Purpose:

Anti-EGFR mAbs are effective in the treatment of metastatic colorectal cancer (mCRC) patients. RAS status and tumor location (sidedness) are predictive markers of patients' response to anti-EGFR mAbs. Recently, low miR-31-3p expression levels have been correlated with clinical benefit from the anti-EGFR mAb cetuximab. Here, we aimed to validate the predictive power of miR-31-3p in a prospective cohort of chemorefractory mCRC patients treated with single-agent anti-EGFR mAbs.

Experimental Design:

miR-31-3p was tested by in situ hybridization (ISH) in 91 pretreatment core biopsies from metastatic deposits of 45 patients with mCRC. Sequential tissue biopsies obtained before treatment, at the time of partial response, and at disease progression were tested to monitor changes in miR-31-3p expression overtreatment. miR-31-3p expression, sidedness, and RAS status in pretreatment cell-free DNA were combined in multivariable regression models to assess the predictive value of each variable alone or in combination.

Results:

Patients with low miR-31-3p expression in pretreatment biopsies showed better overall response rate, as well as better progression-free survival and overall survival, compared to those with high miR-31-3p expression. The prognostic effect of miR-31-3p was independent from age, gender, and sidedness. No significant changes in the expression of miR-31-3p were observed when sequential tissue biopsies were tested in long-term or poor responders to anti-EGFR mAbs. miR-31-3p scores were similar when pretreatment biopsies were compared with treatment-naïve archival tissues (often primary colorectal cancer).

Conclusions:

Our study validates the role of miR-31-3p as potential predictive biomarker of selection for anti-EGFR mAbs.

Translational Relevance

RAS status and sidedness represent negative predictive markers of response to anti-EGFR treatment in metastatic colorectal cancer (mCRC) patients. Recently, miR-31-3p has emerged as a potential biomarker for the selection of candidates to first line treatment with a combination of chemotherapy and anti-EGFR treatment. Here we confirm the predictive value of miR-31-3p in a prospective cohort of chemo-refractory mCRC patients treated with single agent cetuximab in a phase II trial. We show that: miR-31-3p can be scored using ISH on pretreatment biopsies; miR-31-3p expression is comparable between primary colorectal cancer and metastases; miR-31-3p expression does not change during cetuximab treatment; and that patients with low miR-31-3p expression had better disease control, progression-free survival, and overall survival compared to patients with high miR-31-3p expression. We suggest that miR-31-3p analysis might be incorporated in the work-up of mCRC along with tumor sidedness and RAS testing, in order to further refine the selection of potential responders to anti-EGFR treatments.

Colorectal cancer is a leading cause of morbidity and mortality worldwide (1, 2). EGFR mAbs are effective in metastatic colorectal cancer (mCRC) and can be used alone or in combination with chemotherapy (3). Mutations in the RAS pathway are negative predictive biomarkers of response to anti-EGFR antibodies in patients with mCRC, thus RAS testing on tissue is routinely performed in clinical practice for patient selection (3).

We and others have recently shown that implementing RAS genotyping in pretreatment circulating cell-free DNA (cfDNA) can identify patients who are unlikely to benefit from anti-EGFR therapies (4, 5). Furthermore, our mathematical modeling indicated that resistance to anti-EGFR antibodies is often polyclonal, suggesting that multiple genetic and nongenetic drivers might contribute to treatment failure (5).

miRNAs are short noncoding RNAs controlling gene expression at posttranscriptional level (6). miRNAs are involved in developmental and physiologic processes (6), and are often dysregulated in pathologic conditions such as cancer and inflammation (7). miRNA dysregulation is frequently observed in colorectal cancer and multiple lines of evidence suggest that miRNAs affect a number of cancer hallmarks (8), and drive colorectal cancer initiation (9), progression (10), and resistance to treatment (11). Given their relative stability in tissues and other bio-fluids (12, 13), miRNAs have been proposed as potential biomarkers for colorectal cancer early detection (14, 15), diagnosis (16), and prognosis (17).

miRNA upregulation and/or SNPs in miRNA target genes have been postulated as potential determinants of resistance and sensitivity to anti-EGFR mAbs in early and metastatic CRC (18–21). miR-31-3p expression levels have been examined by RT-PCR in retrospective analyses of the FIRE-3, PICCOLO, NEW-EPOC, and PETACC8 trials (22–26); in these studies, low miR-31-3p expression was associated with improved outcome and prolonged benefit from anti-EGFR treatment. Real-time PCR-based assays for the analysis of miR-31-3p on formalin-fixed paraffin embedded (FFPE) tissues are at an advanced stage of validation (23, 27).

The PROSPECT-C trial was a phase II trial of single agent anti-EGFR antibodies in chemo-refractory mCRC. Patients underwent repeated tissue biopsies of metastatic deposits before and after treatment as well as at the time of treatment response in case of partial response (PR; ref. 5).

In this study we aimed to: (i) validate the association between miR-31-3p expression and clinical benefit from anti-EGFR treatment in pre-treatment tissue biopsies; (ii) test changes in miR-31-3p expression in serial tissue biopsies during treatment in order to assess whether miR-31-3p might be a potential biomarker of acquired resistance; (iii) test whether combining miR-31-3p with RAS testing in cfDNA might improve patient selection.

Trial design and patient characteristics

The PROSPECT-C trial (ClinicalTrials.gov identifier: NCT02994888) was a prospective, phase II, open-label, single center, nonrandomized study of biomarkers of response and resistance to anti-EGFR therapies in KRAS/NRAS wild-type (wt) chemo-refractory mCRC. Patients who were at least 18 years old and had a WHO performance status (PS) of 0 to 2 were considered eligible for this study, if they fulfilled all the following criteria: (i) chemo-refractory (at least two lines of chemotherapy) metastatic disease; (ii) KRAS/NRAS wt (on archival material according to clinically accredited molecular testing); (iii) measurable disease; and (iv) metastatic sites amenable to biopsy. Patients received cetuximab/panitumumab through the Cancer Drug Fund. Written informed consent was obtained from all patients. The study was carried out in accordance with the Declaration of Helsinki and was approved by national institutional review boards [National Research Ethics Service (NRES): 12/LO/0914]. The objectives of the study were to validate known mechanisms and identify novel drivers of response/resistance to cetuximab. Treatment consisted of cetuximab 500 mg/m2 once every 2 weeks until progression or intolerable side effects. All but one patient received cetuximab and were anti-EGFR naïve at the time of trial entry; the aforementioned patient was switched to panitumumab due to a Common Toxicity Criteria for Adverse Events (CTCAE) 3.0 Grade II allergic reaction after the first dose of cetuximab, and had previously received three cycles of fluorouracil, oxaliplatin, and cetuximab combination with PR as neo-adjuvant chemotherapy for liver resection in the context of the NewEPOC trial 13 months before entering the PROSPECT-C trial.

All participants were required to have mandatory pre-treatment biopsies [baseline (BL), 6 cores], biopsies at 3 months [if PR by Response Evaluation Criteria In Solid Tumors (RECIST) v1.1 criteria (6 cores)] where clinically and technically feasible, and post-treatment at the time of progressive disease (PD) (6 cores from two suitable progressing metastatic sites). Archival material (primary cancer or original diagnostic biopsies) was assessed where available. Plasma for cfDNA analysis was collected every 4 weeks until disease progression.

Analysis of miR-31-3p expression using ISH

ISH assays for miR-31-3p expression in baseline tissue was performed using miRCURY LNA miRNA ISH Optimization Kit for FFPE (Qiagen). Archival tumor material at diagnosis was tested if available (n = 12). ISH on tissue sections was performed following the Exiqon protocol with some modifications. Initially paraffin was removed with xylene incubation for 5 minutes followed by ethanol 100% incubation for another 5 minutes. Tissue sections were then dehydrated in ThermoBrite hybridizer (Leica Biosystems) containing 20 μg/mL of Proteinase K (Roche) for 15 minutes at 37°C. The dehydration reaction was stopped by immersing the slides in PBS, and a prehybridization step was then performed by adding 1× ISH buffer (Exiqon) and incubating the sections for 15 minutes at 56°C. Following the removal of the prehybridization solution, previously denatured (90°C) miRCURY LNA miRNA detection probe (hsa-miR-31-3p; Catalog No. YD006116560; Exiqon) was added to the sections at a 200 nmol/L concentration; sections were incubated with the detection probe overnight at 56°C. The following day tissue sections were sequentially immersed in 5×, 1×, and 0.2× SCC solutions at hybridization temperature for 5 minutes each, and finally transferred in PBS solution at room temperature. Blocking was performed at 30°C in hybridizer followed by incubation with anti-digoxygenin-AP fragments (Sigma-Aldrich) for 1 hour. The sections were then washed three times with PBS-Tween 0.2% for 5 minutes each and incubated for 2 hours with BCIP/NBT Liquid Substrate System (Sigma-Aldrich) for developing the reaction. The reaction was stopped with by immersing the slides in KTBT buffer and counterstained in Nuclear Fast Red (Vector Laboratories). The sections were then dehydrated by ethanol 100% and xylene incubations (for 5 minutes each) and covered with a coverslip.

miR-31-3p expression was graded by two independent pathologists as follows: 0 = no staining; 1+ = weak staining; 2+ = intermediate staining; and 3+ = strong staining. Patients with a 0 or 1+ expression score were deemed as low expressors whereas those with a score of 2+ or 3+ were deemed as high expressors.

Analysis of miR-31-3p expression using real-time PCR

Prior to RNA extraction, samples were reviewed by the pathologist and cancer areas were marked for subsequent macro-dissection. Total RNA from FFPE slides (4 × 4 μm sections) was extracted using the Ambion Recover All Isolation Kit (Thermo Fisher Scientific), according to manufacturer's instructions. RNA quantity and quality were assessed by NanoDrop2000 (Thermo Fisher Scientific). Ten nanograms of total RNA were retrotranscribed using the TaqMan MicroRNA Reverse Transcription Kit (Thermo Fisher Scientific), and RT-PCR was performed using the TaqMan assay for miR-31-3p (assay ID 002113). RNU48 was used as housekeeping gene for normalization, and relative expression was calculated using the 2ΔCt method. miR-31-3p expression was scored as high or low based on the median of the distribution.

Statistical analysis

Progression-free survival (PFS) was calculated from start of treatment with cetuximab to date of progression assessed radiologically, or clinically. Overall survival (OS) was calculated from start of cetuximab to date of death from any cause or last day of follow-up. Differences in PFS and OS between patients with low expression and high miR-31-3p expression pre-treatment were calculated using the Kaplan–Meier method and compared using the log-rank test. Chi-square test was used to assess the effect of miR expression on overall response to cetuximab treatment. Univariate and multivariate analysis using Cox proportional hazards method were performed to assess effects of age, gender, sidedness of tumor in all 42 patients. In the 34 patients for whom baseline ctDNA results were available, a similar approach was used and multivariate analysis performed. A P value of <0.05 was considered significant.

All the authors reviewed and approved the final manuscript. Researcher performing experiments and scoring tissues were blind to clinical outcome information. Analysis was performed by trial statisticians.

The PROSPECT-C trial recruited 45 eligible patients between November 2012 and December 2016 (Fig. 1). Forty-five percent of patients achieved disease control (PR or stable disease by RECIST 1.1. criteria); median PFS and OS were 2.6 months [95% confidential interval (CI), 1.9–4.2] and 8.2 months (95% CI, 4.2–12.0), respectively. These data have been previously reported by our group (5) and are in keeping with available literature for single agent anti-EGFR treatments in chemo-refractory mCRC (28).

Figure 1.

Schematic overview of the PROSPECT-C trial. Chemorefractory, mCRC patients meeting all the inclusion criteria underwent serial tissue and liquid biopsies from metastatic deposits prior to anti-EGFR treatment, after 3 months of treatment in case of PR, and at time of progression (PD).

Figure 1.

Schematic overview of the PROSPECT-C trial. Chemorefractory, mCRC patients meeting all the inclusion criteria underwent serial tissue and liquid biopsies from metastatic deposits prior to anti-EGFR treatment, after 3 months of treatment in case of PR, and at time of progression (PD).

Close modal

To test the association between miR-31-3p expression and benefit from anti-EGFR treatment, we initially scored miR-31-3p by ISH in 91 baseline tissue core biopsies from 45 patients. Forty-two of those patients (88 cores) could be tested further in this study, as, following extensive previous analyses (5, 29), no cancer was left in three cases.

miR-31-3p was marked as low if scored negative or 1+, and as high if scored 2+ or 3+ in cancer (Fig. 2), whereas stromal miR-31-3p staining due to inflammatory or immune infiltrate (Supplementary Fig. S1) was not taken into account. Positive cells in the stroma were represented by plasma cells, macrophages, and endothelial cells, and most of them showed a faint miR-31-3p expression, with only a limited number of cells characterized by a moderate expression. No significant difference in the proportion of positive stromal cells was observed between anti-EGFR responder and resistant tumors.

Figure 2.

miR-31-3p ISH in pretreatment biopsies. Examples of miR-31-3p expression tested by ISH in tissue biopsies obtained from metastatic deposits prior to anti-EGFR treatment in the PROSPECT-C trial. The left panels show examples of cases scored as miR-31-3p “Low” based on a score of 0 (top) or 1+ (bottom). The right panels show examples of cases scored as miR-31-3p “High” based on a score of 2+ (top) or 3+ (bottom). Original magnifications = 20×. In each case, a higher magnification of miR-31-3p expression in representative neoplastic cells is shown as right bottom inlet.

Figure 2.

miR-31-3p ISH in pretreatment biopsies. Examples of miR-31-3p expression tested by ISH in tissue biopsies obtained from metastatic deposits prior to anti-EGFR treatment in the PROSPECT-C trial. The left panels show examples of cases scored as miR-31-3p “Low” based on a score of 0 (top) or 1+ (bottom). The right panels show examples of cases scored as miR-31-3p “High” based on a score of 2+ (top) or 3+ (bottom). Original magnifications = 20×. In each case, a higher magnification of miR-31-3p expression in representative neoplastic cells is shown as right bottom inlet.

Close modal

At least two different slides for each core were tested and concordance in miR-31-3p scoring among different sections from the same core was 100%. In 32 patients, two different cores from the same metastasis were tested, although, in four cases one of the two cores showed only necrosis and/or inflammation and thus comparison with its parental core was not possible. The concordance in miR-31-3p scoring among different cores in the remaining 28 patients was 89% (three cases scored in different categories and were attributed to the high miR-31-3p category as the average score was above 1+). Overall, 24 patients were scored as miR-31-3p low and 18 as miR-31-3p high; patients' demographics based on miR-31-3p expression are presented in Table 1.

Table 1.

Demographics of the PROSPECT-C Trial based on miR-31-3p expression (n = 42)

miR-31-3p lowmiR-31-3p high
Age at registration: median (IQR) 69.6 (62.5–75.9) 67.9 (59.3–73.3) 
Gender 
 Females 8 (33.3%) 8 (44.4%) 
 Males 16 (66.7%) 10 (55.6%) 
RAS pathway aberration in pretreatment cfDNA 
 Absent 9 (50%) 8 (50%) 
 Present 9 (50%) 8 (50%) 
Side 
 Left 19 (79.2%) 12 (66.7%) 
 Right 5 (20.8%) 6 (33.3%) 
Previous treatment lines: median (IQR) 1 (1–2) 2 (1–2) 
miR-31-3p lowmiR-31-3p high
Age at registration: median (IQR) 69.6 (62.5–75.9) 67.9 (59.3–73.3) 
Gender 
 Females 8 (33.3%) 8 (44.4%) 
 Males 16 (66.7%) 10 (55.6%) 
RAS pathway aberration in pretreatment cfDNA 
 Absent 9 (50%) 8 (50%) 
 Present 9 (50%) 8 (50%) 
Side 
 Left 19 (79.2%) 12 (66.7%) 
 Right 5 (20.8%) 6 (33.3%) 
Previous treatment lines: median (IQR) 1 (1–2) 2 (1–2) 

Abbreviation: IQR, interquartile range.

To validate the results obtained by ISH, we performed miR-31-3p expression analysis using RT-PCR on 46 cores from 18 patients for whom material was available for RNA extraction. Two different core biopsies were available in 19 cases and the average expression (based on RT-PCR) between the two cores was used to determine the miR-31-3p scoring; for the remaining 8 cases only one core biopsy was tested. The 46 cores included four primary tumors, 29 pretreatment (baseline), four on-treatment (at the time of PR), and nine posttreatment (progression) biopsies. A statistically significant correlation (chi-squared exact test P: 0.003) with 77% concordance between the two miR-31-3p expression tests was observed (Supplementary Table S1).

Next we tested the association between miR-31-3p score based on ISH and clinical benefit from anti-EGFR mAbs. Low miR-31-3p expression was associated with better overall response rate (ORR) defined as PR or stable disease, with 58.3% (14/24) patients showing response in the miR-31-3p low group versus 22.2% (4/18) in the miR-31-3p high group (Supplementary Table S2; chi-squared exact test P: 0.029). A significant depth and duration of response was observed in patients with low miR-31-3p expression (Fig. 3A and B). Median PFS was 4.21 months (95% CI, 1.91–5.56) and 2.27 months (95% CI, 1.55–2.53) in patients with low and high miR-31-3p respectively [HR for miR-31-3p high: 2.03 (95% CI, 1.06–3.91); P = 0.03] (Fig. 3C). Similarly, median OS was 8.88 months (95% CI, 5.53–18.36) and 4.14 months (95% CI, 2.96–8.68) in patients with low and high miR-31-3p respectively [HR for miR-31-3p high: 2.20 (95% CI, 1.09–4.43); P = 0.03] (Fig. 3D). Multivariable Cox regression analysis including miR-31-3p expression, age at diagnosis, gender, and sidedness (30) in the trial cohort (n = 42) confirmed that miR-31-3p was an independent predictor of PFS (Supplementary Table S3) and OS (Supplementary Table S4).

Figure 3.

miR-31-3p and clinical benefit from anti-EGFR therapy in the PROSPECT-C trial. Waterfall (A) and spider (B) plots show depth (based on RECIST 1.1 criteria) and duration of response based on miR-31-3p expression. Kaplan–Meier curves for progression-free (C) and overall (D) survival according to miR-31-3p expression.

Figure 3.

miR-31-3p and clinical benefit from anti-EGFR therapy in the PROSPECT-C trial. Waterfall (A) and spider (B) plots show depth (based on RECIST 1.1 criteria) and duration of response based on miR-31-3p expression. Kaplan–Meier curves for progression-free (C) and overall (D) survival according to miR-31-3p expression.

Close modal

Changes in miR-31-3p expression during single-agent cetuximab treatment have never been investigated before. Here we took advantage of repeated serial tissue sampling in our trial and we tested whether miR-31-3p scoring is altered during or after EGFR inhibition. Analysis of nine patients with long-term response (PFS ≥6 months) and patients with primary progression (PFS ≤ 3 months) revealed no changes in miR-31-3p scoring in either of the two groups (Supplementary Table S5; Fig. 4A). Indeed, analysis of liver, nodal, abdominal wall, and pelvic metastases showed consistent miR-31-3p expression even when different metastases were tested (Fig. 4A and B).

Figure 4.

Analysis of miR-31-3p expression in sequential tissue biopsies in the PROSPECT-C trial. A, miR-31-3p expression was tested by ISH in pre- and posttreatment tissue biopsies as well as after 3 months of treatment in case of PR. miR-31-3p scoring did not change over treatment in liver (1024 and 1041) or in nodal (1026) cancer deposits (yellow circles, targeted metastasis). B, Axial enhanced CT images performed at baseline and 20 months later at end of treatment due to RECIST 1.1 progression in nontarget disease in patient 1005. The images show maintained benefit in the target pelvic lesion (yellow arrow) and a mixed response in the nontarget abdominal wall disease with stable appearances of the right para-median lesion (white asterisk) and progression of the left para-median lesion (arrowhead). At baseline the biopsy was obtained from the target pelvic lesion (yellow arrow), at progression the biopsy was of the left para-median lesion (the biopsy tract can be seen in the subcutaneous tissue—white oval). No changes in miR-31-3p staining were observed in sequential biopsies from the different regions. Original magnifications = 20×.

Figure 4.

Analysis of miR-31-3p expression in sequential tissue biopsies in the PROSPECT-C trial. A, miR-31-3p expression was tested by ISH in pre- and posttreatment tissue biopsies as well as after 3 months of treatment in case of PR. miR-31-3p scoring did not change over treatment in liver (1024 and 1041) or in nodal (1026) cancer deposits (yellow circles, targeted metastasis). B, Axial enhanced CT images performed at baseline and 20 months later at end of treatment due to RECIST 1.1 progression in nontarget disease in patient 1005. The images show maintained benefit in the target pelvic lesion (yellow arrow) and a mixed response in the nontarget abdominal wall disease with stable appearances of the right para-median lesion (white asterisk) and progression of the left para-median lesion (arrowhead). At baseline the biopsy was obtained from the target pelvic lesion (yellow arrow), at progression the biopsy was of the left para-median lesion (the biopsy tract can be seen in the subcutaneous tissue—white oval). No changes in miR-31-3p staining were observed in sequential biopsies from the different regions. Original magnifications = 20×.

Close modal

Comparison of miR-31-3p score between archival, treatment naïve tissue (primary colorectal cancer in most of the cases) and pre-cetuximab tissue biopsies (Supplementary Table S6) was concordant in 11 of 12 cases (Fisher exact test P = 0.01) (Supplementary Table S7).

We and others have recently demonstrated the predictive value of RAS testing in pretreatment cfDNA as a valuable and more specific alternative to tissue analysis in the selection of patients eligible for anti-EGFR treatments (4, 5). When we included miR-31-3p in a multivariable Cox regression analysis including age at diagnosis, gender, sidedness, and RAS genotyping in pretreatment cfDNA in patients for whom all the information were available (n = 34), miR-31-3p showed no independent value in predicting PFS (Supplementary Table S8) or OS (Supplementary Table S9). In keeping with these data, when we generated a statistical model combining miR-31-3p status in tissues and RAS genotyping in pretreatment cfDNA (presence/absence of mutations) (Supplementary Fig. S2A–S2C; Supplementary Tables S10 and S11), the interaction tests for ORR, PFS, and OS were nonsignificant (P = 0.213, 0.178, and 0.067, respectively). Among patients who tested as RAS wt in cfDNA from baseline bloods, ORR was 78%, median PFS was 5.10 months (95% CI, 1.91–16.88) and median OS was 15.23 months (95% CI, 1.91–34.08) in patients with low miR-31-3p expression (n = 9). On the contrary, ORR was 25%, median PFS was 2.27 months (95% CI, 1.91–16.88) and median OS was 4.67 months (95% CI, 1.51–12.04) in patients with high miR-31-3p expression (n = 8).

miR-31-3p expression has been tested in a number of retrospective series and retrospective analyses of prospective trials (18, 22–24, 31). Low miR-31-3p expression has been associated with sustained PFS and OS as well as improved ORR in response to EGFR inhibitors. Although these findings have been validated in several studies, the interpretation of these data remains challenging due to the fact that in most of these series anti-EGFR mAbs (cetuximab or panitumumab) were used in combination with different chemotherapy backbones and in different lines of treatment. In the PROSPECT-C trial (5), cetuximab was used as a single agent in a prospective and homogeneous cohort of chemo-refractory mCRC patients; furthermore, miR-31-3p was tested in ad hoc pre- and posttreatment tissues biopsies. Thus, despite a relatively small sample size, the trial provided an excellent opportunity to validate the predictive role of miR-31-3p in a prospective cohort and allowed to test dynamic changes in miR-31-3p expression overtreatment. The results presented here are largely consistent with available literature (22–24, 27) and suggest that low miR-31-3p might be an indicator of response and better prognosis in patients treated with anti-EGFR mAbs.

Even though our data align well with available literature, several questions remain open. First, the biology underpinning a potential role for miR-31-3p in driving resistance to anti-EGFR agents is not clear. Preclinical in vitro and in vivo data in colon and lung cancer respectively suggest that miR-31-3p targets a number of negative regulators of the RAS-MAPK cascade (32, 33). However, despite the link between miR-31-3p overexpression and RAS signaling pathway activation appears solid, no experimental evidence has, as yet, confirmed whether these mechanisms are responsible for resistance to cetuximab.

A second question relates to the source of material and the technology to be used for miR-31-3p testing. Mir-31-3p is overexpressed in early stages of sporadic and inflammation-related colorectal cancer carcinogenesis but expression does not appear to change in more advanced or metastatic stages of disease (34–36). In keeping with these data, no significant changes in miR-31-3p scoring were observed in our trial when comparing primary cancers and metastatic sites. Similarly, in our series, no changes in miR-31-3p scoring were observed in sequential tissues biopsies collected before, during, and after cetuximab treatment. On the contrary, in the NEW-EPOC trial (22), a nonsignificant correlation for miR-31-3p expression was observed between paired primary colorectal cancer specimens and liver metastases in patients receiving preoperative cetuximab, whereas a positive and significant correlation was observed in patients treated with chemotherapy alone, suggesting that cetuximab treatment might induce changes in miR-31-3p expression. One of the potential explanations for the discrepancy between the NEW-EPOC (22) and the PROSPECT-C trial (5) is that, in the former trial, miR-31-3p levels were tested by ISH whereas in the latter, the analysis was performed by RT-PCR. Cetuximab is known to trigger intratumor inflammatory infiltration in liver metastases, with an enrichment of CD3, CD8, and CD56+ cells (37). Given miR-31-3p is also involved in immune and inflammatory cells homeostasis (38–41), its overexpression might sometimes be due to intratumor infiltration from lymphocites or inflammatory cells. Under these circumastances, despite tissue microdissection, contamination by inflammatory cells might potentially lead to a bias in miR-31-3p scoring when using high sensitivity RT-PCR-based assays. In line with this hypothesis, our ISH did detect areas of miR-31-3p overexpression in the stromal compartment of tumors otherwise scored as miR-31-3p low. Furthermore, even though the comparison between ISH-based and RT-PCR-based miR-31-3p scoring in our cohort showed a good concordance, several cases were classified in different miR-31-3p expression categories by the two assays, thus highlighting some hurdles in selecting the best approach for evaluating miR-31-3p expression as a biomarker for anti-EGFR mAbs. Given RT-PCR-based assays have been recently validated for miR-31-3p clinical testing (23, 27), caution in the analysis and interpretation of data should be exerted in cases with intense inflammatory and immune infiltrate as these might affect miR-31-3p classification.

Selection of mCRC patients' candidate to anti-EGFR treatment relies on primary tumor location (sidedness; ref. 30) and RAS testing (3). As we and others have suggested (4, 5), moving RAS testing to plasma cfDNA might represent a more sensitive and cost/effective option than tissue analysis. In our study, we combined RAS genotyping in cfDNA with miR-31-3p expression in order to test whether this would result in a more accurate prediction of response to cetuximab. The test for interaction between the two categorical variables was not significant possibly due to the very small sample size; however, in patients with no cfDNA RAS abnormalities, ORR, PFS, and OS appeared better for patients with miR-31-3p low tumors. Although larger studies will need to confirm these findings, a key question remains open: do we need another test to select mCRC patients for cetuximab treatment, or are we at risk of ultra-selecting patients? Our data, in line with the analyses of FIRE-3 (26) trial, suggest that miR-31-3p expression may be an indicator of depth of response to anti-EGFR inhibition; this, in our opinion, might represent the ideal scenario where a more accurate identification of patients likely to achieve resectability and/or symptom control may justify a more thorough selection of patients (Fig. 5).

Figure 5.

Proposed workflow for the analysis of miR-31-3p expression in mCRC patients. Mir-31-3p might be recommended for left-sided, RAS wt patients eligible for resection and/or metastasectomy or for disease/symptoms control.

Figure 5.

Proposed workflow for the analysis of miR-31-3p expression in mCRC patients. Mir-31-3p might be recommended for left-sided, RAS wt patients eligible for resection and/or metastasectomy or for disease/symptoms control.

Close modal

In conclusion, our results confirm the potential predictive role of miR-31-3p for the selection of patients undergoing anti-EGFR treatment. Further studies are needed to test if miR-31-3p might be combined with RAS testing in cfDNA to further identify best responders in specific clinical niches.

D. Cunningham reports receiving commercial research support from Amgen, Sanofi, Merrimack, AstraZeneca, Celgene, MedImmune, Bayer, Emerge, Clovis, Eli Lilly, and Janssen. K.H. Khan is a consultant/advisory board member for Bayer Oncology Group. S. Rao is a consultant/advisory board member for Roche and Celgene. N. Starling reports receiving speakers bureau honoraria from AstraZeneca, Eli Lilly, and Merck, and reports receiving commercial research grants from AstraZeneca, Bristol-Myers Squibb, and Merck. C. Braconi reports receiving speakers bureau honoraria from Merck Serono, Bayer, and Eli Lilly, and did a consultation with Lumien. I. Chau is a consultant/advisory board member for Eli-Lilly, Bristol-Myers Squibb, MSD, Bayer, Roche, Merck Serono, Five Prime Therapeutics, AstraZeneca, and Oncologie International, and reports receiving commercial research grants from Eli-Lilly, Janssen-Cilag, and Sanofi Oncology. N. Valeri reports receiving speakers bureau honoraria from Bayer, Pfizer, Eli Lilly, and Merck-Serono. No potential conflicts of interest were disclosed by the other authors.

Conception and design: G. Anandappa, D. Cunningham, K.H. Khan, C. Braconi, C. Peckitt, N. Valeri

Development of methodology: G. Anandappa, A. Lampis, D. Cunningham, K.H. Khan, N. Starling, M. Darvish-Damavandi, M. Fassan, N. Valeri

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): G. Anandappa, A. Lampis, D. Cunningham, K.H. Khan, S. Rao, D. Watkins, H. Lote, J. Thomas, N. Khan, N. Fotiadis, R. Begum, I. Rana, J.C. Hahne, I. Chau, N. Valeri

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): G. Anandappa, D. Cunningham, K.H. Khan, K. Kouvelakis, G. Vlachogiannis, N. Tunariu, N. Starling, R. Kalaitzaki, J.C. Hahne, I. Chau, M. Fassan, N. Valeri

Writing, review, and/or revision of the manuscript: G. Anandappa, A. Lampis, D. Cunningham, K.H. Khan, K. Kouvelakis, N. Tunariu, S. Rao, D. Watkins, N. Starling, C. Braconi, M. Darvish-Damavandi, C. Peckitt, R. Kalaitzaki, J.C. Hahne, I. Chau, M. Fassan, N. Valeri

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): K.H. Khan, S. Hedayat, D. Watkins, N. Khan, R. Begum, I. Rana, A. Bryant, N. Valeri

Study supervision: D. Cunningham, K.H. Khan, D. Watkins, M. Rugge, N. Valeri

This work was supported by Cancer Research UK (grant number CEA A18052), the National Institute for Health Research (NIHR) Biomedical Research Centre (BRC) at The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research (grant numbers A62, A100, A101, A159), and the European Union FP7 (grant number CIG 334261) to N. Valeri. The authors acknowledge support from the National Institute for Health Research Biomedical Research Centre at The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2018
.
CA Cancer J Clin
2018
;
68
:
7
30
.
2.
Ferlay
J
,
Colombet
M
,
Soerjomataram
I
,
Dyba
T
,
Randi
G
,
Bettio
M
, et al
Cancer incidence and mortality patterns in Europe: estimates for 40 countries and 25 major cancers in 2018
.
Eur J Cancer
2018
;
103
:
356
87
.
3.
Van Cutsem
E
,
Cervantes
A
,
Adam
R
,
Sobrero
A
,
Van Krieken
JH
,
Aderka
D
, et al
ESMO consensus guidelines for the management of patients with metastatic colorectal cancer
.
Ann Oncol
2016
;
27
:
1386
422
.
4.
Thierry
AR
,
El Messaoudi
S
,
Mollevi
C
,
Raoul
JL
,
Guimbaud
R
,
Pezet
D
, et al
Clinical utility of circulating DNA analysis for rapid detection of actionable mutations to select metastatic colorectal patients for anti-EGFR treatment
.
Ann Oncol
2017
;
28
:
2149
59
.
5.
Khan
K
,
Cunningham
D
,
Werner
B
,
Vlachogiannis
G
,
Spiteri
I
,
Heide
T
, et al
Longitudinal liquid biopsy and mathematical modelling of clonal evolution forecast waiting time to treatment failure in the PROSPECT-C phase II colorectal cancer clinical trial
.
Cancer Discov
2018
;
8
:
1270
85
.
6.
Bartel
DP
. 
Metazoan microRNAs
.
Cell
2018
;
173
:
20
51
.
7.
Croce
CM
. 
Causes and consequences of microRNA dysregulation in cancer
.
Nat Rev Genet
2009
;
10
:
704
14
.
8.
Berindan-Neagoe
I
,
Monroig Pdel
C
,
Pasculli
B
,
Calin
GA
. 
MicroRNAome genome: a treasure for cancer diagnosis and therapy
.
CA Cancer J Clin
2014
;
64
:
311
36
.
9.
Valeri
N
,
Braconi
C
,
Gasparini
P
,
Murgia
C
,
Lampis
A
,
Paulus-Hock
V
, et al
MicroRNA-135b promotes cancer progression by acting as a downstream effector of oncogenic pathways in colon cancer
.
Cancer Cell
2014
;
25
:
469
83
.
10.
Valeri
N
,
Gasparini
P
,
Fabbri
M
,
Braconi
C
,
Veronese
A
,
Lovat
F
, et al
Modulation of mismatch repair and genomic stability by miR-155
.
Proc Natl Acad Sci U S A
2010
;
107
:
6982
7
.
11.
Valeri
N
,
Gasparini
P
,
Braconi
C
,
Paone
A
,
Lovat
F
,
Fabbri
M
, et al
MicroRNA-21 induces resistance to 5-fluorouracil by down-regulating human DNA MutS homolog 2 (hMSH2)
.
Proc Natl Acad Sci U S A
2010
;
107
:
21098
103
.
12.
Anfossi
S
,
Babayan
A
,
Pantel
K
,
Calin
GA
. 
Clinical utility of circulating non-coding RNAs - an update
.
Nat Rev Clin Oncol
2018
;
15
:
541
63
.
13.
Shigeyasu
K
,
Toden
S
,
Zumwalt
TJ
,
Okugawa
Y
,
Goel
A
. 
Emerging role of microRNAs as liquid biopsy biomarkers in gastrointestinal cancers
.
Clin Cancer Res
2017
;
23
:
2391
9
.
14.
Toiyama
Y
,
Okugawa
Y
,
Tanaka
K
,
Araki
T
,
Uchida
K
,
Hishida
A
, et al
A panel of methylated MicroRNA biomarkers for identifying high-risk patients with ulcerative colitis-associated colorectal cancer
.
Gastroenterology
2017
;
153
:
1634
46
.
e8
.
15.
Yamada
A
,
Horimatsu
T
,
Okugawa
Y
,
Nishida
N
,
Honjo
H
,
Ida
H
, et al
Serum miR-21, miR-29a, and miR-125b are promising biomarkers for the early detection of colorectal neoplasia
.
Clin Cancer Res
2015
;
21
:
4234
42
.
16.
Hur
K
,
Toiyama
Y
,
Okugawa
Y
,
Ide
S
,
Imaoka
H
,
Boland
CR
, et al
Circulating microRNA-203 predicts prognosis and metastasis in human colorectal cancer
.
Gut
2017
;
66
:
654
65
.
17.
Ozawa
T
,
Kandimalla
R
,
Gao
F
,
Nozawa
H
,
Hata
K
,
Nagata
H
, et al
A MicroRNA signature associated with metastasis of T1 colorectal cancers to lymph nodes
.
Gastroenterology
2018
;
154
:
844
8
e7
.
18.
Mlcochova
J
,
Faltejskova-Vychytilova
P
,
Ferracin
M
,
Zagatti
B
,
Radova
L
,
Svoboda
M
, et al
MicroRNA expression profiling identifies miR-31-5p/3p as associated with time to progression in wild-type RAS metastatic colorectal cancer treated with cetuximab
.
Oncotarget
2015
;
6
:
38695
704
.
19.
Sclafani
F
,
Chau
I
,
Cunningham
D
,
Peckitt
C
,
Lampis
A
,
Hahne
JC
, et al
Prognostic role of the LCS6 KRAS variant in locally advanced rectal cancer: results of the EXPERT-C trial
.
Ann Oncol
2015
;
26
:
1936
41
.
20.
Sha
D
,
Lee
AM
,
Shi
Q
,
Alberts
SR
,
Sargent
DJ
,
Sinicrope
FA
, et al
Association study of the let-7 miRNA-complementary site variant in the 3′ untranslated region of the KRAS gene in stage III colon cancer (NCCTG N0147 Clinical Trial)
.
Clin Cancer Res
2014
;
20
:
3319
27
.
21.
Saridaki
Z
,
Weidhaas
JB
,
Lenz
HJ
,
Laurent-Puig
P
,
Jacobs
B
,
De Schutter
J
, et al
A let-7 microRNA-binding site polymorphism in KRAS predicts improved outcome in patients with metastatic colorectal cancer treated with salvage cetuximab/panitumumab monotherapy
.
Clin Cancer Res
2014
;
20
:
4499
510
.
22.
Pugh
S
,
Thiebaut
R
,
Bridgewater
J
,
Grisoni
ML
,
Moutasim
K
,
Rousseau
F
, et al
Association between miR-31-3p expression and cetuximab efficacy in patients with KRAS wild-type metastatic colorectal cancer: a post-hoc analysis of the New EPOC trial
.
Oncotarget
2017
;
8
:
93856
66
.
23.
Laurent-Puig
P
,
Grisoni
ML
,
Heinemann
V
,
Liebaert
F
,
Neureiter
D
,
Jung
A
, et al
Validation of miR-31-3p expression to predict cetuximab efficacy when used as first-line treatment in RAS wild-type metastatic colorectal cancer
.
Clin Cancer Res
2019
;
25
:
134
41
.
24.
Laurent-Puig
P
,
Paget-Bailly
S
,
Vernerey
D
,
Vazart
C
,
Decaulne
V
,
Fontaine
K
, et al
Evaluation of miR 31 3p as a biomarker of prognosis and panitumumab benefit in RAS-wt advanced colorectal cancer (aCRC): analysis of patients (pts) from the PICCOLO trial
.
J Clin Oncol
2015
;
33
:
3547
.
25.
Gaston Mathe
Y
,
Martin-Lannerée
S
,
Vazart
C
,
Fontaine
K
,
Mulot
C
,
Caumont
A
, et al
miR-31 as a prognostic and predictive marker of disease-free survival (DFS) in resected stage III colon cancer: a retrospective analysis of the PETACC-8 trial
.
Ann Oncol
2018
;
29
(
suppl_8
):
viii150
viii204
.
26.
Folprecht
G
,
Beer
P
,
Salazar
R
,
Roth
A
,
Aust
D
,
Salgado
R
, et al
Frequency of potentially actionable genetic alterations in EORTC SPECTAcolor
.
Ann Oncol
2016
;
27
:
4580
.
27.
Ramon
L
,
David
C
,
Fontaine
K
,
Lallet
E
,
Marcaillou
C
,
Martin-Lanneree
S
, et al
Technical validation of a reverse-transcription quantitative polymerase chain reaction in vitro diagnostic test for the determination of MiR-31-3p expression levels in formalin-fixed paraffin-embedded metastatic colorectal cancer tumor specimens
.
Biomark Insights
2018
;
13
:
1177271918763357
.
28.
Lee
MS
,
Kopetz
S
. 
Current and future approaches to target the epidermal growth factor receptor and its downstream signaling in metastatic colorectal cancer
.
Clin Colorectal Cancer
2015
;
14
:
203
18
.
29.
Vlachogiannis
G
,
Hedayat
S
,
Vatsiou
A
,
Jamin
Y
,
Fernandez-Mateos
J
,
Khan
K
, et al
Patient-derived organoids model treatment response of metastatic gastrointestinal cancers
.
Science
2018
;
359
:
920
6
.
30.
Boeckx
N
,
Koukakis
R
,
Op de Beeck
K
,
Rolfo
C
,
Van Camp
G
,
Siena
S
, et al
Primary tumor sidedness has an impact on prognosis and treatment outcome in metastatic colorectal cancer: results from two randomized first-line panitumumab studies
.
Ann Oncol
2017
;
28
:
1862
8
.
31.
Manceau
G
,
Imbeaud
S
,
Thiebaut
R
,
Liebaert
F
,
Fontaine
K
,
Rousseau
F
, et al
Hsa-miR-31-3p expression is linked to progression-free survival in patients with KRAS wild-type metastatic colorectal cancer treated with anti-EGFR therapy
.
Clin Cancer Res
2014
;
20
:
3338
47
.
32.
Sun
D
,
Yu
F
,
Ma
Y
,
Zhao
R
,
Chen
X
,
Zhu
J
, et al
MicroRNA-31 activates the RAS pathway and functions as an oncogenic MicroRNA in human colorectal cancer by repressing RAS p21 GTPase activating protein 1 (RASA1)
.
J Biol Chem
2013
;
288
:
9508
18
.
33.
Edmonds
MD
,
Boyd
KL
,
Moyo
T
,
Mitra
R
,
Duszynski
R
,
Arrate
MP
, et al
MicroRNA-31 initiates lung tumorigenesis and promotes mutant KRAS-driven lung cancer
.
J Clin Invest
2016
;
126
:
349
64
.
34.
Ito
M
,
Mitsuhashi
K
,
Igarashi
H
,
Nosho
K
,
Naito
T
,
Yoshii
S
, et al
MicroRNA-31 expression in relation to BRAF mutation, CpG island methylation and colorectal continuum in serrated lesions
.
Int J Cancer
2014
;
135
:
2507
15
.
35.
Nosho
K
,
Igarashi
H
,
Nojima
M
,
Ito
M
,
Maruyama
R
,
Yoshii
S
, et al
Association of microRNA-31 with BRAF mutation, colorectal cancer survival and serrated pathway
.
Carcinogenesis
2014
;
35
:
776
83
.
36.
Drusco
A
,
Nuovo
GJ
,
Zanesi
N
,
Di Leva
G
,
Pichiorri
F
,
Volinia
S
, et al
MicroRNA profiles discriminate among colon cancer metastasis
.
PLoS One
2014
;
9
:
e96670
.
37.
Inoue
Y
,
Hazama
S
,
Suzuki
N
,
Tokumitsu
Y
,
Kanekiyo
S
,
Tomochika
S
, et al
Cetuximab strongly enhances immune cell infiltration into liver metastatic sites in colorectal cancer
.
Cancer Sci
2017
;
108
:
455
60
.
38.
Zhang
L
,
Ke
F
,
Liu
Z
,
Bai
J
,
Liu
J
,
Yan
S
, et al
MicroRNA-31 negatively regulates peripherally derived regulatory T-cell generation by repressing retinoic acid-inducible protein 3
.
Nat Commun
2015
;
6
:
7639
51
.
39.
Whiteoak
SR
,
Claridge
A
,
Balendran
CA
,
Harris
RJ
,
Gwiggner
M
,
Bondanese
VP
, et al
MicroRNA-31 targets thymic stromal lymphopoietin in mucosal infiltrated CD4+ T cells: a role in achieving mucosal healing in ulcerative colitis?
Inflamm Bowel Dis
2018
;
24
:
2377
85
.
40.
Moffett
HF
,
Cartwright
ANR
,
Kim
HJ
,
Godec
J
,
Pyrdol
J
,
Aijo
T
, et al
The microRNA miR-31 inhibits CD8(+) T cell function in chronic viral infection
.
Nat Immunol
2017
;
18
:
791
9
.
41.
Fang
K
,
Law
IKM
,
Padua
D
,
Sideri
A
,
Huang
V
,
Kevil
CG
, et al
MicroRNA-31-3p is involved in substance P (SP)-associated inflammation in human colonic epithelial cells and experimental colitis
.
Am J Pathol
2018
;
188
:
586
99
.