Skip to main content

Advertisement

Log in

Clinical relevance of H-RAS, K-RAS, and N-RAS mRNA expression in primary breast cancer patients

  • Clinical trial
  • Published:
Breast Cancer Research and Treatment Aims and scope Submit manuscript

Abstract

Purpose

The RAS family comprises three proto-oncogenes (H-RAS, K-RAS, and N-RAS) and is among the most widely studied of oncogenes. The present study aimed at investigating the clinical relevance of mRNA levels of the three isoforms in a large group of breast cancer patients with a long-term follow-up.

Methods

198 previously untreated patients were enrolled in the study. mRNA levels of K-RAS, H-RAS, and N-RAS were measured using microarray (Affymetrix HG-U133A).

Results

Elevated H-RAS levels were found significantly more frequently in patients with larger (p = 0.021) and ER-positive tumors (p = 0.048), while elevated K-RAS levels were associated with nodal positivity (p = 0.001) and HER2-positivity (p = 0.010). Patients with high N-RAS mRNA levels were more likely to be diagnosed with triple-negativity (p < 0.001) and higher grading (p = 0.001). Patients with high K-RAS levels were more likely to show an elevated H-RAS (p = 0.003). After a median follow-up of 183 months, patients with high N-RAS expression had significantly reduced overall survival (OS) compared with patients with low N-RAS (mean: 146.9 vs. 211.0 months; median 169.3 vs. not reached; p = 0.009). In patients with non-metastatic disease at the time of tissue sampling, mean disease-free survival (DFS) was 150.1 months for patients with high N-RAS versus 227.7 months with low N-RAS; median DFS was not reached (p = 0.004). The expression of H-RAS and K-RAS was not associated with DFS/OS. In the multivariable analysis, distant metastasis, HER2 positivity, and elevated N-RAS mRNA levels independently predicted reduced OS, while nodal status, HER2 status, and N-RAS predicted reduced DFS.

Conclusions

Elevated N-RAS mRNA levels predict impaired clinical outcome; hypothetically, further exploration of the RAS signaling pathway might enable identifying potential targeted treatment strategies. The association between high N-RAS levels and the most aggressive among breast cancer subtypes, the triple-negative phenotype, for which targeted approaches are still lacking, underlines the need to further investigate the RAS family.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Data availability

The datasets generated during the current study are available from the corresponding author on reasonable request.

References

  1. Downward J (2003) Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer 3:11–22. https://doi.org/10.1038/nrc969

    Article  CAS  PubMed  Google Scholar 

  2. Shields JM, Pruitt K, McFall A, Shaub A, Der CJ (2000) Understanding Ras: ‘it ain’t over ‘til it’s over’. Trends Cell Biol 10:147–154

    Article  CAS  PubMed  Google Scholar 

  3. Siewertsz van Reesema LL, Lee MP, Zheleva V, Winston JS, O’Connor CF, Perry RR, Hoefer RA, Tang AH (2016) RAS pathway biomarkers for breast cancer prognosis. Clin Lab Int 40:18–23

    PubMed  PubMed Central  Google Scholar 

  4. Gysin S, Salt M, Young A, McCormick F (2011) Therapeutic strategies for targeting ras proteins. Genes Cancer 2:359–372. https://doi.org/10.1177/1947601911412376

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Castellano E, Santos E (2011) Functional specificity of ras isoforms: so similar but so different. Genes cancer 2:216–231. https://doi.org/10.1177/1947601911408081

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Walsh AB, Bar-Sagi D (2001) Differential activation of the Rac pathway by Ha-Ras and K-Ras. J Biol Chem 276:15609–15615. https://doi.org/10.1074/jbc.M010573200

    Article  CAS  PubMed  Google Scholar 

  7. Voice JK, Klemke RL, Le A, Jackson JH (1999) Four human ras homologs differ in their abilities to activate Raf-1, induce transformation, and stimulate cell motility. J Biol Chem 274:17164–17170

    Article  CAS  PubMed  Google Scholar 

  8. Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, Cerami E, Sander C, Schultz N (2013) Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6:pl1. https://doi.org/10.1126/scisignal.2004088

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Spandidos DA, Agnantis NJ (1984) Human malignant tumours of the breast, as compared to their respective normal tissue, have elevated expression of the Harvey ras oncogene. Anticancer Res 4:269–272

    CAS  PubMed  Google Scholar 

  10. Agnantis NJ, Parissi P, Anagnostakis D, Spandidos DA (1986) Comparative study of Harvey-ras oncogene expression with conventional clinicopathologic parameters of breast cancer. Oncology 43:36–39

    Article  CAS  PubMed  Google Scholar 

  11. Agnantis NJ, Apostolikas NA, Zolotas VG, Spandidos DA (1994) Immunohistochemical detection of ras p21 oncoprotein in breast cancer imprints. Acta Cytol 38:335–340

    CAS  PubMed  Google Scholar 

  12. Agnantis NJ, Petraki C, Markoulatos P, Spandidos DA (1986) Immunohistochemical study of the ras oncogene expression in human breast lesions. Anticancer Res 6:1157–1160

    CAS  PubMed  Google Scholar 

  13. Dati C, Muraca R, Tazartes O, Antoniotti S, Perroteau I, Giai M, Cortese P, Sismondi P, Saglio G, De Bortoli M (1991) c-erbB-2 and ras expression levels in breast cancer are correlated and show a co-operative association with unfavorable clinical outcome. Int J Cancer 47:833–838

    Article  CAS  PubMed  Google Scholar 

  14. Banys-Paluchowski M, Fehm T, Janni W, Aktas B, Fasching PA, Kasimir-Bauer S, Milde-Langosch K, Pantel K, Rack B, Riethdorf S, Solomayer EF, Witzel I, Müller V (2018) Elevated serum RAS p21 is an independent prognostic factor in metastatic breast cancer. BMC Cancer. In press

  15. McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM (2005) REporting recommendations for tumour MARKer prognostic studies (REMARK). Br J Cancer 93:387–391. https://doi.org/10.1038/sj.bjc.6602678

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Milde-Langosch K, Karn T, Muller V, Witzel I, Rody A, Schmidt M, Wirtz RM (2013) Validity of the proliferation markers Ki67, TOP2A, and RacGAP1 in molecular subgroups of breast cancer. Breast Cancer Res Treat 137:57–67. https://doi.org/10.1007/s10549-012-2296-x

    Article  CAS  PubMed  Google Scholar 

  17. Milde-Langosch K, Karn T, Schmidt M, Zu Eulenburg C, Oliveira-Ferrer L, Wirtz RM, Schumacher U, Witzel I, Schutze D, Muller V (2014) Prognostic relevance of glycosylation-associated genes in breast cancer. Breast Cancer Res Treat 145:295–305. https://doi.org/10.1007/s10549-014-2949-z

    Article  CAS  PubMed  Google Scholar 

  18. Ihnen M, Muller V, Wirtz RM, Schroder C, Krenkel S, Witzel I, Lisboa BW, Janicke F, Milde-Langosch K (2008) Predictive impact of activated leukocyte cell adhesion molecule (ALCAM/CD166) in breast cancer. Breast Cancer Res Treat 112:419–427. https://doi.org/10.1007/s10549-007-9879-y

    Article  CAS  PubMed  Google Scholar 

  19. Zheng ZY, Tian L, Bu W, Fan C, Gao X, Wang H, Liao YH, Li Y, Lewis MT, Edwards D, Zwaka TP, Hilsenbeck SG, Medina D, Perou CM, Creighton CJ, Zhang XH, Chang EC (2015) Wild-type N-Ras, overexpressed in basal-like breast cancer, promotes tumor formation by inducing IL-8 secretion via JAK2 activation. Cell Rep 12:511–524. https://doi.org/10.1016/j.celrep.2015.06.044

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, Speed D, Lynch AG, Samarajiwa S, Yuan Y, Graf S, Ha G, Haffari G, Bashashati A, Russell R, McKinney S, Group M, Langerod A, Green A, Provenzano E, Wishart G, Pinder S, Watson P, Markowetz F, Murphy L, Ellis I, Purushotham A, Borresen-Dale AL, Brenton JD, Tavare S et al (2012) The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature 486:346–352. https://doi.org/10.1038/nature10983

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Olsen SN, Wronski A, Castano Z, Dake B, Malone C, De Raedt T, Enos M, DeRose YS, Zhou W, Guerra S, Loda M, Welm A, Partridge AH, McAllister SS, Kuperwasser C, Cichowski K (2017) Loss of RasGAP tumor suppressors underlies the aggressive nature of luminal B breast cancers. Cancer Discov 7:202–217. https://doi.org/10.1158/2159-8290.CD-16-0520

    Article  CAS  PubMed  Google Scholar 

  22. Hoadley KA, Weigman VJ, Fan C, Sawyer LR, He X, Troester MA, Sartor CI, Rieger-House T, Bernard PS, Carey LA, Perou CM (2007) EGFR associated expression profiles vary with breast tumor subtype. BMC Genom 8:258. https://doi.org/10.1186/1471-2164-8-258

    Article  CAS  Google Scholar 

  23. Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Pergamenschikov A, Williams C, Zhu SX, Lonning PE, Borresen-Dale AL, Brown PO, Botstein D (2000) Molecular portraits of human breast tumours. Nature 406:747–752. https://doi.org/10.1038/35021093

    Article  CAS  PubMed  Google Scholar 

  24. Zhang T, Li Q, Chen S, Luo Y, Fan Y, Xu B (2017) Phase I study of QLNC120, a novel EGFR and HER2 kinase inhibitor, in pre-treated patients with HER2-overexpressing advanced breast cancer. Oncotarget 8:36750–36760. https://doi.org/10.18632/oncotarget.13581

    Article  PubMed  Google Scholar 

  25. Ye Q, Qi F, Bian L, Zhang SH, Wang T, Jiang ZF (2017) Circulating-free DNA mutation associated with response of targeted therapy in human epidermal growth factor receptor 2-positive metastatic breast cancer. Chin Med J 130:522–529. https://doi.org/10.4103/0366-6999.200542

    Article  PubMed  PubMed Central  Google Scholar 

  26. Hah JH, Zhao M, Pickering CR, Frederick MJ, Andrews GA, Jasser SA, Fooshee DR, Milas ZL, Galer C, Sano D, William WN Jr, Kim E, Heymach J, Byers LA, Papadimitrakopoulou V, Myers JN (2014) HRAS mutations and resistance to the epidermal growth factor receptor tyrosine kinase inhibitor erlotinib in head and neck squamous cell carcinoma cells. Head Neck 36:1547–1554. https://doi.org/10.1002/hed.23499

    Article  PubMed  PubMed Central  Google Scholar 

  27. Kim RK, Suh Y, Yoo KC, Cui YH, Kim H, Kim MJ, Gyu Kim I, Lee SJ (2015) Activation of KRAS promotes the mesenchymal features of basal-type breast cancer. Exp Mol Med 47:e137. https://doi.org/10.1038/emm.2014.99

    Article  PubMed  PubMed Central  Google Scholar 

  28. Subik K, Lee JF, Baxter L, Strzepek T, Costello D, Crowley P, Xing L, Hung MC, Bonfiglio T, Hicks DG, Tang P (2010) The expression patterns of ER, PR, HER2, CK5/6, EGFR, Ki-67 and AR by immunohistochemical analysis in breast cancer cell lines. Breast Cancer 4:35–41

    PubMed  PubMed Central  Google Scholar 

  29. Sanchez-Munoz A, Gallego E, de Luque V, Perez-Rivas LG, Vicioso L, Ribelles N, Lozano J, Alba E (2010) Lack of evidence for KRAS oncogenic mutations in triple-negative breast cancer. BMC Cancer 10:136. https://doi.org/10.1186/1471-2407-10-136

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bamford S, Dawson E, Forbes S, Clements J, Pettett R, Dogan A, Flanagan A, Teague J, Futreal PA, Stratton MR, Wooster R (2004) The COSMIC (Catalogue of Somatic Mutations in Cancer) database and website. Br J Cancer 91:355–358. https://doi.org/10.1038/sj.bjc.6601894

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Birkeland E, Wik E, Mjos S, Hoivik EA, Trovik J, Werner HM, Kusonmano K, Petersen K, Raeder MB, Holst F, Oyan AM, Kalland KH, Akslen LA, Simon R, Krakstad C, Salvesen HB (2012) KRAS gene amplification and overexpression but not mutation associates with aggressive and metastatic endometrial cancer. Br J Cancer 107:1997–2004. https://doi.org/10.1038/bjc.2012.477

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wan X, Liu R, Li Z (2017) The prognostic value of HRAS mRNA expression in cutaneous melanoma. Biomed Res Int 2017:5356737. https://doi.org/10.1155/2017/5356737

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Cancer Genome Atlas N (2012) Comprehensive molecular portraits of human breast tumours. Nature 490:61–70. https://doi.org/10.1038/nature11412

    Article  CAS  Google Scholar 

  34. Miles WO, Lembo A, Volorio A, Brachtel E, Tian B, Sgroi D, Provero P, Dyson N (2016) Alternative polyadenylation in triple-negative breast tumors allows NRAS and c-JUN to bypass PUMILIO posttranscriptional regulation. Cancer Res 76:7231–7241. https://doi.org/10.1158/0008-5472.CAN-16-0844

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: KML, VM, MBP, and BS; Methodology: KML, VM, LOF; Software: KLM, VM; Validation: KLM, LOF; Writing-Original & Draft Preparation: MBP, VM; Writing-Review & Editing: MBP, VM, TF, IW, LOF, KLM, BS; Project Administration: KLM, VM, MBP, BS.

Corresponding author

Correspondence to Volkmar Müller.

Ethics declarations

Conflict of interest

MBP received lecture honoraria or consultant fees from Roche, Novartis, Eli Lilly, Eisai, and Pfizer. VM received speaker honoraria from Amgen, AstraZeneca, Celgene, Daiichi-Sankyo, Eisai, Pfizer, Pierre Fabre, Novartis, Roche, Teva, and Janssen-Cilag, and served as a consultant/advisor to Genomic Health, Roche, Pierre Fabre, Amgen, Daiichi-Sankyo, and Eisai, and received research grants from Genomic Health, Roche, Pierre Fabre, Amgen, Daiichi-Sankyo, and Eisai. IW received honoraria from MSD, Roche, Pfizer, Novartis, and Daichii Sankyo. TF, BS, LOF, KLM declare no competing interests.

Informed consent

Informed consent was obtained from all individual participants included in this study.

Research involving human participants

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee (Ethik-Kommission der Ärztekammer Hamburg, #OB/V/03) and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. The experiments conducted within this study comply with the current laws of the country in which they were performed.

Additional information

Publisher's Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 44 kb)

REMARK checklist

REMARK checklist

Item to be reported

Page no.

Introduction

1

State the marker examined, the study objectives, and any pre-specified hypotheses

3

Materials and methods

 Patients

  2

Describe the characteristics (e.g., disease stage or co-morbidities) of the study patients, including their source and inclusion and exclusion criteria

5

  3

Describe treatments received and how chosen (e.g., randomized or rule-based)

5

 Specimen characteristics

  4

Describe type of biological material used (including control samples) and methods of preservation and storage

5

 Assay methods

  5

Specify the assay method used and provide (or reference) a detailed protocol, including specific reagents or kits used, quality control procedures, reproducibility assessments, quantitation methods, and scoring and reporting protocols. Specify whether and how assays were performed blinded to the study endpoint

5

 Study design

  6

State the method of case selection, including whether prospective or retrospective and whether stratification or matching (e.g., by stage of disease or age) was used. Specify the time period from which cases were taken, the end of the follow-up period, and the median follow-up time

5

  7

Precisely define all clinical endpoints examined

5–6

  8

List all candidate variables initially examined or considered for inclusion in models

6

  9

Give rationale for sample size; if the study was designed to detect a specified effect size, give the target power and effect size.

5–6

 Statistical analysis methods

  10

Specify all statistical methods, including details of any variable selection procedures and other model-building issues, how model assumptions were verified, and how missing data were handled

6

  11

Clarify how marker values were handled in the analyses; if relevant, describe methods used for cut point determination

6 Table 2

Results

 Data

  12

Describe the flow of patients through the study, including the number of patients included in each stage of the analysis (a diagram may be helpful) and reasons for dropout. Specifically, both overall and for each subgroup extensively examined report the numbers of patients and the number of events

7–8

  13

Report distributions of basic demographic characteristics (at least age and sex), standard (disease-specific) prognostic variables, and tumor marker, including numbers of missing values

7–8, Table 1

 Analysis and presentation

  14

Show the relation of the marker to standard prognostic variables

7–8

Table 1

  15

Present univariable analyses showing the relation between the marker and outcome, with the estimated effect (e.g., hazard ratio and survival probability). Preferably provide similar analyses for all other variables being analyzed. For the effect of a tumor marker on a time-to-event outcome, a Kaplan–Meier plot is recommended

7–8

Figures 4, 5, and 6

  16

For key multivariable analyses, report estimated effects (e.g., hazard ratio) with confidence intervals for the marker and, at least for the final model, all other variables in the model

8

Table 3

  17

Among reported results, provide estimated effects with confidence intervals from an analysis in which the marker and standard prognostic variables are included, regardless of their statistical significance

7–8

  18

If done, report results of further investigations, such as checking assumptions, sensitivity analyses, and internal validation

 

Discussion

 19

Interpret the results in the context of the pre-specified hypotheses and other relevant studies; include a discussion of limitations of the study

9–11

 20

Discuss implications for future research and clinical value

9–11

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Banys-Paluchowski, M., Milde-Langosch, K., Fehm, T. et al. Clinical relevance of H-RAS, K-RAS, and N-RAS mRNA expression in primary breast cancer patients. Breast Cancer Res Treat 179, 403–414 (2020). https://doi.org/10.1007/s10549-019-05474-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10549-019-05474-8

Keywords

Navigation