karimat A. Asadulaeva
1,
Mikhail G. Bubnov
1,
Ekaterina A. Otradnova
1,
Darya M. Terina
1,
Sofia S. Shulga
1,
Darya E. Martynenko
1,
Maria V. Semina
1,
Evgeniya V. Belogubova
1,
Vladislav I. Tiurin
1,
Priscilla S. Amankwah
1,
Aleksandr S. Martianov
1,2 and
Evgeny N. Imyanitov
1,2,*
1
Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
2
Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(17), 13610; https://doi.org/10.3390/ijms241713610
Submission received: 14 August 2023 / Revised: 31 August 2023 / Accepted: 31 August 2023 / Published: 2 September 2023
(This article belongs to the Section Molecular Oncology)

Abstract

:
This study aimed to conduct a comprehensive analysis of actionable gene rearrangements in tumors with microsatellite instability (MSI). The detection of translocations involved tests for 5′/3′-end expression imbalance, variant-specific PCR and RNA-based next generation sequencing (NGS). Gene fusions were detected in 58/471 (12.3%) colorectal carcinomas (CRCs), 4/69 (5.8%) gastric cancers (GCs) and 3/65 (4.6%) endometrial cancers (ECs) (ALK: 8; RET: 12; NTRK1: 24; NTRK2: 2; NTRK3: 19), while none of these alterations were observed in five cervical carcinomas (CCs), four pancreatic cancers (PanCs), three cholangiocarcinomas (ChCs) and two ovarian cancers (OCs). The highest frequency of gene rearrangements was seen in KRAS/NRAS/BRAF wild-type colorectal carcinomas (53/204 (26%)). Surprisingly, as many as 5/267 (1.9%) KRAS/NRAS/BRAF-mutated CRCs also carried tyrosine kinase fusions. Droplet digital PCR (ddPCR) analysis of the fraction of KRAS/NRAS/BRAF mutated gene copies in kinase-rearranged tumors indicated that there was simultaneous co-occurrence of two activating events in cancer cells, but not genetic mosaicism. CRC patients aged above 50 years had a strikingly higher frequency of translocations as compared to younger subjects (56/365 (15.3%) vs. 2/106 (1.9%), p = 0.002), and this difference was particularly pronounced for tumors with normal KRAS/NRAS/BRAF status (52/150 (34.7%) vs. 1/54 (1.9%), p = 0.001). There were no instances of MSI in 56 non-colorectal tumors carrying ALK, ROS1, RET or NTRK1 rearrangements. An analysis of tyrosine kinase gene translocations is particularly feasible in KRAS/NRAS/BRAF wild-type microsatellite-unstable CRCs, although other categories of tumors with MSI also demonstrate moderate occurrence of these events.

1. Introduction

Microsatellite instability (MSI), being a consequence of deficient mismatch repair (dMMR), is manifested by multiple mutations affecting repetitive DNA sequences [1,2]. MSI may occur in tumors associated with Lynch hereditary cancer syndrome. These malignancies arise in subjects with inherited pathogenic variants in the MLH1, MSH2, MSH6, PMS2 or EPCAM genes, and their development involves a somatic second-hit inactivation of the involved member of the dMMR pathway [3]. MSI is also characteristic of some sporadic malignancies, being attributed to MLH1 promoter hypermethylation [4]. Microsatellite-unstable carcinomas have an increased tumor mutation burden (TMB) and are responsive to inhibitors of immune checkpoints [5,6].
MSI is particularly common for colorectal carcinomas, with approximately 5–15% of tumors displaying this phenotype. Microsatellite-unstable CRCs often carry mutations leading to MAPK signaling pathway activation, particularly amino acid substitutions in the KRAS, NRAS and BRAF genes [4,7]. Somewhat unexpectedly, MSI-CRCs were repeatedly shown to contain rearrangements in genes encoding receptor tyrosine kinases [2,8,9,10]. A recent study revealed that these translocations are related to an increased frequency of mutations within the G:C-reach intronic regions of the involved genes [11]. There are several drugs targeting ALK, ROS1, RET and NTRK1–3 tyrosine kinases; therefore, the detection of these gene fusions is of high clinical importance [10,12]. In addition to CRC, MSI is characteristic of several non-colorectal cancer types, particularly gastric and endometrial carcinomas [13,14]. It has not yet been systematically studied whether tyrosine kinase gene rearrangements occur at noticeable frequencies in microsatellite-unstable cancers arising in organs other than the colon.
A comprehensive analysis of ALK, ROS1, RET and NTRK1–3 gene rearrangements requires RNA-based next generation sequencing, which is an expensive technique [2]. We have developed an efficient laboratory screening procedure for ALK, ROS1, RET and NTRK1–3 translocations, which is largely based on an analysis of 5′/3′-end unbalanced expression of these genes [15,16]. When the gene is not affected, the number of transcripts corresponding to its kinase portion and to the upstream nucleotide sequences are equal. ALK, ROS1, RET and NTRK1–3 rearrangements usually result in fusion of the kinase domain to the actively transcribed gene. In the latter case, the expression of the kinase-domain-related portion of the receptor tyrosine kinase is elevated as compared to sequences located upstream to the breakpoint. This pipeline, being coupled with the identification of the ALK, ROS1, RET and NTRK1–3 fusion variants, allows for an analysis of a large number of tumor samples

2. Results

2.1. MSI Detection

MSI was detected in 743/14,111 (5.3%) CRCs, 84/1756 (4.8%) GCs, 91/506 (18%) ECs, 7/459 (1.5%) cervical carcinomas, 4/64 (6.3%) cholangiocarcinomas, 5/339 (1.5%) ovarian cancers and 4/474 (0.8%) pancreatic cancers (Supplementary Table S1). These frequencies are in good agreement with the published data [1]. Since we utilized both single-marker BAT26 testing and the pentaplex panel (BAT25, BAT26, NR21, NR22 and NR24), we evaluated whether the differences between these two methods are likely to influence the results. We considered the BAT26 length in 471 CRCs, 69 GCs and 66 ECs that were microsatellite-unstable by the pentaplex assay. Strikingly, only 3 out of 586 of these tumors had normal BAT26 status, indicating that the methodology of MSI testing had no significant influence on the results.

2.2. Analysis of Gene Rearrangements

An analysis of gene rearrangements was considered for 659 tumors carrying MSI (501 CRCs, 73 GCs, 69 ECs, 6 CCs, 4 ChCs, 4 PanCs and 2 OCs). There were 40/659 (6.1%) cases that failed to pass the RNA quality controls, as the referee gene in these samples emerged after the 35th PCR cycle (30/501 (6.0%) CRCs, 4/73 (5.5%) GCs, 4/69 (5.8%) ECs, 1/6 (16.7%) CCs and 1/4 (25%) ChCs). The remaining 619 MSI-positive samples (471 CRCs, 69 GCs, 65 ECs, 5 CCs, 4 PanCs, 3 ChCs and 2 OCs) were subjected to an analysis of gene fusions.
The 5′/3′-end expression imbalance for receptor tyrosine kinases was observed in 59 tumors (ALK: 8; RET: 8; NTRK1: 24; NTRK2: 2; NTRK3: 17). All these samples were available for variant-specific PCR, and the latter procedure resulted in the identification of rearrangements in 28/59 (47.5%) samples. Variant-specific PCR failed to reveal translocations in 31/59 (52.5%) tumors with the 5′/3′-end unbalanced expression. Only 13 of these tumors were analyzed by NGS, as the majority of samples were returned to primary hospitals after the completion of MSI testing and, therefore, were not available for the new round of RNA isolation. Strikingly, NGS revealed gene translocations in all these tumors (SPTBN1::ALK (S7;A20) (n = 2); ETV6::NTRK2 (E5;N15) (n = 2); NCOA4::RET (N9;R12) (n = 2) and one each NCOA4::RET (N9del501;R12); STRN::ALK (S5;ins53A20); RBBP8::ALK (R8;A20); TPR::NTRK1 (T15;N10); TPR::NTRK1 (T20;N11); ZKSCAN1::NTRK1 (Z3;N12) and LMNA::NTRK1 (L9ins44;del74N11)) (Table 1). Taken together, the above data allow for the assumption that the majority of the remaining 18 tumors, which were not available for NGS, also carry rare tyrosine kinase gene rearrangements.
Our screening procedure involved simultaneous determination of 5′/3′-end unbalanced expression and the use of variant-specific PCR for common translocations. All tumors carrying fusions in the ALK, NTRK1 and NTRK2 kinases demonstrated expression imbalance. However, the 5′/3′-end expression had relatively poor performance for RET and NTRK3 gene fusions, as 4/9 (44.4%) and 2/11 (18.2%) tumors with rearrangements of these genes, respectively, showed normal expression of these genes.
In addition to the above-mentioned well-known gene rearrangements, we performed an analysis of novel fusions (BCR::PKHD1 and CLIP1::LTK) that were recently identified in lung carcinomas [17,18]. None of the microsatellite-unstable carcinomas carried these translocations.

2.3. Pattern of Gene Rearrangement in Various Categories of Microsatellite-Unstable Tumors

There were 47 microsatellite-unstable tumors with identified translocation variants (ALK: 6; RET: 9; NTRK1: 19; NTRK2: 2; NTRK3: 11) (Figure 1). In addition, there were 18 tumors with 5′/3′-end unbalanced expression (ALK: 2; RET: 3; NTRK1: 5; NTRK3: 8), which were highly likely to carry rearrangements in the above genes, but the involved translocation variants could not be identified due to the above-mentioned technical limitations.
The frequency of gene fusions in microsatellite-unstable tumors approached 58/471 (12.3%) for CRC, 4/69 (5.8%) for GC and 3/65 (4.6%) for EC. The study included 204 CRCs negative for mutations in KRAS, NRAS or BRAF oncogenes; when considering samples with identified translocation variants, 42 (20.6%) of these tumors carried gene rearrangements (ALK: 6; RET: 9; NTRK1: 16; NTRK2: 1; NTRK3: 10). In addition, 11 KRAS/NRAS/BRAF mutation-negative CRCs demonstrated 5′/3′-end expression imbalances for one of the receptor tyrosine kinases (ALK: 2; RET: 3; NTRK1: 3; NTRK3: 3); however, the fusion variants could not be identified due to technical reasons. In total, 53/204 (26%) KRAS/NRAS/BRAF mutation-negative CRCs appeared to carry actionable gene fusions (ALK: 8; RET: 12; NTRK1: 19; NTRK2: 1; NTRK3: 13).
Somewhat surprisingly, as many as 5/267 (1.9%) CRCs with activating mutations in the KRAS, NRAS or BRAF genes were found to have kinase gene rearrangements (Figure 2, Table 1). In addition, the gastric tumor with TPM3::NTRK1 (T8;N10) rearrangement also simultaneously carried the p.G12C mutation in the KRAS oncogene. Activating genetic lesions in genes involved in the MAPK pathway are usually mutually exclusive; therefore, the coincident occurrence of gene fusions and KRAS/NRAS/BRAF mutations is intriguing. This coincidence may occur due to the presence of the above events in distinct cells, i.e., the mosaicism of activating genetic lesions, or due to the simultaneous occurrence of two mutations in the same cell. We evaluated the fraction of KRAS/BRAF-mutated cells in these tumors using digital droplet PCR (Supplementary Table S2). The obtained data were highly concordant with the results of the visual inspection of the slides, thus suggesting that KRAS/BRAF mutations are not mosaic but present in all tumor cells. It is noteworthy that all but one of the above-described tumors with tyrosine kinase rearrangements showed 5′/3′-end unbalanced expression of the affected gene; this imbalance could not be detected if only minor fractions of the tumor cells carried a fusion (Table 1). Furthermore, the only tumor with a non-altered expression pattern carried a translocation in the NTRK3 gene; as mentioned above, alterations in this kinase are not always accompanied by changes in the 5′/3′-end transcript ratio.
A high number of microsatellite-unstable CRCs carrying gene rearrangements permitted the analysis of clinical correlations. Male/female ratios were similar in fusion-positive and fusion-negative tumors (Supplementary Table S3, Figure S1). Patients aged above 50 years had a strikingly higher frequency of translocations as compared to younger subjects (56/365 (15.3%) vs. 2/106 (1.9%), p = 0.002). This difference was even more pronounced for tumors with normal KRAS/NRAS/BRAF status (52/150 (34.7%) vs. 1/54 (1.9%), p = 0.001; Supplementary Figures S2–S4).

2.4. MSI Analysis in Non-Colorectal Tumors Carrying ALK/ROS/RET/NTRK Rearrangements

This and other studies have demonstrated that a subset of microsatellite-unstable tumors carries tyrosine kinase gene fusions. We questioned whether the same overlap between these two events is observed when the MSI testing is applied to non-colorectal kinase-rearranged carcinomas. We utilized pentaplex panel testing for 23 lung carcinomas, 23 sarcomas, 4 thyroid carcinomas, 3 salivary gland tumors, and 3 pancreatic carcinomas with oncogenic gene translocations (ALK: 31; ROS1 10; RET: 9; NTRK1: 2; NTRK3: 4). None of the above tumors demonstrated microsatellite alterations.

3. Discussion

This is apparently the largest single-center study to systematically evaluate gene rearrangements in microsatellite-unstable tumors belonging to various cancer types. It has been confirmed that ALK, RET and NTRK1–3 gene rearrangements are very frequent in KRAS/NRAS/BRAF mutation-negative CRCs, especially in patients aged above 50 years, although these events may also occur in colorectal tumors carrying RAS/RAF mutations as well as in non-colorectal MSI-positive CRCs.
The results of this report are consistent with a recent study by Madison et al. [11], who demonstrated that the emergence of gene rearrangements in CRCs is attributed to the mutagenic effects of microbiota-derived butyrate and the consequent generation of 8-oxoguanine. These data convincingly explain the significant differences in the incidence of gene rearrangements between colorectal and non-colorectal malignancies. However, moderate frequencies of tyrosine kinase fusions were observed in gastric and endometrial tumors, suggesting that microsatellite-unstable cells may acquire translocations even in the absence of the influence of gut microbes.
Our report confirms that alterations in the genes belonging to the MAPK pathway are generally mutually exclusive. Indeed, activation of a single member of this molecular cascade, be it a receptor tyrosine kinase or KRAS, NRAS or BRAF oncogene, is usually sufficient to drive the signaling. Furthermore, the above-mentioned genetic alterations are usually considered to be more or less equivalent in terms of phenotypic consequences [7]. Interestingly, despite this apparent equivalence, the frequencies of alterations in particular genes vary considerably between MSI-positive and MSI-negative CRCs. Microsatellite-unstable CRCs have approximately a twice lower frequency of KRAS mutations but approximately a four times higher incidence of BRAF mutations as compared to MSI-negative tumors [7,19]. Furthermore, while kinase gene rearrangements are common in CRCs with MSI, they are exceptionally rare in MSI-negative colorectal carcinomas [11]. Overall, the cumulative frequency of the activation of the MAPK cascade is similar in CRCs with and without MSI; for example, 320/471 (67.9%) microsatellite-unstable CRCs analyzed in this study had evidence of genetic alteration of the MAPK pathway (Figure 2), which is very close to the estimates obtained for microsatellite-stable colorectal carcinomas [19]. The remaining 30–40% of CRCs do not have overt genetic alterations within this signaling cascade and deserve further investigations [7]. Interestingly, the analysis of our dataset revealed a few instances of the simultaneous occurrence of genetic events affecting two distinct oncogenes. Several prior investigations produced similar examples [19,20,21]. Single-cell sequencing may permit reliable discrimination between mutation mosaicism and the true co-occurrence of several MAPK activating events within the same cell [22]. Here, we utilized ddPCR for a rough analysis of the fraction of KRAS/BRAF-mutated cells, which strongly suggested that these mutations are truncal but not mosaic (Supplementary Table S2). Interestingly, there were two CRCs with simultaneous occurrences of BRAF p.V600E mutations and NTRK translocations. These tumors are unique with regard to clinical opportunities, as they have three highly actionable targets (MSI for immune therapy, BRAF p.V600E substitution for combined EGFR/BRAF inhibition and NTRK activation for the use of TRK inhibitors) [4,7,12].
Gene fusions occurred at high frequencies in CRC patients aged above 50 years, but were uncommon in younger subjects (Supplementary Tables S3 and S4). This age threshold is commonly utilized for discrimination between sporadic CRCs, which develop microsatellite instability due to somatic hypermethylation of the MLH1 promoter, and hereditary CRCs, which are attributed to the biallelic mutation-driven inactivation of DNA mismatch repair genes [3,4,7]. In this respect, tyrosine kinase translocations are similar to the BRAF p.V600E mutation, which is a validated marker for the exclusion of microsatellite-unstable CRCs from Lynch syndrome germline testing [3,7].
The spectrum of gene fusions in microsatellite-unstable tumors has some characteristic features. NTRK1–3 gene fusions are relatively frequent in some pediatric tumors and sarcomas, although they are exceptionally rare in common cancer types [23,24,25]. However, NTRK1–3 rearrangements compose the majority of the gene translocations observed in MSI-positive carcinomas. Similar to other tumor types [25], TPM3::NTRK1, EML4::NTRK3 and ETV6::NTRK3 fusion variants represented the majority of NTRK1–3 translocations identified in this study. ALK and RET gene rearrangements are particularly frequent in lung carcinomas. While common translocation variants constitute over 90% of ALK fusions in pulmonary malignancies [26], only one such rearrangement was detected in our dataset (EML4::ALK (E6;A20)). Similarly, KIF5B::RET fusions represent more than 70% of RET alterations in lung cancers [16]; however, none of the RET-rearranged microsatellite-unstable tumors carried this variant.
This study has some limitations. In particular, we did not have survival data for the patients; therefore, we could not evaluate whether the presence of gene rearrangements affects disease outcomes. MSI detection was based on a standard PCR protocol. Although this approach is reliable for the detection of tumors with highly unstable microsatellite repeats, it may miss a subset of mismatch repair-deficient carcinomas, which have low rates of cell proliferation and, therefore, low numbers of mutations in mononucleotide tracks [5,27]. The discordance between the immunohistochemical (IHC) evaluation of MMR proteins and the PCR analysis of MSI is mainly observed for tumors arising outside the gastrointestinal tract [5,27]. Therefore, given the preferential occurrence of gene fusions in CRCs, it is highly unlikely that these MMR-deficient but microsatellite-stable tumors would carry kinase-activating rearrangements.
This study detected 31 samples with 5′/3′-end unbalanced expression, in which variant-specific PCR failed to identify known gene rearrangements. Only 13 of these tumors were available for NGS, and all these samples contained rare fusion variants. It is highly likely that the majority of the remaining 18 tumors (ALK: 2; RET: 3; NTRK1: 5; NTRK3: 8) also carry uncommon types of rearrangements in the mentioned genes. The poor availability of microsatellite-unstable tumor samples for RNA-based NGS is attributed to the study design. The standard procedure for sample processing in our laboratory involves the simultaneous isolation of DNA and RNA, followed by cDNA synthesis in the same tube. This protocol is not compatible with subsequent RNA-based NGS sequencing; therefore, we needed to retrieve the tissue samples and subject them to a new round of RNA isolation. This effort turned out to be inefficient, as the majority of archival blocks were returned to the primary hospital immediately after the completion of standard molecular testing. This described drawback can be easily resolved if the aliquot of the DNA/RNA sample is stored without subsequent cDNA synthesis and, therefore, used for NGS whenever necessary. We have now incorporated this amendment in the processing of those tumors that may potentially require NGS testing. It also has to be acknowledged that the administration of ALK and NTRK inhibitors is not necessarily based on the identification of particular translocation variants, as the FISH assay is an approved method for the analysis of these genes [28]. Overall, 5′/3′-end unbalanced expression very rarely produces false-positive results, i.e., virtually all tumors identified by this assay indeed carry gene rearrangement [15,16]. Therefore, in theory, the results of 5′/3′-end unbalanced expression per se may be sufficient to guide therapy in some circumstances.

4. Materials and Methods

The analysis of MSI was applied to 14,111 CRCs, 1756 gastric carcinomas and 506 endometrial carcinomas, which were diagnosed on the basis of current World Health Organization (WHO) classification [29] and referred for molecular analysis to the N.N. Petrov Institute of Oncology (St. Petersburg, Russia) within the years 2013–2023. The majority of the CRCs included in this study, as well as a subset of GCs, were analyzed for mutations in the KRAS, NRAS and BRAF oncogenes [19]. In addition, MSI testing was applied to 459 cervical carcinomas, 64 cholangiocarcinomas, 339 ovarian cancers and 474 pancreatic cancers. In the years 2013–2021, microsatellite analysis was largely based on the use of a single marker, BAT26, given the evidence for its high accuracy for MSI detection [30]. In the years 2022–2023, this assay was replaced by the standard MSI test involving 5 markers (BAT25, BAT26, NR21, NR22 and NR24). MSI analysis was generally performed using tumor tissues only; in exceptionally rare instances of ambiguous results, corresponding normal cells were utilized for comparison. The primers and probes for these assays are described in Supplementary Table S5. For the pentaplex panel, tumors with two or more shifts were classified as MSI-positive [31]. Capillary electrophoresis was performed using the GenomeLab GeXP Genetic Analysis System (Beckman Coulter, Brea, CA, USA) or the Nanophore-05 instrument (Syntol, Moscow, Russia).
The detection of ALK, ROS1, RET and NTRK1–3 rearrangements was carried out for 619 microsatellite-unstable tumors. The methodology of the analysis of translocations in the above genes has been described in [15,16]. Briefly, tumor blocks were compared against corresponding histological slides, and areas with sufficient content of malignant cells were dissected from the specimens. The extraction of nucleic acids from manually dissected tumor cells involved the simultaneous isolation of both DNA and RNA, followed by cDNA synthesis. The quality of cDNA was controlled by PCR amplification of the SDHA-specific transcript; samples with a cycle threshold (Ct) above 35 were considered unreliable for further analysis. The ALK, ROS1, RET, NTRK1, NTRK2 and NTRK3 genes were subjected to tests for 5′/3′-end unbalanced expression. The primers and probes for these assays are described in Supplementary Table S6. In addition to the 5′/3′-end unbalanced expression screening test, all MSI-positive tumors were analyzed by variant-specific PCR for the most common translocations affecting the ALK (4 variants), ROS1 (10 variants) and RET (11 variants) genes, as well as BCR::PKHD1 and CLIP1::LTK gene fusions [17,18]. The design and multiplexing of the variant-specific tests are described in Supplementary Tables S7–S10. Tumors with unbalanced 5′/3′-end ALK, ROS1, RET, NTRK1, NTRK2 or NTRK3 expression, which lacked the above-mentioned common rearrangements, were further tested for rare translocation variants (see the list in Supplementary Tables S11–S15). In addition, we applied this testing for all MSI+ tumors with detectable RET and NTRK1–3 expression, even in the absence of 5′/3′-end expression imbalance. Finally, samples with unbalanced expression, which did not have PCR-detectable rearrangements, were subjected to RNA-based NGS.
PCR reactions were performed using the CFX-96 Real-Time PCR Detection System (Bio-Rad, Hercules, CA, USA). The PCR mix contained 1 μL of cDNA sample, 1× GeneAmp PCR Buffer I (Thermo Fisher Scientific Baltics UAB, Vilnius, Lithuania, cat. #4379876), 250 μM of each dNTP, 200 nM of each primer and probe, 2.5 mM MgCl2 and 1 U of TaqM polymerase (AlkorBio, St. Petersburg, Russia, cat. #751-100) in a total volume of 20 μL. The PCR reactions were initiated by enzyme activation (95 °C, 10 min) and included 38 cycles (95 °C for 15 s followed by 58 °C for 1 min).
NGS RNA sequencing was performed using the custom QIAseq RNA-Scan Targeted Panel (Qiagen, Hilden, Germany, cat. #CFHS10753Z-81), which is capable of detecting rearrangements in 6 genes (ALK, ROS1, RET, NTRK1, NTRK2 and NTRK3). This panel relies on the technology of Single Primer Extension (SPE). RNA was obtained using the PureLink FFPE RNA Isolation kit (Invitrogen, Carlsbad, CA, USA, cat. #45-7015). NGS was carried out on the Illumina MiSeq or NextSeq 550 instruments. Fusion calling was performed with the STAR-Fusion pipeline (V.1.4.0).

5. Conclusions

Overall, this study demonstrates that the comprehensive analysis of ALK, RET, NTRK1, NTRK2 and NTRK3 rearrangements is particularly feasible in MSI-positive KRAS/NRAS/BRAF mutation-negative CRCs, although there is also a moderate frequency of these events in other categories of microsatellite-unstable tumors. The spectrum of the involved tyrosine kinases and their partners is characterized by a high level of diversity; therefore, the utilization of indirect methods, such as IHC or FISH, may be associated with some uncertainty. While RNA-based NGS is the gold standard for the detection of gene rearrangements, the diagnostic pipeline presented here may be considered as a cost-efficient alternative for facilities with limited access to massive parallel sequencing.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms241713610/s1.

Author Contributions

Conceptualization, E.N.I., R.S.M. and E.V.P.; methodology, V.I.T., R.S.M. and M.G.B.; investigation, R.S.M., E.S.S., K.A.A., E.A.O., D.M.T., S.S.S., A.S.M., D.E.M. and E.V.B.; analysis and interpretation of data, E.V.P., R.S.M., E.S.S., M.V.S. and P.S.A.; writing—original draft preparation, R.S.M., E.V.P. and E.N.I.; writing—review and editing, P.S.A. and E.N.I.; visualization, E.V.P.; supervision, E.N.I. All authors have read and agreed to the published version of the manuscript.

Funding

This research has been supported by the Russian Science Foundation (grant number 23-15-20032). Funding sources did not influence how the study was conducted or the description of its results.

Institutional Review Board Statement

This study was conducted in accordance with the Declaration of Helsinki and approved by the local ethical committee of N.N. Petrov Institute of Oncology (Approval Code: 20. Approval Date: 23 November 2017).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Shia, J. The diversity of tumours with microsatellite instability: Molecular mechanisms and impact upon microsatellite instability testing and mismatch repair protein immunohistochemistry. Histopathology 2020, 78, 485–497. [Google Scholar] [CrossRef] [PubMed]
  2. Wang, J.; Li, R.; Li, J.; Yi, Y.; Liu, X.; Chen, J.; Zhang, H.; Lu, J.; Li, C.; Wu, H.; et al. Comprehensive analysis of oncogenic fusions in mismatch repair deficient colorectal carcinomas by sequential DNA and RNA next generation sequencing. J. Transl. Med. 2021, 19, 1–13. [Google Scholar] [CrossRef]
  3. Gallon, R.; Gawthorpe, P.; Phelps, R.L.; Hayes, C.; Borthwick, G.M.; Santibanez-Koref, M.; Jackson, M.S.; Burn, J. How Should We Test for Lynch Syndrome? A Review of Current Guidelines and Future Strategies. Cancers 2021, 13, 406. [Google Scholar] [CrossRef] [PubMed]
  4. Taieb, J.; Svrcek, M.; Cohen, R.; Basile, D.; Tougeron, D.; Phelip, J.M. Deficient mismatch repair/microsatellite unstable colorectal cancer: Diagnosis, prognosis and treatment. Eur. J. Cancer 2022, 175, 136–157. [Google Scholar] [CrossRef] [PubMed]
  5. Eso, Y.; Shimizu, T.; Takeda, H.; Takai, A.; Marusawa, H. Microsatellite instability and immune checkpoint inhibitors: Toward precision medicine against gastrointestinal and hepatobiliary cancers. J. Gastroenterol. 2019, 55, 15–26. [Google Scholar] [CrossRef]
  6. Jin, Z.; Sinicrope, F.A. Mismatch Repair-Deficient Colorectal Cancer: Building on Checkpoint Blockade. J. Clin. Oncol. 2022, 40, 2735–2750. [Google Scholar] [CrossRef] [PubMed]
  7. Imyanitov, E.; Kuligina, E. Molecular testing for colorectal cancer: Clinical applications. World J. Gastrointest. Oncol. 2021, 13, 1288–1301. [Google Scholar] [CrossRef]
  8. Vaňková, B.; Vaněček, T.; Ptáková, N.; Hájková, V.; Dušek, M.; Michal, M.; Švajdler, P.; Daum, O.; Daumová, M.; Mezencev, R.; et al. Targeted next generation sequencing of MLH1 -deficient, MLH1 promoter hypermethylated, and BRAF/RAS -wild-type colorectal adenocarcinomas is effective in detecting tumors with actionable oncogenic gene fusions. Genes Chromosom. Cancer 2020, 59, 562–568. [Google Scholar] [CrossRef] [PubMed]
  9. Bocciarelli, C.; Caumont, C.; Samaison, L.; Cariou, M.; Aline-Fardin, A.; Doucet, L.; Roudié, J.; Terris, B.; Merlio, J.P.; Marcorelles, P.; et al. MSI-High RAS-BRAF wild-type colorectal adenocarcinomas with MLH1 loss have a high frequency of targetable oncogenic gene fusions whose diagnoses are feasible using methods easy-to-implement in pathology laboratories. Hum. Pathol. 2021, 114, 99–109. [Google Scholar] [CrossRef]
  10. Singh, H.; Li, Y.Y.; Spurr, L.F.; Shinagare, A.B.; Abhyankar, R.; Reilly, E.; Brais, L.K.; Nag, A.; Ducar, M.D.; Thorner, A.R.; et al. Molecular Characterization and Therapeutic Targeting of Colorectal Cancers Harboring Receptor Tyrosine Kinase Fusions. Clin. Cancer Res. 2021, 27, 1695–1705. [Google Scholar] [CrossRef]
  11. Madison, R.W.; Hu, X.; Ramanan, V.; Xu, Z.; Huang, R.S.; Sokol, E.S.; Frampton, G.M.; Schrock, A.B.; Ali, S.M.; Ganesan, S.; et al. Clustered 8-Oxo-Guanine Mutations and Oncogenic Gene Fusions in Microsatellite-Unstable Colorectal Cancer. JCO Precis. Oncol. 2022, 6, e2100477. [Google Scholar] [CrossRef] [PubMed]
  12. Okano, S.; Yamashiro, Y.; Onagi, H.; Sasa, K.; Hayashi, T.; Takahashi, M.; Sugimoto, K.; Sakamoto, K.; Yao, T.; Saito, T. Tyrosine kinase alterations in colorectal cancer with emphasis on the distinct clinicopathological characteristics. Histopathology 2023. [Google Scholar] [CrossRef]
  13. Kurnit, K.C.; Westin, S.N.; Coleman, R.L. Microsatellite instability in endometrial cancer: New purpose for an old test. Cancer 2019, 125, 2154–2163. [Google Scholar] [CrossRef] [PubMed]
  14. Puliga, E.; Corso, S.; Pietrantonio, F.; Giordano, S. Microsatellite instability in Gastric Cancer: Between lights and shadows. Cancer Treat. Rev. 2021, 95, 102175. [Google Scholar] [CrossRef] [PubMed]
  15. Preobrazhenskaya, E.V.; Suleymanova, A.M.; Bizin, I.V.; Zagrebin, F.A.; Romanko, A.A.; Saitova, E.S.; Mulkidzhan, R.S.; Imyanitov, E.N. Spectrum of kinase gene rearrangements in a large series of paediatric inflammatory myofibroblastic tumours. Histopathology 2023, 83, 109–115. [Google Scholar] [CrossRef]
  16. Tiurin, V.I.; Preobrazhenskaya, E.V.; Mitiushkina, N.V.; Romanko, A.A.; Anuskina, A.A.; Mulkidjan, R.S.; Saitova, E.S.; Krivosheyeva, E.A.; Kharitonova, E.D.; Shevyakov, M.P.; et al. Rapid and Cost-Efficient Detection of RET Rearrangements in a Large Consecutive Series of Lung Carcinomas. Int. J. Mol. Sci. 2023, 24, 10530. [Google Scholar] [CrossRef] [PubMed]
  17. Izumi, H.; Matsumoto, S.; Liu, J.; Tanaka, K.; Mori, S.; Hayashi, K.; Kumagai, S.; Shibata, Y.; Hayashida, T.; Watanabe, K.; et al. The CLIP1–LTK fusion is an oncogenic driver in non-small-cell lung cancer. Nature 2021, 600, 319–323. [Google Scholar] [CrossRef]
  18. Imyanitov, E.; Zagrebin, F.; Mulkidzhan, R.; Krivosheeva, E.; Saitova, E.; Iyevleva, A.; Bizin, I.; Preobrazhenskaya, E. 25P Identification of novel kinase-activating fusions in non-small cell lung carcinomas (NSCLCs). Ann. Oncol. 2022, 33, S553–S554. [Google Scholar] [CrossRef]
  19. Martianov, A.S.; Mitiushkina, N.V.; Ershova, A.N.; Martynenko, D.E.; Bubnov, M.G.; Amankwah, P.; Yanus, G.A.; Aleksakhina, S.N.; Tiurin, V.I.; Venina, A.R.; et al. KRAS, NRAS, BRAF, HER2 and MSI Status in a Large Consecutive Series of Colorectal Carcinomas. Int. J. Mol. Sci. 2023, 24, 4868. [Google Scholar] [CrossRef]
  20. Afrăsânie, V.A.; Gafton, B.; Marinca, M.V.; Alexa-Stratulat, T.; Miron, L.; Rusu, C.; Ivanov, A.V.; Balan, G.G.; Croitoru, A.E. The Coexistence of RAS and BRAF Mutations in Metastatic Colorectal Cancer: A Case Report and Systematic Literature Review. J. Gastrointest. Liver Dis. 2020, 29, 251–256. [Google Scholar] [CrossRef]
  21. Uchida, S.; Kojima, T.; Sugino, T. Frequency and Clinicopathological Characteristics of Patients With KRAS/BRAF Double-Mutant Colorectal Cancer: An In Silico Study. Pathol. Oncol. Res. 2022, 28, 1610206. [Google Scholar] [CrossRef] [PubMed]
  22. Mustachio, L.M.; Roszik, J. Single-Cell Sequencing: Current Applications in Precision Onco-Genomics and Cancer Therapeutics. Cancers 2022, 14, 657. [Google Scholar] [CrossRef]
  23. O’haire, S.; Franchini, F.; Kang, Y.J.; Steinberg, J.; Canfell, K.; Desai, J.; Fox, S.; Ijzerman, M. Systematic review of NTRK 1/2/3 fusion prevalence pan-cancer and across solid tumours. Sci. Rep. 2023, 13, 1–10. [Google Scholar] [CrossRef] [PubMed]
  24. Marchetti, A.; Ferro, B.; Pasciuto, M.P.; Zampacorta, C.; Buttitta, F.; D’angelo, E. NTRK gene fusions in solid tumors: Agnostic relevance, prevalence and diagnostic strategies. Pathologica 2022, 114, 199–216. [Google Scholar] [CrossRef] [PubMed]
  25. Qi, C.; Zhou, T.; Bai, Y.; Chen, H.; Yuan, J.; Zhao, F.; Liu, C.; Ma, M.; Bei, T.; Chen, S.; et al. China special issue on gastrointestinal tumor—NTRK fusion in a large real-world population and clinical utility of circulating tumor DNA genotyping to guide TRK inhibitor treatment. Int. J. Cancer 2023. [Google Scholar] [CrossRef] [PubMed]
  26. Fois, S.S.; Paliogiannis, P.; Zinellu, A.; Fois, A.G.; Cossu, A.; Palmieri, G. Molecular Epidemiology of the Main Druggable Genetic Alterations in Non-Small Cell Lung Cancer. Int. J. Mol. Sci. 2021, 22, 612. [Google Scholar] [CrossRef]
  27. Wang, C.; Zhang, L.; Vakiani, E.; Shia, J. Detecting mismatch repair deficiency in solid neoplasms: Immunohistochemistry, microsatellite instability, or both? Mod. Pathol. 2022, 35, 1515–1528. [Google Scholar] [CrossRef] [PubMed]
  28. Marchiò, C.; Scaltriti, M.; Ladanyi, M.; Iafrate, A.; Bibeau, F.; Dietel, M.; Hechtman, J.; Troiani, T.; López-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]
  29. Nagtegaal, I.D.; Odze, R.D.; Klimstra, D.; Paradis, V.; Rugge, M.; Schirmacher, P.; Washington, K.M.; Carneiro, F.; Cree, I.A.; the WHO Classification of Tumours Editorial Board. The 2019 WHO classification of tumours of the digestive system. Histopathology 2020, 76, 182–188. [Google Scholar] [CrossRef] [PubMed]
  30. Zhou, X.P.; Hoang, J.M.; Li, Y.J.; Seruca, R.; Carneiro, F.; Sobrinho-Simoes, M.; Lothe, R.A.; Gleeson, C.M.; Russell, S.H.; Muzeau, F.; et al. Determination of the replication error phenotype in human tumors without the requirement for matching normal DNA by analysis of mononucleotide repeat microsatellites. Genes Chromosom. Cancer 1998, 21, 101–107. [Google Scholar] [CrossRef]
  31. Suraweera, N.; Duval, A.; Reperant, M.; Vaury, C.; Furlan, D.; Leroy, K.; Seruca, R.; Iacopetta, B.; Hamelin, R. Evaluation of tumor microsatellite instability using five quasimonomorphic mononucleotide repeats and pentaplex PCR. Gastroenterology 2002, 123, 1804–1811. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flowchart of the analysis of gene rearrangements in MSI-positive samples. RAS/RAF +: CRCs carrying mutations in KRAS, NRAS or BRAF oncogenes. RAS/RAF −: KRAS/NRAS/BRAF wild-type CRCs.
Figure 1. Flowchart of the analysis of gene rearrangements in MSI-positive samples. RAS/RAF +: CRCs carrying mutations in KRAS, NRAS or BRAF oncogenes. RAS/RAF −: KRAS/NRAS/BRAF wild-type CRCs.
Ijms 24 13610 g001
Figure 2. Flowchart of the detection of gene rearrangements in MSI-positive samples of colorectal cancers. RAS/RAFKRAS/NRAS/BRAF mutation status.
Figure 2. Flowchart of the detection of gene rearrangements in MSI-positive samples of colorectal cancers. RAS/RAFKRAS/NRAS/BRAF mutation status.
Ijms 24 13610 g002
Table 1. Clinical data for MSI-positive tumors with tyrosine kinase gene fusions.
Table 1. Clinical data for MSI-positive tumors with tyrosine kinase gene fusions.
IDSexAge, YearsDiagnosisRAS/RAF Mutations5′/3′-End Expression ImbalanceFusionMethod of Fusion Identification
1.A9365m74CRCnegativeALKSTRN::ALK (S5;ins53A20)NGS
2.C1182f71CRCnegativeALKEML4::ALK (E6;A20)PCR
3.C3234f49CRCnegativeALKSPTBN1::ALK (S7;A20)NGS
4.D0934m70CRCnegativeALKRBBP8::ALK (R8;A20)NGS
5.P28580f66CRCnegativeALKSPTBN1::ALK (S7;A20)NGS
6.P30811m80CRCnegativeALKEML4::ALK (E20;A20)PCR
7.B9775f62CRCnegative-CCDC6::RET (C1;RE12)PCR
8.C7622m58CRCnegative-CCDC6::RET (C1;RE12)PCR
9.A7310m65CRCnegative-CCDC6::RET (C1;RE12)PCR
10.P22395m62CRCnegative-CCDC6::RET (C1;RE12)PCR
11.B8013f63CRCnegativeRETNCOA4::RET (N8;R12)PCR
12.C2542f67CRCnegativeRETNCOA4::RET (N9del501;R12)NGS
13.C4286m54CRCnegativeRETNCOA4::RET (N9;R12)NGS
14.P16667f65CRCnegativeRETNCOA4::RET (N9;R12)NGS
15.P24158f66CRCnegativeRETCCDC6::RET (C1;RE12)PCR
16.A4695f64CRCnegativeNTRK1TPM3::NTRK1 (T8;N10)PCR
17.B2240f60CRCnegativeNTRK1TPM3::NTRK1 (T8;N10)PCR
18.B9472m52CRCnegativeNTRK1TPM3::NTRK1 (T8;N10)PCR
19.C1589f64CRCnegativeNTRK1TPM3::NTRK1 (T8;N10)PCR
20.C6335f69CRCnegativeNTRK1TPM3::NTRK1 (T8;N10)PCR
21.C6755m72CRCnegativeNTRK1TPM3::NTRK1 (T8;N10)PCR
22.D0273m68CRCnegativeNTRK1TPM3::NTRK1 (T8;N10)PCR
23.D1407f58CRCnegativeNTRK1TPM3::NTRK1 (T8;N10)PCR
24.E2169m58CRCnegativeNTRK1TPM3::NTRK1 (T8;N10)PCR
25.P18891f59CRCnegativeNTRK1TPM3::NTRK1 (T8;N12)PCR
26.P21693m86CRCnegativeNTRK1TPM3::NTRK1 (T8;N10)PCR
27.E4114f69CRCnegativeNTRK1LMNA::NTRK1 (L3;N10)PCR
28.B9540m72CRCnegativeNTRK1TPR::NTRK1 (T15;N10)NGS
29.C4763m65CRCnegativeNTRK1TPR::NTRK1 (T20;N11)NGS
30.C7112f65CRCnegativeNTRK1ZKSCAN1::NTRK1 (Z3;N12)NGS
31.D0155f81CRCnegativeNTRK1LMNA::NTRK1 (L9ins44;del74N11)NGS
32.B9665m74CRCnegativeNTRK2ETV6::NTRK2 (E5;N15)NGS
33.A4890f65CRCnegativeNTRK3EML4::NTRK3 (E2;N14)PCR
34.B1263f58CRCnegative-EML4::NTRK3 (E2;N14)PCR
35.B4631f68CRCnegativeNTRK3EML4::NTRK3 (E2;N14)PCR
36.C3968f70CRCnegativeNTRK3EML4::NTRK3 (E2;N14)PCR
37.C5878m57CRCnegativeNTRK3ETV6::NTRK3 (E5;N15)PCR
38.C6430f72CRCnegativeNTRK3EML4::NTRK3 (E2;N14)PCR
39.C7912f54CRCnegativeNTRK3EML4::NTRK3 (E2;N14)PCR
40.D7461f61CRCnegativeNTRK3ETV6::NTRK3 (E5;N15)PCR
41.P22904f68CRCnegativeNTRK3EML4::NTRK3 (E2;N14)PCR
42.P23972f67CRCnegativeNTRK3ETV6::NTRK3 (E5;N15)PCR
43.B4561m31CRCKRAS p.A146TNTRK2ETV6::NTRK2 (E5;N15)NGS
44.C1274f73CRCBRAF p.V600E-ETV6::NTRK3 (E5;N15)PCR
45.C3231f76CRCBRAF p.V600ENTRK1LMNA::NTRK1 (L3;N10)PCR
46.P29082f85CRCNRAS p.Q61KNTRK1TPM3::NTRK1 (T8;N10)PCR
47.C7527f71GCKRAS p.G12CNTRK1TPM3::NTRK1 (T8;N10)PCR
48.C4292f72CRCNegativeALKnot definedn/a for NGS
49.C8576m66CRCNegativeALKnot definedn/a for NGS
50.C2716m80CRCNegativeRETnot definedn/a for NGS
51.C5821m83CRCNegativeRETnot definedn/a for NGS
52.P15387m68CRCNegativeRETnot definedn/a for NGS
53.B9814f65CRCNegativeNTRK1not definedn/a for NGS
54.C4831f60CRCNegativeNTRK1not definedn/a for NGS
55.D0795m74CRCNegativeNTRK1not definedn/a for NGS
56.C5665f68CRCNegativeNTRK3not definedn/a for NGS
57.C9396m32CRCNegativeNTRK3not definedn/a for NGS
58.D3390f80CRCNegativeNTRK3not definedn/a for NGS
59.D5144m66CRCKRAS p.G12DNTRK3not definedn/a for NGS
60.A9663m66GCNegativeNTRK1not definedn/a for NGS
61.C1642m83GCNegativeNTRK1not definedn/a for NGS
62.E3046m34GCNegativeNTRK3not definedn/a for NGS
63.C8708f67ECNegativeNTRK3not definedn/a for NGS
64.D5486f71ECNegativeNTRK3not definedn/a for NGS
65.D6096f64ECNegativeNTRK3not definedn/a for NGS
CRC—colorectal cancer, EC—endometrial cancer, GC—gastric cancer, f—female, m—male, n/a for NGS—material not available for next generation sequencing.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mulkidjan, R.S.; Saitova, E.S.; Preobrazhenskaya, E.V.; Asadulaeva, K.A.; Bubnov, M.G.; Otradnova, E.A.; Terina, D.M.; Shulga, S.S.; Martynenko, D.E.; Semina, M.V.; et al. ALK, ROS1, RET and NTRK1–3 Gene Fusions in Colorectal and Non-Colorectal Microsatellite-Unstable Cancers. Int. J. Mol. Sci. 2023, 24, 13610. https://doi.org/10.3390/ijms241713610

AMA Style

Mulkidjan RS, Saitova ES, Preobrazhenskaya EV, Asadulaeva KA, Bubnov MG, Otradnova EA, Terina DM, Shulga SS, Martynenko DE, Semina MV, et al. ALK, ROS1, RET and NTRK1–3 Gene Fusions in Colorectal and Non-Colorectal Microsatellite-Unstable Cancers. International Journal of Molecular Sciences. 2023; 24(17):13610. https://doi.org/10.3390/ijms241713610

Chicago/Turabian Style

Mulkidjan, Rimma S., Evgeniya S. Saitova, Elena V. Preobrazhenskaya, Karimat A. Asadulaeva, Mikhail G. Bubnov, Ekaterina A. Otradnova, Darya M. Terina, Sofia S. Shulga, Darya E. Martynenko, Maria V. Semina, and et al. 2023. "ALK, ROS1, RET and NTRK1–3 Gene Fusions in Colorectal and Non-Colorectal Microsatellite-Unstable Cancers" International Journal of Molecular Sciences 24, no. 17: 13610. https://doi.org/10.3390/ijms241713610

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