Skip to main content
British Journal of Cancer logoLink to British Journal of Cancer
. 2021 Oct 12;125(11):1544–1551. doi: 10.1038/s41416-021-01568-7

Clinical assessment of the miR-34, miR-200, ZEB1 and SNAIL EMT regulation hub underlines the differential prognostic value of EMT miRs to drive mesenchymal transition and prognosis in resected NSCLC

Simon Garinet 1,2, Audrey Didelot 2, Thomas Denize 1, Alexandre Perrier 2, Guillaume Beinse 2, Jean-Baptiste Leclere 3, Jean-Baptiste Oudart 1, Laure Gibault 4, Cecile Badoual 4, Françoise Le Pimpec-Barthes 3, Pierre Laurent-Puig 1,2, Antoine Legras 2,3,#, Helene Blons 1,2,✉,#
PMCID: PMC8609001  PMID: 34642464

Abstract

Background

Patients with non-small cell lung cancer (NSCLC) receiving curative surgery have a risk of relapse, and adjuvant treatments only translate into a 5% increase in 5-year survival. We assessed the clinical significance of epithelial–mesenchymal transition (EMT) and explored its association with the [SNAIL/miR-34]:[ZEB/miR-200] regulation hub to refine prognostic information.

Methods

We validated a 7-gene EMT score using a consecutive series of 176 resected NSCLC. We quantified EMT transcription factors, microRNAs (miRs) of the miR-200, miR-34 families and miR-200 promoter hypermethylation to identify outcome predictors.

Results

Most tumours presented with an EMT-hybrid state and the EMT score was not predictive of outcome. Individually, all miR-200 were inversely associated with the EMT score, but only chromosome-1 miRs, miR-200a, b, 429, were associated with disease-free survival (p = 0.08, 0.05 and 0.025) and overall survival (p = 0.013, 0.003 and 0.006). We validated these associations on The Cancer Genome Atlas data. Tumour unsupervised clustering based on miR expression identified two good prognostic groups, unrelated to the EMT score, suggesting that miR profiling may have an important clinical value.

Conclusion

miR-200 family members do not have similar predictive value. Core EMT-miR, regulators and not EMT itself, identify NSCLC patients with a low risk of relapse after surgery.

Subject terms: Prognostic markers, Non-small-cell lung cancer

Introduction

Despite major advances, non-small cell lung cancer (NSCLC) remains the major cause of cancer-related death in developed countries [1]. Metastasis and drug resistance are the main factors contributing to relapse and death [2]. Patients with stage I to IIIA are at risk of relapse and death after surgical resection, with a 40–70% risk of recurrence depending on the stage. Adjuvant treatments have been shown to reduce cancer recurrence risk; however, patients with low relapse risk have a moderate benefit from adjuvant therapy. With various upcoming practice changes in adjuvant options as the use of immunotherapies or targeted therapies [3], there is a need for identifying markers to guide treatment selection. The identification and validation of molecular factors linked to relapse risk that predicts which patients are most likely to benefit from adjuvant treatment remain a challenge. Recurrence after complete resection of NSCLC has been attributed to micrometastatic cancer cells and disseminated tumour cells that may have acquired a new phenotype with increased expression of mesenchymal markers and decreased expression of epithelial markers, referred to as epithelial-to-mesenchymal transition (EMT). Although the exact role of EMT in tumour metastasis and cancer prognosis remains a matter of debate, there is a consensus on considering tumour plasticity as the major key point as it allows cells to switch back and forth from E to M states. EMT is a complex molecular and cellular process of tissue remodelling that plays essential roles in cell invasion, migration and drug resistance in many cancer types including NSCLC [4]. During EMT, cells acquire increased mobility, invasiveness, stem-cell properties and resistance to apoptosis [5]. Many signalling pathways control EMT according to different cellular contexts [6, 7]. Pathways involved in EMT are transforming growth factor-β [8], epidermal growth factor (EGF) [9], which activate transcription factors (TFs) [10], including SNAIL superfamily members, ZEB family members, E47, the Krüpple-like factor 8 [11, 12], TWIST family members, Goosecoid, E2.2/TCF4 and FOXC2. Up-regulation of these TFs and loss of E-cadherin are hallmarks of EMT and are related to carcinogenesis and metastasis [13].

MicroRNAs (miRNAs or miRs) are important regulators of EMT-driver genes and are involved in cancer progression and metastasis [5, 14, 15]. MiR-34 and miR-200 are the two main miR families involved in the regulation of cell fate and plasticity through EMT. The regulatory unit of miRs and EMT-TFs [SNAIL/miR-34]:[ZEB/miR-200] is the core regulatory system for the EMT process where miRs regulate EMT-TF, and, in turn, EMT-TF regulates miRs through complex regulation loops. Besides their involvement in driving EMT, miR-200 was described to regulate angiogenesis [16], to restrict metastasis and to modulate intratumoral immunosuppression [17]. In some studies, a prometastatic and protumoral effect of miR-200 was also reported, emphasising the complexity of EMT regulation [16, 18]. MiR-200 family splits into two clusters located on chromosome 1 (chr-1) (miR-200a, b and 429) and chr-12 (miR-200c and 141). Those two clusters are differently expressed and their effects on EMT may depend among tumour types. In an insulinoma mouse model, it was shown that the miR-200c family—and not the closely related miR-141—was responsible for EMT regulation through ZEB1 activation [19]. In breast cancer, miR-200c silencing and ZEB1 regulation were associated with aggressiveness [20]. In colorectal cancer, low miR-200a expression was related to poor prognosis [21]. In the lung, nearly all miR-200s were repressed in metastases as compared to primary tumours in a mouse metastatic lung adenocarcinoma, suggesting a global miR-200 involvement in lung cancer progression [22]. However, only miR-141 and miR-200c were markers of overall survival (OS) in early-stage NSCLC [23], suggesting that the prognostic value of miR-200 family members remains a matter of debate.

To assess their proper relevance to survival, we quantified all members of the miR-200/ZEB/miR-34/SNAIL circuit and downstream EMT targets in a series of 176 resected NSCLC. We validated and used a 7-genes score to classify samples. We correlated miRs and TFs with the EMT score and assessed their value in predicting OS and relapse-free survival (RFS) individually or using hierarchical clustering. Finally, this study underlines highly significant differences between chr-1 and chr-12 miR-200 in predicting relapse and death in NSCLC.

Materials and methods

Study design and patients

This study, conducted at the European Georges Pompidou hospital, was approved by CPP Ile de France 2 ethics committee (nos. 2012-08-09 and 2012-08-09 A1) and registered in clinical trial.gov (NCT03509779). Patients with NSCLC treated by surgery for curative intent signed informed consent for research and tumour tissues banking. A series of 176 primary lung cancer were prospectively collected from October 2011 to December 2014. Samples were stored frozen (−80 °C) at the Biological Resources centre and Tumour Bank Platform (PRB-HEGP BB-0033-00063) before nucleic acid extraction. Baseline demographics and clinical variables were collected using the Epithor national database, and survival data were updated using patients’ medical records.

Molecular characterisation

Tumours were cut prepared on a cryostat and reviewed by the pathologist before DNA and RNA extractions. Mean tumour cell content was 52% ± 25; all samples with <20% were excluded. DNA and RNA were extracted using QIAamp DNA Mini Kit (Qiagen) and miRNeasy Mini Kit (Qiagen) extraction kits; DNAs and RNAs were quantified by Qubit Fluorometric Quantitation (Thermo Fisher Scientific) and stored frozen.

Samples were characterised for molecular alterations by targeted next-generation sequencing (NGS) (Ion AmpliSeq™ Colon-Lung Cancer Research Panel v2, Life Technologies™, Carlsbad, CA). Briefly, the multiplex barcoded libraries are generated from 10 ng of DNA following the manufacturer’s recommendations (Ion Ampliseq Library Kit V2) and are normalised using the Ion Library Equalizer™ Kit. The pooled libraries (max 96) are processed on Ion Chef™ System for template preparation and chip loading (Ion PI HI-Q Chef Kit, Ion PI Chip V3), and sequenced on the Ion Proton™ System (Life Technologies™). The FASTQs sequencing data are aligned to the human genome (hg19) and processed using IonTorrent Suite V5.0.4.0 This package included the Torrent Variant Caller V5.0.4.0 using the built-in ‘Somatic-low stringency’ with optimised parameters to automatically call variants with allelic ratio >2%.

Real-time quantitative PCRs (qPCRs) were performed to characterise EMT and stem-cell markers in tumours using a set of molecular markers; five mesenchymal markers (TWIST1, N-CADHERIN, ZEB1, SNAI1 and VIMENTIN), two epithelial markers (E-CADHERIN and JUP) and two stem-cells markers (TCF3 and CD44), as recommended by guidelines [24]. RNAs (1 μg) were reverse transcribed using the High Capacity cDNA Reverse Transcription Kit (Applied Biosystem, Foster City, CA). E-CADHERIN, JUP, TWIST1, N-CADHERIN, ZEB1, SNAI1, VIMENTIN, TCF3 and CD44 messenger RNA (mRNA) levels were quantified using FAM Taqman probes on an ABI Prism 7900 Sequence Detector System (TaqMan™ Gene Expression Assay, Applied Biosystems). Measures were performed in triplicate with 10 ng/µL of complementary DNA (cDNA) using Taqman Gene Expression Master Mix (Applied Biosystems). In each run, normal human lung RNAs from a pool of normal lung tissues (n = 10) was used as calibrator. We performed duplex analysis using 18S RNAs (VIC probe, Applied Biosystems) as an endogenous gene control. Low-quality RNA samples (18S RNA Ct > 16) were discarded from the analysis. Expression levels were calculated as described by Livak and Schmittgen using the ΔΔCt method [25]. Data were analysed using the RT2 profiler PCR array Data Analysis Web portal (Qiagen). Fold change between controls and samples defined down (between 0 and 0.5), stable (0.5–2) or up-regulated (>2) markers. As we quantified two epithelial markers and five mesenchymal markers, we weighted results to create an EMT score to classify tumours: for epithelial markers (E-CADHERIN and JUP), down, stable and up-regulation were weighted as 10, 5 and 0 points; for mesenchymal markers (TWIST1, N-CADHERIN, ZEB1, SNAI1 and VIMENTIN), down, stable and up-regulation were weighted as 0, 2 and 6 points. EMT score was the sum of epithelial and mesenchymal markers. Low to high EMT scores classify sample phenotypes from epithelial to mesenchymal. This classification was established a priori, and tumours were classified blindly of miR levels and clinical data. This score was validated by comparison to two previously published EMT signatures using linear regression (Spearman’s correlation) 76GS: 76 Gene signature method [26] (Supplemental Fig. 1A) and KS: a two-sample Kolmogorov–Smirnov test (Supplemental Fig. 1B) [27]. We found a good correlation between our EMT score and published signatures (r = −0.58, p < 10−5 and r = 0.65, p < 10−5, respectively), consistent with the one existing between the two validated signatures (r = −0.77, p < 10−5, Supplemental Fig. 1C) [28]. Our 7-genes score is an approximation of minima of the EMT status and can be used as a surrogate marker to score EMT.

MiRs were quantified by quantitative reverse transcription-PCR. RNAs (10 ng/µL) were reversed transcribed using the TaqMan Advanced miRNA cDNA Synthesis Kit (Applied Biosystem). MiR levels were quantified by qPCR using Taqman probes (Taqman Advanced miRNA Assay, clone ID 478490_mir and clone ID 477849_mir, Thermo Fisher Scientific) on an ABI Prism 7900 System (Applied Biosystems). Real-time PCR was performed in triplicate with 3.3 ng of cDNA and the Taqman Fast Advanced Master Mix (Applied Biosystems). In each run, normal human lung RNA as described above was used as a calibrator. MiR-423 and miR-425 (clone ID 478327_mir and clone ID 478094_mir, Thermo Fisher Scientific) were used for normalisation and expression levels were calculated as described for EMT markers.

DNA methylation

DNA methylation level at miR-200 promoters was measured by fragment analysis. DNA underwent bisulfite treatment. Bisulfite and methylation-sensitive primers were designed with Methprimer. For a specific CpG site, two couples were designed with R primer localised on a CpG site and F outside of any CpG site. R primers were HEX tagged for methylated fragments and FAM tagged for non-methylated fragments. Migration was performed on ABI3730xl sequencer data and analysed with Genemapper software (Thermo Fisher diagnostics). Methylation level was defined as the ratio of methylated peak height/(methylated + non-methylated peak height). Methylation states were grouped in quartiles for analysis.

Statistical analyses

The association between continuous variables was assessed by Pearson’s correlation and the association between categorical variables was assessed by Wilcoxon’s test. Survival curves were estimated by the Kaplan–Meier method. Statistical comparisons between survival distributions were made using the log-rank test. Multivariate analyses were performed using the Cox proportional hazards model for overall and disease-free survival (DFS) analyses. All data analyses were conducted with two-sided tests; a p value < 0.05 was statistically significant (R V3.5.2). miR clustering was done using the Ward method with JMP (SAS Institute JMP, France).

Results

Patients and tumour characteristics

Clinical characteristics are shown in Table 1: the average age of patients was 65 years. The disease stage was distributed as follows: 78 patients stage I (44%), 45 (25%) stage II, 47 stage III (26%) and 6 (3%) stage IV. Patients had lobectomy (n = 131), bilobectomy (n = 7), pneumonectomy (n = 17) or atypical resection (n = 11). The median follow-up duration was 42 months. Less than a half of the patients had adjuvant chemotherapy (n = 64) or radiotherapy (n = 26) and 53 patients had relapsed or died at 3 years. The most frequent genetic alteration was, as expected, TP53 mutations followed by KRAS and EGFR (Table 2A). Molecular alterations in TP53, KRAS or EGFR were not linked to age, stage, relapse or death at 3 years. EGFR mutations were more frequent in women (p = 0.03) and in non-smokers (p = 0.006) and TP53 mutations were more frequent in smokers (p = 0.03), no relation with tobacco exposure was identified in the KRAS-mutated group.

Table 1.

Clinical, surgical and oncological characteristics of samples.

Clinics
Sex ratio (F/M) 55/121
Age (yr) ± SD 65 ± 11
IMC (kg/m2) ± SD 25 ±  4
VEMS (L/min) ± SD 81 ± 21
Comorbidities
Cardiovascular 81 (46%)
Diabetes 23 (13%)
Cancer (other) 45 (26%)
Surgery
Surgical approach
Thoracotomy, n (%) 157 (89%)
Video-assisted, n (%) 17 (10%)
Robotic, n (%) 2 (1%)
Surgical resection
Sub-lobar, n (%) 12 (7%)
Lobectomy, n (%) 136 (77%)
Bilobectomy, n (%) 9 (6%)
Pneumonectomy, n (%) 19 (10%)
Complete resection (R0), n (%) 176 (100%)
Side (D/G) 103/73
Lymph nodes resection
Radical, n (%) 166 (94%)
Sampling, n (%) 10 (6%)
Duration (min) ± SD 182 ± 64
Post-operative course
Infection, n (%) 25 (14%)
Cardiovascular, n (%) 16 (9%)
Prolonged air leaks, n (%) 21 (12%)
Death, n (%) 4 (2%)
Length of stay (days) ±  SD 13 ± 8
Oncology
Histological type
Adenocarcinomas, n (%) 117 (66%)
Squamous cell, n (%) 46 (26%)
Large cell, n (%) 9 (5%)
Carcinosarcomas n (%) 4 (2%)
TNM (IASLC 2009)
Tx, n (%) 3 (2%)
T1, n (%) 55 (31%)
T2, n (%) 77 (44%)
T3, n (%) 35 (20%)
T4, n (%) 6 (3%)
Nx 6 (3%)
N0, n (%) 99 (56%)
N1, n (%) 32 (18%)
N2, n (%) 39 (22%)
M0, n (%) 170 (97%)
M1, n (%) 6 (3%)
Stage
IA, n (%) 43 (24%)
IB, n (%) 35 (20%)
IIA, n (%) 9 (5%)
IIB, n (%) 36 (20%)
IIIA, n (%) 43 (24%)
IIIB, n (%) 4 (2%)
IV, n (%) 6 (3%)
Perioperative treatment
Neoadjuvant chemotherapy 20 (11%)
Adjuvant chemotherapy 66 (38%)
Adjuvant radiation therapy 26 (15%)

Table 2.

Description of molecular alterations.

(A) Mutations
Mutated genes Alteration in n cases (%)
KRAS 51 (31%)
BRAF 5 (3%)
NRAS 4 (2%)
TP53 75 (45%)
PIK3CA 7 (4%)
CTNNB1 3 (2%)
SMAD4 2 (1%)
FBXW7 1 (1%)
STK11 10 (6%)
EGFR 17 (10%)
ERBB2 2 (1%)
AKT1 2 (1%)
ERBB4 3 (2%)
FGFR3 1 (1%)
PTEN 2 (1%)
DDR2 1 (1%)
MAP2K1 1 (1%)
At least 1 alteration 138 (84%)
1 alteration 75 (45%)
2 alterations 49 (30%)
3 alterations or more 14 (8%)
(B) MiRs and EMT marker
Up Stable Down
miR
  miR-200a-3p 52 (32%) 91 (55%) 21 (13%)
  miR-200b-3p 91 (57%) 54 (34%) 14 (9%)
  miR-429-3p 83 (51%) 64 (39%) 17 (10%)
  miR-200c-3p 72 (45%) 70 (44%) 18 (11%)
  miR-141-3p 104 (65%) 42 (26%) 14 (9%)
Epithelial markers
  E-cadherin 51 (35%) 57 (39%) 37 (26%)
  JUP 58 (40%) 59 (41%) 28 (19%)
Mesenchymal markers
  Twist1 72 (50%) 32 (22%) 41 (28%)
  N-cadherin 62 (43%) 43 (30%) 40 (28%)
  ZEB1 24 (17%) 46 (32%) 75 (52%)
  SNAI1 20 (14%) 43 (30%) 82 (57%)
  VIM 24 (17%) 44 (30%) 77 (53%)
Stem-cell markers
  TCF3 44 (30%) 62 (43%) 39 (27%)
  CD44 35 (24%) 53 (37%) 57 (39%)
 EMT score (mean ± SD) 19.7 ± 7.6

Mutations, using NGS Colon-Lung v2 Cancer Panel (A); miR-200a, miR-429, miR-200b, miR-200c, miR-141, EMT and stem-cell markers expression levels (B) defined as up, stable and low as described in ‘Materials and methods’.

(A) Mutations and (B) MiRs and EMT marker.

EMT tumour profiles

EMT gene expression and miR quantification are described in Table 2B. Because EMT progresses through dynamic intermediate states, we explored EMT’s value using a 7-genes score as described in the ‘Materials and methods’ section. EMT score ranged from 0 to 40 (theoretical maximum: 0–50); mean was 19.7 [18.5–21]IC95% (quartiles 15–24). The existence of tumours expressing epithelial and mesenchymal markers and the existence of a correlation between epithelial and mesenchymal markers illustrates the predominance of hybrid states (Fig. 1).

Fig. 1. Correlation matrix of miR-34, miR-200, promoter methylation, epithelial and mesenchymal marker expression.

Fig. 1

Circles represent the correlation coefficient when significant; the cross represents the absence of correlation.

EMT score inversely correlated with miR-429 (p = 0.004), miR-200b (p = 0.004), miR-200a (p < 0.0001), miR-200c (p = 0.003) and miR-141 (p = 0.001) was associated with methylation at chr-1 (p = 0.0003) and chr-12 (p = 0.0002) (Supplemental Fig. 2) and with high expression of stem-cell markers (p < 0.0001). EMT score was not linked to smoking, tumour mutational status, sex, age or stage (data not shown).

EMT’s prognostic value was explored using the EMT score either tested as a continuous variable or grouped in quartiles with the lower quartile defining the epithelial state, the two intermediate quartiles the transition state and the higher quartile the mesenchymal state. No association was found between the EMT score and RFS or OS (Supplemental Fig. 3).

MiR tumour profiles

Expression of miR-200 family members strongly correlated together within chromosome clusters: miR-200a, b and miR-429 on chr-1, and miR-200c and miR-141 on chr-12 (Fig. 1). However, correlations were weak or absent between chr-1 or chr-12. MiR-200a inversely correlated with SNAI1, TCF3 and CD44, whereas miR-34a positively correlates with CDH1, ZEB1, VIM, TCF7 and CD44. MiR promoter methylation at chromosome 1 was related to low miR-200a, b and 429, methylation at chr-12 was not related to low chr-12 miR-200s. High miR-429 (p = 0.02) and miR-200b (p = 0.002) associate with the female gender; no links were found between miRs and other clinical data including stage and node metastasis. Up-regulation of chr-12 miRs, miR-200c (p = 0.04) and miR-141 (p = 0.03), were associated with KRAS mutations at the opposite; up-regulation of chr-1 miRs, miR-200b (p = 0.02) and miR-429 (p = 0.004), were associated with EGFR mutations. TP53 mutations were inversely correlated with miR-200a (p = 0.004), miR-200c (p = 0.005) and miR-141 (p = 0.03). As miRs analysed individually moderately associate with EMT markers, we tested whether miR profiles might be more informative.

miR profile analyses

We used hierarchical clustering to classify tumours based on miR expression and explored whether identified clusters exhibited different prognostic profiles (Fig. 2). MiR expression data and methylation status at chr-1 and chr-12 were used to create clusters. Parallel plots were generated to identify gene profiles within each cluster. Cluster 1 is defined by high expression of miR-429, miR-200a and intermediate miR-200b, but low or absence of expression of miR-141 and miR-200c; cluster 2 has lower expression of miR-429, miR-200a and higher expression of miR-200b, miR-141 and miR-200c; cluster 3 is characterised by high to intermediate methylation at chr-1 and miR-34a expression; cluster 4 is characterised by high methylation at chr-1 and chr-12 and very low expression of all miRs markers, and cluster 5 is defined by high expression of miR-34b and miR-34c (Fig. 2). MiR clusters were associated with the EMT score; lower scores defining epithelial samples are seen in clusters 1 and 2 (Supplemental Fig. 4). Expression of ZEB1, SNAIL and TWIST was statistically different between miR clusters (p = 0.02, 0.03 and 0.002, respectively) and TP53 mutations were more frequent in cluster 4 (p = 0.03) (Supplemental Fig. 5). No association was found with sex, age, stage or node metastasis.

Fig. 2. Unsupervised hierarchical clustering based on miR-34, miR-200 expression and promoter methylation status.

Fig. 2

Five different clusters with differential methylation or miRs expression profiles are identified.

Core EMT miRs and prognosis in NSCLC

We assessed links between miRs, clinical data and tumour phenotype. Univariate analysis showed that age, TP53, EGFR or KRAS mutations, individual EMT markers or EMT score had no impact on OS or RFS (data not shown). RFS was associated to stage (p < 0.0001) and to chr-1-miR-200 expression miR-200a-3p (p = 0.08), miR-200b (p = 0.05) and miR-429 (p = 0.025) (Supplemental Fig. 6A–C). No association was identified with chr-12-miR-200s or miR-34s. OS was associated to stage (p < 0.0001), neoadjuvant chemotherapy (p < 0.001), lymph nodes invasion (p < 0.001), to chr-1-miR-200 expression miR-200a (p = 0.016), miR-429 (p = 0.034) and miR-200b (p = 0.08) (Supplemental Fig. 6D–F). Multivariate analysis including stage showed that chr-1 miR-200s were independent predictors of OS and a trend was seen for DFS, especially for miR-429 (p = 0.059) (Supp Table 1). No prognostic value was found for chr-12 miR-200s or miR-34s. Kaplan–Meir survival curves confirmed that low expression of miR-200a, mir-200b and miR-429 was linked to shorter OS (p = 0,013, p = 0.003, p = 0.006, respectively) and shorter DFS (p = 0.08, 0.05 and 0.025, respectively) (Fig. 3). Our results were validated by The Cancer Genome Atlas (TCGA) data showing that indeed chr-1 miR-200s and not chr-12 miR-200s had a predictive value for OS in NSCLC (Supplemental Fig. 7).

Fig. 3. Kaplan–Meier OS and RFS plots.

Fig. 3

Disease-free survival according to miR-200a (a), miR-200b (b) and miR-429 (c) expressions. Overall survival according to miR-200a (d), miR-200b (e) and miR-429 (f) expressions.

Methylation status was associated with low miR-200 expression (Fig. 1). Using 80% methylation ratio as a cut-off, lower OS was identified in the high methylated group (p = 0.0079), and a trend was identified for lower DFS (p = 0.057) (Fig. 4a, b).

Fig. 4. Kaplan–Meier OS and RFS plots.

Fig. 4

Disease-free survival (a) and overall survival (b) according to different miR clusters showing two sub-groups, clusters 1 and 5, with low relapse risk (a) and longer OS (b). Disease-free survival (c) and overall survival (d) according to miR-200-chromosome-1 methylation status.

Considering miR clusters, we showed that clusters 2, 3 and 4 were linked to higher death (p = 0.002) and relapse (p = 0.02) at 3 years, and nearly 50% of patients had died or relapsed as compared to clusters 1 and 5, for which only 2 out of 18 and 1 out of 17 had died at 3 years in clusters 1 and 5, respectively. Kaplan–Meir survival curves confirmed that tumours with miR profiles corresponding to clusters 1 and 5 are of low risk of relapse (p = 0.064) (Fig. 4c) and death (p = 0.021) (Fig. 4d).

Discussion

Patients treated by surgery for localised lung cancer have distinct prognosis and relapse risk. Perioperative treatments have an unsatisfactory benefit, with a 5-year OS rate increase ranging from 5 to 8% [29, 30]. The management of patients with localised diseases is rapidly evolving with new perioperative treatments available as immunotherapies and targeted therapies [3]. With the diversification of treatment options, tumour molecular characterisation will be necessary for patients with localised diseases to identify patients with a high risk of relapse. Epithelial-to-mesenchymal transition is a cellular process that drives metastasis and resistance to treatment, suggesting that the tumour EMT phenotype could be a valuable marker of aggressiveness. The absence of prognosis value of the EMT status itself may seem contradictory; however, due to EMT plasticity, the strength of EMT evaluation to predict survival might be low. Moreover, low miR may reflect the propensity of cells to adapt through an EMT programme, and the hybrid E/M phenotype may have multiple advantages as compared to a complete mesenchymal state to favour metastasis due to collective migrating cell clusters [31]. Gene signatures were used to score EMT. A 76-gene signature [26] predicted resistance to EGFR inhibitors in vitro, but had no prognostic value in vivo; a 16-gene signature [32] was linked to immune tumour infiltration, but not to prognosis, and finally, a 12-gene signature predicted responses to immune checkpoint inhibitors and was linked to OS and RFS in patients receiving immune checkpoint inhibitors [33]. Our EMT score was not associated with OS or RFS in line with the previous series. EMT signatures may not capture well enough cell plasticity that may be of importance to evaluate tumour aggressiveness. Indeed, most tumours showed co-expression of epithelial and mesenchymal markers corresponding to different hybrid states. Analysing upstream EMT regulatory loops might drive important information on cell capacities to undergo E to M switches. The miR-200, miR-34, ZEB1, SNAIL regulation circuit is a hub that governs cell fate [34]. We hypothesis that quantifying core circuit regulators may help identify tumours that are prone to acquire EM plasticity. In this series, miR-200 down-regulation and high EMT scores were observed in broadly 10% of samples. No association between miR-200s down-regulation and up-regulation of their direct target ZEB1 was found. Bracken et al. showed that the promoter shared by miR-200a, miR-200b and miR-429 was repressed in mesenchymal cells by ZEB1 and SIP1 [35, 36], and Lindner et al. showed that ZEB1 alters the epigenetic landscape in colorectal cancer cells [37]. Depending on extracellular stimulations, the miR-200/ZEB1/2 equilibrium may turn on epithelial or mesenchymal markers, and feedback loops may explain the absence of association between miR-200s and ZEB1/2 in this series. Our study showed that miRs belonging to the miR-200 family but not miR belonging to the miR-34 family were linked to the EMT score. The miR-34/SNAIL loop may act as a side regulator that does not give rise to phenotypic transition by itself. However, hierarchical clustering showed that the miR-34b- and miR-34c-positive sub-group was of good prognosis and low relapse risk, even though these samples had low miR-200s and high EMT scores. In line with this, differences between miR-34a, c and b were shown and miR-34b and c were found more effective tumour suppressors as compared to miR-34a [38]. Here we found that miR-34b/c expression could rescue miR-200 loss. One of the main results brought by this study is the identification of major differences between both miR-200 clusters. We showed that the molecular background, as for KRAS, EGFR and TP53 associates with miR-200s expression. TP53 mutations were more frequent in cluster 4, characterised by high methylation and low miR-200s. Links between miR-200 and TP53 exist: in TP53-mutated breast cancer, loss of miR-200c promotes carcinogenesis. Moreover, wild-type TP53 transactivates miR-200s, and, in turn, regulates ZEB1/2 [39]. Our results suggested that in NSCLC, mutant TP53 could lose its ability to up-regulate miR-200s and therefore be an important regulator of EMT. The seed miR sequences that differ among miR-200 may direct at different 3′-untranslated region sequences of target mRNAs. In lung cancer models, miR-200 exhibits a dual function in lung cancer progression: often described as tumour suppressors, they have also been associated with tumour progression as for miR-429 in cellular models [18, 40] and for miR-141 [41]. This suggests that first the biological functions of miR-200s members may be different and second may depend on environment and tissue types. Up-regulation of miR-200c and miR-141 has been associated with poor survival rates in early-stage NSCLC [23, 42]. Here we showed, in accordance with TCGA data, that miR-141 and miR-200c do not predict survival in NSCLC. In line with that, a meta-analysis testing the prognostic value of miR-141 in cancer showed large differences between cancer types and studies [43]. In our study, chr-12 miRs seemed much less regulated by promoter methylation as compared to chr-1 miRs. Considering the miR-ZEB1-SNAIL regulation hub, methylation might unbalance the regulation loops towards the mesenchymal side. The hypothesis that a methylator phenotype or DNMT dysregulation could be at the origin of miR-200a, b and miR429 down-regulation in lung cancer needs to be validated. In colorectal cancer, miR-200 promoter methylation was associated with the CMS4 subtype that is known to have a mesenchymal phenotype and a worse prognosis [44]. The importance of methylation in driving EMT and prognosis through down-regulation of chr-1 miR-200s brings new opportunities. Indeed, miR-200 promoter hypermethylation may be used as a surrogate marker of miR quantification and a target to reverse the EMT process. As EMT was largely associated with resistance to treatments, combinations might increase response rates. MiR-200 was implicated in immunomodulation by targeting PDL-1 [45], in angiogenesis [16] and in targeted therapies resistance [46], suggesting a wide range of situations in which EMT-miR evaluation might be of interest to personalise treatment options.

This work is the first identification of a different impact on prognosis between chr-1 and chr-12 miR-200 clusters confirmed on the TCGA data set. We show an unrevealed link between down-regulation of chr-1 miR-200s (miR-200a, miR-200b and miR-429) and survival or relapse in localised NSCLC. Moreover, using miR clustering, we identified a group of samples with low relapse risk and good overall survival characterised by high EMT score, low miR-200 but high miR-34b and c. This study provides new insights on the clinical impact of miR-200s family members in localised NSCLC. We believe that using EMT markers with the TNM classification could trigger adjuvant treatment decisions and that EMT itself could be targeted in a subset of lung cancer patients. Here we show that miR-200 should no longer be considered as a homogeneous group, and that core EMT regulators quantification is promising to identify NSCLC patients with a low risk of relapse after surgery.

Supplementary information

Checklist form (1.8MB, pdf)
Supplemental Figure 1 (168.5KB, pptx)
Supplemental Figure 2 (264.7KB, pptx)
Supplemental Figure 3 (82.5KB, ppt)
Supplemental Figure 4 (69KB, ppt)
Supplemental Figure 5 (55KB, pptx)
Supplemental Figure 6 (153.8KB, pptx)
Supplemental Figure 7 (305.1KB, pptx)
Supplemental Table 1 (10.1KB, xlsx)

Acknowledgements

We thank Claudia DeToma, Marine Largeau, Elodie Michel and Lauriane Chambolle for their work and implication at the Biological Resources centre and Tumour Bank Platform PRB-HEGP (BB-0033-00063).

Author contributions

Conception or design: HB, SG and AL. Data collection: J-BL, FLP-B, AL, LG, CB, TD and AP. Data analysis: SG and AL. Redaction: AL, SG and HB. Final approval: SG, AD, TD, AP, GB, J-BL, J-BO, LG, CB, FLP-B, PL-P, AL and HB.

Funding information

This study was supported by a grant from the integrated site of cancer research ‘cancer research for personalised medicine’ (SIRIC CARPEM). AL received personal research grants from Fondation de la Recherche Médicale. We thank the Tumour Bank Platform (PRB-HEGP BB-0033-00063) for its support.

Ethics approval and consent to participate

This study, conducted at the European Georges Pompidou hospital, was approved by CPP Ile de France 2 ethics committee (nos. 2012-08-09 and 2012-08-09 A1) and registered in clinical trial.gov (NCT03509779). Patients with NSCLC treated by surgery for curative intent signed informed consent for research and tumour tissues banking. Samples were stored frozen (−80 °C) at the Biological Resources centre and Tumour Bank Platform (PRB-HEGP BB-0033-00063).

Competing interests

AL reports grants from Fondation de la Recherche Médicale, during the conduct of the study. HB reports a grant from Site de la Recherche Intégrée sur le Cancer (SIRIC) CARPEM for this study.

Footnotes

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

These authors contributed equally: Antoine Legras, Helene Blons.

Supplementary information

The online version contains supplementary material available at 10.1038/s41416-021-01568-7.

References

  • 1.Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108. doi: 10.3322/caac.21262. [DOI] [PubMed] [Google Scholar]
  • 2.Noone A, Howlader N, Krapcho M, Miller D, Brest A, Yu M, et al. (eds). SEER Cancer Statistics Review, 1975–2015. Bethesda:National cancer Institute; 2018.
  • 3.Wu Y-L, Tsuboi M, He J, John T, Grohe C, Majem M, et al. Osimertinib in resected EGFR-mutated non-small-cell lung cancer. N. Engl J Med. 2020;383:1711–23. doi: 10.1056/NEJMoa2027071. [DOI] [PubMed] [Google Scholar]
  • 4.Legras A, Pécuchet N, Imbeaud S, Pallier K, Didelot A, Roussel H, et al. Epithelial-to-mesenchymal transition and micrornas in lung cancer. Cancers. 2017;9:101. [DOI] [PMC free article] [PubMed]
  • 5.Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119:1420–8. doi: 10.1172/JCI39104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Thiery JP, Acloque H, Huang RYJ, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–90. doi: 10.1016/j.cell.2009.11.007. [DOI] [PubMed] [Google Scholar]
  • 7.Zaravinos A. The regulatory role of microRNAs in EMT and cancer. J Oncol. 2015;2015:865816. doi: 10.1155/2015/865816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Zavadil J, Böttinger EP. TGF-beta and epithelial-to-mesenchymal transitions. Oncogene. 2005;24:5764–74. doi: 10.1038/sj.onc.1208927. [DOI] [PubMed] [Google Scholar]
  • 9.Lo H-W, Hsu S-C, Xia W, Cao X, Shih J-Y, Wei Y, et al. Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelial-mesenchymal transition in cancer cells via up-regulation of TWIST gene expression. Cancer Res. 2007;67:9066–76. doi: 10.1158/0008-5472.CAN-07-0575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Tania M, Khan MA, Fu J. Epithelial to mesenchymal transition inducing transcription factors and metastatic cancer. Tumour Biol. 2014;35:7335–42. doi: 10.1007/s13277-014-2163-y. [DOI] [PubMed] [Google Scholar]
  • 11.Zhang P, Sun Y, Ma L. ZEB1: at the crossroads of epithelial-mesenchymal transition, metastasis and therapy resistance. Cell Cycle. 2015;14:481–7. doi: 10.1080/15384101.2015.1006048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Gemmill RM, Roche J, Potiron VA, Nasarre P, Mitas M, Coldren CD, et al. ZEB1-responsive genes in non-small cell lung cancer. Cancer Lett. 2011;300:66–78. doi: 10.1016/j.canlet.2010.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Pallier K, Cessot A, Côté J-F, Just P-A, Cazes A, Fabre E, et al. TWIST1 a new determinant of epithelial to mesenchymal transition in EGFR mutated lung adenocarcinoma. PLoS ONE. 2012;7:e29954. doi: 10.1371/journal.pone.0029954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gregory PA, Bracken CP, Bert AG, Goodall GJ. MicroRNAs as regulators of epithelial-mesenchymal transition. Cell Cycle. 2008;7:3112–8. doi: 10.4161/cc.7.20.6851. [DOI] [PubMed] [Google Scholar]
  • 15.Meng F, Wu G. The rejuvenated scenario of epithelial-mesenchymal transition (EMT) and cancer metastasis. Cancer Metastasis Rev. 2012;31:455–67. doi: 10.1007/s10555-012-9379-3. [DOI] [PubMed] [Google Scholar]
  • 16.Pecot CV, Rupaimoole R, Yang D, Akbani R, Ivan C, Lu C, et al. Tumour angiogenesis regulation by the miR-200 family. Nat Commun. 2013;4:2427. doi: 10.1038/ncomms3427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Chen L, Gibbons DL, Goswami S, Cortez MA, Ahn Y-H, Byers LA, et al. Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression. Nat Commun. 2014;5:5241. doi: 10.1038/ncomms6241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lang Y, Xu S, Ma J, Wu J, Jin S, Cao S, et al. MicroRNA-429 induces tumorigenesis of human non-small cell lung cancer cells and targets multiple tumor suppressor genes. Biochem Biophys Res Commun. 2014;450:154–9. doi: 10.1016/j.bbrc.2014.05.084. [DOI] [PubMed] [Google Scholar]
  • 19.Title AC, Hong S-J, Pires ND, Hasenöhrl L, Godbersen S, Stokar-Regenscheit N, et al. Genetic dissection of the miR-200–Zeb1 axis reveals its importance in tumor differentiation and invasion. Nat Commun. 2018;9:4671. doi: 10.1038/s41467-018-07130-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Damiano V, Brisotto G, Borgna S, di Gennaro A, Armellin M, Perin T, et al. Epigenetic silencing of miR-200c in breast cancer is associated with aggressiveness and is modulated by ZEB1: MIR-200C LOCUS METHYLATION IN BREAST CANCER. Genes Chromosomes Cancer. 2017;56:147–58. doi: 10.1002/gcc.22422. [DOI] [PubMed] [Google Scholar]
  • 21.Pichler M, Ress AL, Winter E, Stiegelbauer V, Karbiener M, Schwarzenbacher D, et al. MiR-200a regulates epithelial to mesenchymal transition-related gene expression and determines prognosis in colorectal cancer patients. Br J Cancer. 2014;110:1614–21. doi: 10.1038/bjc.2014.51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Xue B, Chuang C-H, Prosser HM, Fuziwara CS, Chan C, Sahasrabudhe N, et al. miR-200 deficiency promotes lung cancer metastasis by activating cancer-associated fibroblasts. bioRxiv:2020.09.02.276550 [Preprint]. 2020. 10.1101/2020.09.02.276550. [DOI] [PMC free article] [PubMed]
  • 23.Tejero R, Navarro A, Campayo M, Viñolas N, Marrades RM, Cordeiro A, et al. miR-141 and miR-200c as markers of overall survival in early stage non-small cell lung cancer adenocarcinoma. PLoS ONE. 2014;9:e101899. [DOI] [PMC free article] [PubMed]
  • 24.On behalf of the EMT International Association (TEMTIA. Yang J, Antin P, Berx G, Blanpain C, Brabletz T, et al. Guidelines and definitions for research on epithelial–mesenchymal transition. Nat Rev Mol Cell Biol. 2020;21:341–52. doi: 10.1038/s41580-020-0237-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 2001;25(Dec):402–8. doi: 10.1006/meth.2001.1262. [DOI] [PubMed] [Google Scholar]
  • 26.Byers LA, Diao L, Wang J, Saintigny P, Girard L, Peyton M, et al. An epithelial-mesenchymal transition gene signature predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhibitor resistance. Clin Cancer Res. 2013;19:279–90. doi: 10.1158/1078-0432.CCR-12-1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Tan TZ, Miow QH, Miki Y, Noda T, Mori S, Huang RY-J, et al. Epithelial-mesenchymal transition spectrum quantification and its efficacy in deciphering survival and drug responses of cancer patients. EMBO Mol Med. 2014;6:1279–93. doi: 10.15252/emmm.201404208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Chakraborty P, George JT, Tripathi S, Levine H, Jolly MK. Comparative study of transcriptomics-based scoring metrics for the epithelial-hybrid-mesenchymal spectrum. Front Bioeng Biotechnol. 2020;8:220. [DOI] [PMC free article] [PubMed]
  • 29.Pignon J-P, Tribodet H, Scagliotti GV, Douillard J-Y, Shepherd FA, Stephens RJ, et al. Lung adjuvant cisplatin evaluation: a pooled analysis by the LACE Collaborative Group. J Clin Oncol. 2008;26:3552–9. doi: 10.1200/JCO.2007.13.9030. [DOI] [PubMed] [Google Scholar]
  • 30.NSCLC Meta-analyses Collaborative Group. Arriagada R, Auperin A, Burdett S, Higgins JP, Johnson DH, et al. Adjuvant chemotherapy, with or without postoperative radiotherapy, in operable non-small-cell lung cancer: two meta-analyses of individual patient data. Lancet. 2010;375:1267–77. doi: 10.1016/S0140-6736(10)60059-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Jolly MK, Boareto M, Huang B, Jia D, Lu M, Ben-Jacob E, et al. Implications of the hybrid epithelial/mesenchymal phenotype in metastasis. Front Oncol. 2015;5:155. doi: 10.3389/fonc.2015.00155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Young KC, Sangmin C, Taeyeong K, Jonathan A, Sarita A, Wade I, et al. Epithelial-mesenchymal transition (EMT) signature is inversely associated with T-cell infiltration in non-small cell lung cancer (NSCLC). Sci Rep. 2018;8:2918. [DOI] [PMC free article] [PubMed]
  • 33.Thompson JC, Hwang W-T, Davis C, Deshpande C, Jeffries S, Rajpurohit Y, et al. Gene signatures of tumor inflammation and epithelial-to-mesenchymal transition (EMT) predict responses to immune checkpoint blockade in lung cancer with high accuracy. Lung Cancer. 2020;139:1–8. doi: 10.1016/j.lungcan.2019.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Jolly MK, Boareto M, Huang B, Jia D, Lu M, Ben-Jacob E, et al. Implications of the hybrid epithelial/mesenchymal phenotype in metastasis. Front Oncol. 2015;5:155. doi: 10.3389/fonc.2015.00155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bracken CP, Gregory PA, Kolesnikoff N, Bert AG, Wang J, Shannon MF, et al. A double-negative feedback loop between ZEB1-SIP1 and the microRNA-200 family regulates epithelial-mesenchymal transition. Cancer Res. 2008;68:7846–54. doi: 10.1158/0008-5472.CAN-08-1942. [DOI] [PubMed] [Google Scholar]
  • 36.Hill L, Browne G, Tulchinsky E. ZEB/miR-200 feedback loop: at the crossroads of signal transduction in cancer. Int J Cancer. 2013;132:745–54. doi: 10.1002/ijc.27708. [DOI] [PubMed] [Google Scholar]
  • 37.Lindner P, Paul S, Eckstein M, Hampel C, Muenzner JK, Erlenbach-Wuensch K, et al. EMT transcription factor ZEB1 alters the epigenetic landscape of colorectal cancer cells. Cell Death Dis. 2020;11:147. doi: 10.1038/s41419-020-2340-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kim JS, Kim EJ, Lee S, Tan X, Liu X, Park S, et al. MiR-34a and miR-34b/c have distinct effects on the suppression of lung adenocarcinomas. Exp Mol Med. 2019;51:9. [DOI] [PMC free article] [PubMed]
  • 39.Kim T, Veronese A, Pichiorri F, Lee TJ, Jeon Y-J, Volinia S, et al. p53 regulates epithelial–mesenchymal transition through microRNAs targeting ZEB1 and ZEB2. J Exp Med. 2011;208:875–83. doi: 10.1084/jem.20110235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Xiao P, Liu W, Zhou H. miR-429 promotes the proliferation of non-small cell lung cancer cells via targeting DLC-1. Oncol Lett. 2016;12:2163–8. doi: 10.3892/ol.2016.4904. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 41.Mei Z, He Y, Feng J, Shi J, Du Y, Qian L, et al. MicroRNA-141 promotes the proliferation of non-small cell lung cancer cells by regulating expression of PHLPP1 and PHLPP2. FEBS Lett. 2014;588:3055–61. doi: 10.1016/j.febslet.2014.06.020. [DOI] [PubMed] [Google Scholar]
  • 42.Zhu W, He J, Chen D, Zhang B, Xu L, Ma H, et al. Expression of miR-29c, miR-93, and miR-429 as potential biomarkers for detection of early stage non-small lung cancer. PLoS ONE. 2014;9:e87780. doi: 10.1371/journal.pone.0087780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Wang F, Zhang L, Xu H, Li R, Xu L, Qin Z, et al. The significance role of microRNA-200c as a prognostic factor in various human solid malignant neoplasms: a meta-analysis. J Cancer. 2019;10:277–86. doi: 10.7150/jca.27536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.De Sousa E, Melo F, Wang X, Jansen M, Fessler E, Trinh A, et al. Poor-prognosis colon cancer is defined by a molecularly distinct subtype and develops from serrated precursor lesions. Nat Med. 2013;19:614–8. doi: 10.1038/nm.3174. [DOI] [PubMed] [Google Scholar]
  • 45.Noman MZ, Janji B, Abdou A, Hasmim M, Terry S, Tan TZ, et al. The immune checkpoint ligand PD-L1 is upregulated in EMT-activated human breast cancer cells by a mechanism involving ZEB-1 and miR-200. Oncoimmunology. 2017;6:e1263412. doi: 10.1080/2162402X.2016.1263412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Tulchinsky E, Demidov O, Kriajevska M, Barlev NA, Imyanitov E. EMT: a mechanism for escape from EGFR-targeted therapy in lung cancer. Biochim Biophys Acta. 2019;1871:29–39. doi: 10.1016/j.bbcan.2018.10.003. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Checklist form (1.8MB, pdf)
Supplemental Figure 1 (168.5KB, pptx)
Supplemental Figure 2 (264.7KB, pptx)
Supplemental Figure 3 (82.5KB, ppt)
Supplemental Figure 4 (69KB, ppt)
Supplemental Figure 5 (55KB, pptx)
Supplemental Figure 6 (153.8KB, pptx)
Supplemental Figure 7 (305.1KB, pptx)
Supplemental Table 1 (10.1KB, xlsx)

Articles from British Journal of Cancer are provided here courtesy of Cancer Research UK

RESOURCES