ArticlePublisher preview available

Detecting anaplastic lymphoma kinase (ALK) gene rearrangements with next-generation sequencing remains a reliable approach in patients with non-small-cell lung cancer

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Next-generation sequencing (NGS) is rapidly becoming routine in clinical oncology practice to identify therapeutic biomarkers, including gene rearrangements in anaplastic lymphoma kinase (ALK). Our study investigated the concordance of ALK positivity evaluated by DNA-based NGS with orthogonal ALK testing methods such as fluorescence in situ hybridization (FISH), immunohistochemistry (IHC), and RNA-based NGS (RNA-NGS). Thirty-eight patients with lung adenocarcinoma who were detected with ALK rearrangements using DNA-NGS and also had adequate tissue samples submitted for FISH, IHC, and RNA-NGS, were included in this study. Of the 38 patients, RNA samples from 3 patients failed quality control for RNA-NGS. The concordance of ALK positivity was calculated relative to DNA-NGS results. The concordance rates were 97.1% (34/35) for RNA-NGS, 94.7% (36/38) for IHC, and 97.4% (37/38) for FISH. DNA-NGS detected single ALK rearrangements in 14 (35.0%) patients and complex ALK rearrangements in 26 (65.0%). RNA-NGS detected only single transcripts of the primary ALK fusions. A novel LANCL1-ALK (L7:A20) detected using DNA-NGS was detected as EML4-ALK (E13:A20) transcripts using RNA-NGS. Interestingly, patients with single ALK rearrangements were more likely to be detected with atypical isolated red signals (p < 0.001), while patients with complex ALK rearrangements were more likely to be detected with atypical split red and green signals less than 2 signal diameters apart (p < 0.001). Our study highlights the reliability of NGS in the accurate detection of specific ALK fusion variants and concomitant mutations that are crucial for individualized treatment decisions in patients with lung cancer.
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
1 3
https://doi.org/10.1007/s00428-022-03339-y
ORIGINAL ARTICLE
Detecting anaplastic lymphoma kinase (ALK) gene rearrangements
withnext‑generation sequencing remains areliable approach
inpatients withnon‑small‑cell lung cancer
YingDing1· ChangSun2· WeiSu2· ChenMiao1· XiaoHe1· Jin‑SongWang3· Zhi‑HongZhang1
Received: 5 January 2022 / Revised: 12 April 2022 / Accepted: 12 May 2022
© The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature 2022
Abstract
Next-generation sequencing (NGS) is rapidly becoming routine in clinical oncology practice to identify therapeutic biomark-
ers, including gene rearrangements in anaplastic lymphoma kinase (ALK). Our study investigated the concordance of ALK
positivity evaluated by DNA-based NGS with orthogonal ALK testing methods such as fluorescence insitu hybridization
(FISH), immunohistochemistry (IHC), and RNA-based NGS (RNA-NGS). Thirty-eight patients with lung adenocarcinoma
who were detected with ALK rearrangements using DNA-NGS and also had adequate tissue samples submitted for FISH,
IHC, and RNA-NGS, were included in this study. Of the 38 patients, RNA samples from 3 patients failed quality control
for RNA-NGS. The concordance of ALK positivity was calculated relative to DNA-NGS results. The concordance rates
were 97.1% (34/35) for RNA-NGS, 94.7% (36/38) for IHC, and 97.4% (37/38) for FISH. DNA-NGS detected single ALK
rearrangements in 14 (35.0%) patients and complex ALK rearrangements in 26 (65.0%). RNA-NGS detected only single
transcripts of the primary ALK fusions. A novel LANCL1-ALK (L7:A20) detected using DNA-NGS was detected as EML4-
ALK (E13:A20) transcripts using RNA-NGS. Interestingly, patients with single ALK rearrangements were more likely to be
detected with atypical isolated red signals (p < 0.001), while patients with complex ALK rearrangements were more likely
to be detected with atypical split red and green signals less than 2 signal diameters apart (p < 0.001). Our study highlights
the reliability of NGS in the accurate detection of specific ALK fusion variants and concomitant mutations that are crucial
for individualized treatment decisions in patients with lung cancer.
Keywords ALK rearrangement· FISH· NGS· NSCLC
Abbreviations
ALK
Anaplastic lymphoma kinase
EML4
Echinoderm microtubule-associated protein-
like 4
FDA
Food and Drug Administration
FFPE Formalin-fixed, paraffin-embedded
FISH
Fluorescence insitu hybridization
IHC Immunohistochemistry
NGS Next-generation sequencing
NSCLC
Non-small-cell lung cancer
qRT-PCR
Quantitative reverse transcriptase-polymerase
chain reaction
TKI
Tyrosine kinase inhibitor
Introduction
Between 2 and 7% of unselected non-small-cell lung cancer
(NSCLC) harbor chromosomal translocations involving the
anaplastic lymphoma kinase (ALK) gene [15]. A majority
of ALK-rearranged NSCLCs harbor gene fusion between the
5 portion of the echinoderm microtubule-associated protein-
like 4 (EML4) gene and the 3 portion of the ALK gene that
contains its tyrosine kinase domain [1]. EML4-ALK fusion
results from the paracentric inversion in the short arm of
chromosome 2 (between 2p21 and 2p23) and leads to the
constitutive activation of downstream signaling pathways
that promote cell proliferation [1]. At least 15 variants of
* Zhi-Hong Zhang
zhangzh@njmu.edu.cn
1 Department ofPathology, the First Affiliated Hospital
ofNanjing Medical University, Nanjing210029, China
2 The First School ofClinical Medicine, Nanjing Medical
University, Nanjing210029, China
3 Department ofPathology, Nanjing First Hospital, Nanjing
Medical University, Nanjing210000, China
/ Published online: 28 May 2022
Virchows Archiv (2022) 481:405–419
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Article
Objectives Gene rearrangements frequently act as oncogenic driver mutations and determine the onset and progression of cancer. RNA-based next-generation sequencing (NGS) is being used with increasing frequency for solid tumors. The purpose of our study is to investigate the feasibility and utility of an RNA-based NGS fusion panel for solid tumors. Methods We conducted a retrospective, single-institution review of fusion panels requested between May 2022 and March 2023. Demographic, clinical, pathologic, and molecular findings of the patients were reviewed. The utility of the RNA-based NGS fusion panel for the pathologic diagnosis of solid tumors was assessed. Results Our study included 345 cases, and a fusion event was identified in 24.3% (78/321) of cases. Among the 110 cases submitted for diagnostic purposes, a fusion event was detected in 42.7% (47/110) of cases. The results led to refinement or clarification of the initial diagnosis in 31.9% (15/47) of cases and agreement or support for the initial diagnosis in 59.6% (28/47) of cases. Furthermore, our study indicated that the overall cellularity (tumor and normal tissue) of the tested specimen influences the success of the testing process. Conclusions In summary, this study demonstrated the feasibility and utility of an RNA-based NGS fusion panel for a wide variety of solid tumors in the appropriate clinicopathologic context. These findings warrant further validation in larger studies involving multiple institutional patient cohorts.
Article
Full-text available
Background: Targeted treatment modalities for non-small cell lung carcinoma (NSCLC) patients are expanding rapidly and demand a constant adaptation of molecular testing strategies. In this regard, broad reflex testing via next-generation sequencing (NGS) might have several advantages. However, real-world data regarding practical feasibility and clinical relevance are scarce, especially for RNA-based NGS. Methods: We performed a retrospective study comparing NGS use in two consecutive years (2019 and 2020). In 2019, reflex testing mainly consisted of DNA-based NGS for mutations and immunohistochemistry (IHC) for ALK, ROS1, and NTRK fusion products. At the beginning of 2020, our approach has changed, with DNA- and RNA-based NGS panels now being simultaneously performed. This change in protocol allowed us to retrospectively evaluate if broad molecular reflex testing brings additional value to lung cancer patients. Results: Within the whole cohort (n=432), both DNA- and RNA-based NGS yielded almost always evaluable results. Only in 6 cases, the RNA content was too little for an appropriate analysis. After integrating RNA-based NGS in the reflex testing approach, the number of detected fusions increased significantly (2.6% vs. 8.2%; P=0.0021), but also more patients received targeted therapies. Furthermore, exceedingly rare alterations were more likely to be detected, including the so far undescribed EGFR-NUP160 fusion. Conclusions: Our study demonstrates that a comprehensive approach to reflex NGS testing is practically feasible and clinically relevant. Including RNA-based panels in the reflex testing approach results in more detected fusions and more patients receiving targeted therapies. Additionally, this broad molecular profiling strategy identifies patients with emerging biomarkers, underscoring its usefulness in the rapidly evolving landscape of targeted therapies.
Article
Full-text available
Background Receptor tyrosine kinases (RTKs) are a class of tyrosine kinases that regulate cell-to-cell communication and control a variety of complex biological functions. Dysregulation of RTK signaling partly due to chromosomal rearrangements leads to novel tyrosine kinase fusion oncoproteins that are possibly driver alterations to cancers. Targeting some RTK fusions with specific tyrosine kinases inhibitors (TKIs) is an effective therapeutic strategy across a spectrum of RTK fusion-related cancers. However, there is still a paucity of extensive RTK fusion investigations in breast cancer. This study aims to characterize RTK fusions in Chinese breast cancer patients. Methods An in-house DNA sequencing database of 1440 Chinese breast cancer patients with a capture-based panel (520 gene or 108 gene-panel) was thoroughly reviewed. A total of 2,229 samples including 1,045 tissues and 1,184 plasmas were analyzed. RTK fusion was defined as an in-frame fusion with the tyrosine kinase domain of the RTK completely retained. Concomitant mutations were also analyzed and tumor mutational burden (TMB) was calculated. Patients’ clinical characteristics were retrieved from case records. Results A total of 30 RTK fusion events were identified from 27 breast cancer patients with a prevalence of 1.875%%. FGFR2 fusions were seen the most commonly (n=7), followed by RET (n=5), ROS1 (n=3), NTRK3 (n=3), BRAF (n=2), and NTRK1 (n=2). Other RTK fusions including ALK, EGFR, FGFR1, FGFR3, MET, and NTRK2 were identified in one patient each. A total of 27 unique resultant fusion proteins (22 with a novel partner) were discovered including 19 intrachromosomal rearrangements and 8 interchromosomal ones. Twenty-one fusions had the tyrosine kinase domain in-frame fused with a partner gene and six were juxtaposed with an intergenic space. Among the 27 fusions, FGFR2-WDR11 (E17: intergenic) (n=3) and ETV6-NTRK3 (E5:E15) (n=2) occurred recurrently. Of note, the normalized abundance of RTK fusion (fusion AF/max AF) correlated negatively with TMB (r=-0.48, P=0.017). Patients with TMB < 8 (Mutations/Mb) displayed a higher fusion abundance than those with TMB ≥ 8 (Mutations/Mb) (P=0.025). Moreover, CREBBP mutation only co-occurred with FGFR2 fusion (P=0.012), while NTRK3 fusion and TP53 mutation were mutually exclusive (P=0.019). Conclusion This is the first study comprehensively delineating the prevalence and spectrum of RTK fusions in Chinese breast cancers. Further study is ongoing to identify the enriched subpopulation who may benefit from RTK fusion inhibitors.
Article
Full-text available
The presence of anaplastic lymphoma kinase (ALK) rearrangement defines a molecular subtype of non-small-cell lung cancer (NSCLC). ALK inhibitors confer significant clinical benefits in patients with ALK-positive advanced NSCLC; therefore, it is of great clinical significance to select accurate, rapid, and appropriate ALK testing methods to screen for patients who are suitable for anti-ALK treatment. In recent years, great progress has been made in the development and clinical application of ALK inhibitors, as well as in our understanding of acquired drug resistance mechanisms. Meanwhile, new ALK companion diagnostic platforms have been developed and applied in clinical practice. Although many studies have shown that there is a high rate of concordance among these platforms, new problems continue to appear during testing. To maximize the benefit for patients, accurate testing results can be obtained by first selecting the appropriate testing method and then formulating, optimizing, and complying with the standardized testing process in accordance with the testing population and specimen types. With the ongoing accumulation of clinical practice data, experience from quality control of ALK testing, and results from multicenter research, an updated expert consensus is necessary. The experts who participated in the discussion and development of this consensus have a rich background in theoretical and clinical testing experience, which ensures the practical value of the information presented in this recommendation guideline.
Article
Full-text available
Since the discovery of echinoderm microtubule-associated protein-like 4 (EML4) and anaplastic lymphoma kinase (ALK) gene fusion in non-small cell lung carcinoma (NSCLC) in 2007, more than 10 EML4-ALK variants based on the exon breakpoints in EML4 have been identified. Unlike other receptor tyrosine kinase fusion positive NSCLC such as ROS1 or RET fusion, EML4-ALK is the dominant fusion variant in ALK+ NSCLC accounting for approximately 85% of all fusion variants in ALK+ NSCLC. Currently, eight EML4-ALK variants are generally recognized with a number (1, 2, 3a/b, 4’, 5a/b, 5’, 7, 8) with EML4-ALK variants 1 and 3 being the two most common variants accounting for 75-80% of the total EML4-ALK variants. Preclinical, retrospective analyses of institutional databases, and global randomized phase 3 trials have demonstrated differential clinical response (overall response rate, progression-free survival) to ALK tyrosine kinase inhibitors (TKIs) between the “short” (v3 and v5) and “long” (v1, v2, v5’, v7, and v8) EML4-ALK variants. We discuss in more details how EML4-ALK variant structure influences protein stability and response to ALK TKIs, Additionally, the most recalcitrant single solvent-front mutation ALK G1202R is more prone to develop among EML4-ALK v3 following sequential use of next-generation ALK TKIs. Furthermore, TP53 mutations being the most common genomic co-alterations in ALK+ NSCLC also contribute to the heterogeneous response to ALK TKIs. Recognizing ALK+ NSCLC is not one homogeneous disease entity but comprised of different ALK fusion variants with different underlying genomic alterations in particular TP53 mutations that modulate treatment response will provide insight into the further optimization of treatment of ALK+ NSCLC patients potentially leading to improvement in survival.
Article
Full-text available
Introduction Reliable diagnostic approaches to detect ALK rearrangement are critical for selecting patients eligible for crizotinib therapy. This study aimed to compare next-generation sequencing (NGS) and Ventana immunohistochemistry (IHC) in evaluating ALK rearrangements and evaluate their impact on first-line crizotinib efficacy. Patients and Methods A total of 472 NSCLC patients were identified as ALK-positive by NGS and/or IHC between March 2014 and February 2020. The concordance of ALK detection, overall response rate (ORR), and progression-free survival (PFS) were analyzed for 319 patients who received front-line crizotinib. Results First-line crizotinib (n=319) significantly prolonged PFS in comparison with chemotherapy (n=46; 12.0 vs 6.8 months; p<0.0001). Of the 76 crizotinib-treated patients whose ALK status was assessed by both NGS and IHC, 78.9% of the patients had concordant ALK status (NGS-positive/IHC-positive), 18.4% patients were NGS-positive but IHC-negative, and 2 patients were IHC-positive but NGS-negative. Different detection assays confer no statistical difference in ORR and PFS with first-line crizotinib. The ORR in NGS only, IHC only, and both NGS and IHC was 84.3%, 90.1%, and 88.1%, respectively, while PFS was 11.4, 13.0, and 11.0 months, respectively. The ORR in NGS-positive/IHC-positive and NGS-positive/IHC-negative patients was 85.4% and 92.8%, respectively. Compared to NGS-positive/IHC-positive patients, those with NGS-positive/IHC-negative results had a trend of shorter PFS but statistical significance was not reached (mPFS, 5.9 months vs 11.5 months, p=0.43). Conclusion Our results demonstrate that ALK status detected by NGS and/or IHC is reliable in identifying patients with ALK-positive NSCLC who will benefit from ALK inhibitor therapy.
Article
Full-text available
Background During non-reciprocal/reciprocal translocation process, 5’-ALK sometimes get retained in the genome and are detectable by next-generation sequencing (NGS); however, no report have investigated its clinical significance. Our study aimed to assess the impact of harboring 5’-ALK on the efficacy of crizotinib. Patients and methods A total of 150 patients with NGS-identified ALK-rearranged NSCLC from March 2014 to July 2018 at the Hunan Cancer Hospital were enrolled in this study. The efficacy of crizotinib as first-line therapy was evaluated in 112 patients according to the retention of 5’-ALK. Results Among 150 NSCLC patients, non-reciprocal/reciprocal translocation was detected in 18.7% (28/150) and 3’-ALK fusion alone was detected in 81.3% (122/150). Among the 112 patients who received first-line crizotinib, 89 patients had 3’-ALK fusion alone (79 EML4-ALK and 10 non-EML4 ALK) and 23 patients had non-reciprocal/reciprocal ALK translocation. Among the patients with non-reciprocal/reciprocal ALK translocation, 3 patients harbored dual concurrent 3’-ALK fusions. Patients with non-reciprocal/reciprocal ALK translocation had higher incidence of brain metastasis at baseline than those with 3’-ALK fusion alone (39.1% vs. 13.4%, p=0.028). Crizotinib-treated patients with non-reciprocal/reciprocal ALK translocation had significantly shorter mPFS compared with patients carrying 3’-ALK fusion alone (6.1m vs. 12.0m, p=0.001) or with EML4-ALK fusion alone (6.1m vs. 12.6m, p=0.001). Multivariate analysis revealed that harboring non-reciprocal/reciprocal ALK translocation was an independent predictor of worse PFS for crizotinib-treated ALK-rearranged NSCLC (p=0.0046). Conclusions Presence of non-reciprocal/reciprocal ALK translocation was predictive for worse PFS and greater likelihood of baseline brain metastases in patients with ALK-rearranged NSCLC who received first-line crizotinib.
Article
RET fusion has emerged as a targetable driver in non-small-cell lung cancer (NSCLC). A comparative analysis on RET fusions at DNA (DNA-seq) and RNA (RNA-seq) levels was performed in this study. Archived tumor samples from 54 NSCLC patients were selected for RNA-seq, including patients who concurrently harbored a canonical RET fusion (KIF5B-RET or CCDC6-RET) and non-canonical RET fusion(s) and patients who harbored non-canonical RET fusion(s) at DNA level. RNA-seq identified RET fusion transcripts in 41/44 samples passing quality control. In the subset of cases harboring RET 3’-end fusions and predicted to produce in-frame proteins (Group A, n=33), RNA-seq identified the same 3’-end fusions in 32 (96.9%). 26/32 also had a reciprocal RET 5’-end fusion detected by DNA-seq that were not transcribed. In the subset with DNA-level out-of-frame RET fusions (group B, n=9), RNA-seq identified in-frame RET fusion transcripts in 8 cases (88.9%). In the subset only identified with a RET 5’-end fusion by DNA-seq (Group C, n=2), RNA-seq detected the corresponding 3’-end fusion in one case. The discordant DNA- and RNA-level fusions observed in Group B may be mediated by complex genomic rearrangement events and transcriptional or post-transcriptional processes. In conclusion, DNA-seq demonstrates a high concordance of 96.9% on detecting in-frame RET fusion, but shows a low concordance on detecting out-of-frame RET fusion and RET 5’-end fusion compared with RNA-seq.
Article
Next-generation sequencing (NGS) allows sequencing a high number of nucleotides in a short time frame at an affordable cost. While this technology has been widely implemented, there are no recommendations from scientific societies about their use in oncology practice. The European Society for Medical Oncology (ESMO) is proposing three levels of recommendations for the use of NGS. Based on the current evidence, ESMO recommends routine use of NGS on tumour samples in advanced non-squamous NSCLC, prostate cancers, ovarian cancers and cholangiocarcinoma. In these tumours, large multigene panels could be used if they add acceptable extra cost compared to small panels. In colon cancers, NGS could be an alternative to polymerase chain reaction (PCR). In addition, based on KN158 trial and considering that patients with endometrial and small cell lung cancers should have broad access to anti-PD1 antibodies, it is recommended to test tumour mutational burden (TMB) in cervical cancers, well- and moderately-differentiated neuroendocrine tumours, salivary cancers, thyroid and vulvar cancers, as TMB-high predicted response to pembrolizumab in these cancers. Outside the indications of multigene panels, and considering that the use of large panel of genes could lead to few clinically meaningful responders, ESMO acknowledges that a patient and a doctor could decide together to order a large panel of genes, pending no extra cost for the public healthcare system and if the patients is informed about the low likelihood of benefit. ESMO recommends that the use of off-label drugs matched to genomics is done only if an access programme and a procedure of decision has been developed at the national or regional level. Finally, ESMO recommends that clinical research centres develop multigene sequencing as a tool to screen patients eligible for clinical trials and to accelerate drug development, and prospectively capture the data that could further inform how to optimise the use of this technology.
Article
Introduction Next-generation sequencing (NGS) based on genomic DNA has been widely applied for gene rearrangement detection in patients with non-small cell lung cancer (NSCLC). However, intergenic-breakpoint fusions, in which one or both genomic breakpoints localize to intergenic regions, confound kinase fusion detection. We evaluated the function of intergenic-breakpoint fusions with multiplex molecular testing approaches. Methods NSCLCs with intergenic-breakpoint fusion identified by DNA-based NGS were analyzed by RNA-based NGS, immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH). Results Twenty-six cases with single intergenic-breakpoint fusion were identified from a large cohort of NSCLCs using DNA-based NGS. Of the 26 cases, RNA-based NGS detected expressed fusion transcripts in 11 cases, and the genomic breakpoint position did not logically predict breakpoint of the fusion transcript in these cases, possibly due to complex rearrangements (n=5), alternative splicing (n=2) and reciprocal rearrangement (n=4). Nonetheless, no expressed fusion transcript was detected in 5 cases. Moreover, positive ALK IHC was observed in 3 of the remaining 10 cases without RNA-based NGS results. Three intergenic-breakpoint ALK fusion cases with or without RNA-based NGS/IHC confirmation receiving crizotinib treatment showed partial responses. However, one intergenic-breakpoint ROS1 case, given the positive FISH result, received crizotinib but developed progressive disease within one month, possibly owing to no functional fusion transcript detected by RNA-based NGS. Conclusions Intergenic-breakpoint fusions detected by DNA sequencing confound kinase fusion detection in NSCLC, as functional fusion transcripts may be generated or not. Additional validation testing using RNA/protein assay should be performed in intergenic-breakpoint fusion cases to guide optimal treatment.