Radiol Oncol 2023; 57(1): 103-110. doi: 10.2478/raon-2023-0014 103 research article The five-year KRAS, NRAS and BRAF analysis results and treatment patterns in daily clinical practice in Slovenia in 1st line treatment of metastatic colorectal (mCRC) patients with RAS wild-type tumour (wtRAS) – a real- life data report 2013–2018 Tanja Mesti1,2, Martina Rebersek1,2, Janja Ocvirk1,2 1 Department of Medical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia 2 Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia Radiol Oncol 2023; 57(1): 103-110. Received 6 September 2022 Accepted 30 September 2022 Correspondence to: Prof. Janja Ocvirk, M.D., Ph.D., Department of Medical Oncology, Institute of Oncology Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia. E-mail: jocvirk@onko-i.si Disclosure: No potential conflicts of interest were disclosed. This is an open access article distributed under the terms of the CC-BY license (https://creativecommons.org/licenses/by/4.0/). Background. We preformed a Phase IV non-interventional study to assess KRAS, NRAS and BRAF status in metastatic colorectal cancer (mCRC) patients suitable for 1st line treatment and to evaluate the decisions for 1st line treatment considering the treatment goals in the RAS wild type (wt) patients. The aim of our study was also to evaluate the influ- ence of a waiting period for biomarkers analysis on the start of first-line treatment. Patients and methods. Patients with histologically confirmed mCRC adenocarcinoma suitable for first-line treat- ment fulfilling all inclusion criteria were included in the study. The KRAS, NRAS and BRAF analysis was performed from tissue samples of primary tumor site or metastatic site. All included patients have given consent to participate in the study by signing the informed consent form. Results. From April 2013 to March 2018 at the Institute of Oncology Ljubljana 650 patients were included, 637 of them were treated with first- line systemic treatment according to RAS and BRAF status. Remaining 13 patients with mCRC did not receive systemic first-line treatment. The distribution of patients with KRAS mutated and wild-type tumors, was almost equal, 48.8% and 47.9% respectively, 89 % of the patients had wt NRAS tumours and 86.1% had wt BRAF tu- mours. The most frequently prescribed treatment was bevacizumab-based therapy (53.1%), either in combination with doublet chemotherapy or with mono-chemotherapy. EGFR inhibitors cetuximab and panitumumab were prescribed in wt RAS mCRC patients (30.9%). The waiting period for biomarkers analysis was two weeks. Conclusions. Our real-world data, single centre 5-year analysis showed that the distribution between wild type and mutated type tumors of the patients with mCRC was approximately the same, as worldwide, so the Slovenian popula- tion with mCRC has the same ratio distribution of KRAS, NRAS and BRAF wild and mutated genes. We concluded that a two-week waiting period for biomarkers analysis did not influence the first line treatment decision, so it was in the accordance with the worldwide treatment guidelines based on evidence-based medicine. Key words: metastatic colorectal cancer; RAS and BRAF biomarkers; systemic treatment Introduction Colorectal cancer (CRC) is one of the most com- mon cancers and one of the leading causes of cancer death in the world and also in Slovenia. According to the Cancer Registry of Slovenia 2021, 1349 new patients were diagnosed with CRC in 2018.1 Approximately 25% of patients present with Radiol Oncol 2023; 57(1): 103-110. Mesti T et al. / Metastatic colorectal cancer and RAS and BRAF biomarkers104 metastatic disease at diagnosis, and about 50% of patients with CRC will eventually develop metas- tases.2,3 Metastatic disease is still incurable, with 5% five-year overall survival (OS) without treat- ment. Until 1996 five-fluorouracil (5-FU) was the only approved drug for this disease. Since then, five new agents have been approved in the United States and in Europe for this disease, among them irinotecan, oxaliplatin and the targeted therapies cetuximab, bevacizumab and panitumumab.2-16 With the current management of metastatic dis- ease, with chemotherapy with oxaliplatin and irinotecan in combination with biologicals, tar- geting epidermal growth factor mediated growth regulatory pathway and the vascular endothelial growth factor mediated angiogenesis pathway, the progression-free survival (PFS) and OS of these patients can be prolonged. The CRC-related 5-year survival rate approaches 60%.2,3 CRC represents a heterogeneous group of dis- eases. They are promoted by environmental risk factors and various molecular pathways, which in- fluence individual susceptibility to cancer. About 70% of CRCs are sporadic, while 20–30% have a hereditary component, such as Lynch syndrome and familial adenomatous polyposis (FAP).2,3,15-17 Classification of CRC can be divided in anatomic, genetic and molecular transcriptomic classifica- tion. The race, foods, nutrients, carcinogenic agents and increasingly more important the gut microbi- ome act in a specific manner determined by the primary tumour location to promote carcinogen- esis right-sided tumours (RCC) account about to 35% of cases, while left-sided (LCC) and rectal can- cer represent about 65% of cases.17,18 According to genetic classification approximately 85% of CRC, comprises of non-hypermutated, microsatellite stable (MSS) tumours with chromosomal instabil- ity with a high frequency of DNA somatic altera- tions.17,18 Several oncogenes play key roles in promot- ing colorectal cancer.17-20 Among them firstly in clinical practice of mCRC management, onco- genic mutations of Rat sarcoma virus (RAS) and b-Raf murine sarcoma viral oncogene homolog B (BRAF), which activate the mitogen-activated protein kinase (MAPK) signalling pathway, are the most important.21-24 Oncogenic RAS mutations have historically been present in approximately 40–50% of CRC patients. According to additional subsequent analysis, the prevalence of RAS mu- tations in mCRC has been shown to be higher, in 55.9% with mutations in KRAS exon 2 being the most common, they represent 42.6%, followed by KRAS exon 3 in 3.8%, KRAS exon 4 in 6.2%, NRAS exon 2 in 2.9%, NRAS exon3 in 4.2% and NRAS exon4 in 0.3% mutations.16 NRAS mutant tumours are more frequently in older patients and located on the proximal colon. KRAS and NRAS mutant tumours exhibit similar metastasis patterns. RAS mutations are encountered in approximately 50% of the total population worldwide but it also dem- onstrates geographical variations. Therefore, such a country-by-country determination of the mu- tation status of the RAS gene in mCRC patients would be meaningful for personalised treatment decision-making. Selection of patients for anti-epidermal growth factor receptor (EGFR) antibodies based on mo- lecular characteristics of the tumour is very im- portant, because the activity of the anti-EGFR antibodies was confined to wtKRAS tumours (tra- ditionally mutations on codon 12 and 13 of exon 2).22-26 Than the testing was expanded to the other more rare RAS mutations: codon 61 of exon 3 and codon 117 and 146 of exon 4 of KRAS and exons 2, 3 and 4 of N-Rat sarcoma virus (NRAS), which are also predictive to response to anti-EGFR anti- bodies.2 Exon 2 KRAS mutations occur in 40% of CRC cases, and the other KRAS and NRAS muta- tions in 10%–15% of CRC patients. Those muta- tions influence the response to the combinations of cetuximab or panitumumab alone or with irinotecan- and oxaliplatin-based regimens.27-42 Treatment with anti-EGFR antibodies may even harm patients with a RAS mutation, especially when combined with oxaliplatin.4 BRAF mutations (mt BRAF) are present in about 10–15% of CRCs, with mt BRAFV600E mutations being the most frequent.15,16 BRAF encodes a ser- ine/threonine protein kinase, a downstream ef- fector of the KRAS protein, and mt BRAF results in constitutive activation of the MAPK signal- ling pathway. RAS mutations (mt RAS) and mt BRAF mutations are usually mutually exclusive. The most common mutation of the BRAF gene is V600E.15-18 The same was reported in our previ- ous study carried on Slovenian patients with CRC where the mt BRAF V600E was found in 5.1% of pa- tients.22,43,44 A mt BRAF is a strong negative prog- nostic biomarker. The patients with a mt BRAF mCRC have a very poor prognosis. In prognostic analyses, patients with mt BRAF had significantly shorter PFS (7.0 months vs. 12.2 months, HR 2.78; p < 0.001) and OS (13.4 months vs. 37.9 months, HR 5.67; p < 0.001) compared with BRAF wild-type (wt BRAF) patients, whilst patients with mt RAS experienced no difference in PFS (11.0 months vs. Radiol Oncol 2023; 57(1): 103-110. Mesti T et al. / Metastatic colorectal cancer and RAS and BRAF biomarkers 105 12.2 months, HR 1.15; p = 0.241); however, OS was significantly shorter compared with wtRAS pa- tients (26.3 months vs. 37.9 months, HR 1.44; p = 0.015).2,3,41,44 Other, non-V600 mt BRAF are a very rare and occur in 2% of metastatic CRC patients, most frequently in younger male patients, with pa- tients, mostly males. Tumours are generally well differentiated tumours, more frequently harbour concurrent mt RAS. Survival of these patients was shown to be improved compared to metastatic CRC patients with V600E mt BRAF or RAS wild- type (wt RAS).15,16 As the role of targeted therapy for treatment of advanced or mCRC has become increasingly imported, so currently, determination of tumour gene status for KRAS/NRAS and BRAF mutations, as well as HER2 amplifications and MSI/MMR sta- tus, neurotrophic tyrosine receptor kinase (NTRK) fusions are recommended for patients with mCRC. Testing may be carried out for individual genes or as part of an NGS panel.2,3 In order to get the real-life data for Slovenian population with mCRC treated at the Oncology Institute of Ljubljana from 30 April 2013 to 5 March 2018 we have performed a Phase IV non- interventional study to assess KRAS, NRAS and BRAF status in mCRC patients suitable for 1st line treatment. This prospective cohort study was approved by the Institutional Review Board Committee (Approval number: KESOPKR-6 and 03-Z/ KESOPKR-6) and was conducted following the ethical standards defned by the Declaration of Helsinki. The study was conducted with the un- derstanding and the consent of the patients. Prior to treatment patients have signed an informed consent for treatment and that their data could be used for scientific purposes. The aim of this prospective cohort study was to determine in our mCRC patient population the biomarkers status and distribution in the RAS and BRAF genes, and to determine the time from re- ceipt of the histological sample to the determina- tion of the molecular status. Patients and methods We performed a prospective, single-arm, single- centre, non-comparative, case-based, observation- al study from 30 April 2013 to 5 March 2018. The study was comprised of one centre which covers the treatment of all mCRC patients and reflects Slovenian demographic distribution. Originally, all samples for biomarker examination were to be evaluated for KRAS status by the certificated Molecular laboratory at the Institute of Oncology Ljubljana. The data was to be recorded in a collec- tive data base to provide reliability. As from 2013 before initiating treatment with cetuximab, the testing for RAS status included not only KRAS, but also NRAS mutations by an experienced labora- tory using validated test methods for detection of KRAS and NRAS. According to this in November 2013 in addition to KRAS we included also NRAS mutations analysis in all patients included in this study. RAS testing had previously been performed for all patients already included. In this study, 650 mCRC patients treated at the Institute of Oncology Ljubljana were enrolled. Initially, the planned number of patients for the study was 300. Between April 2013 and February 2014, 300 patients were recruited and underwent KRAS testing. The protocol was then amended in view of full RAS testing becoming standard clini- cal practice. The protocol was amended again in November 2015, to include an extra 350 above the original plan – a total of 650, and BRAF testing was added to the list of biomarkers analysed. From November 2015 until March 2018, 350 additional patients were recruited, making a total recruit- ment of 650. Thirteen patients that were included and signed consent to be part of this study, and underwent RAS and BRAF testing, did not present for the first-line systemic treatment afterwards, re- sulting in complete data for 637 patients. The basic data about each patient were record- ed, as date of birth, gender, performance status Eastern Cooperative Oncology Group (ECOG) performance status and smoking habits. The dis- ease characteristics were also included, including date of diagnosis of the primary tumour, tumour location – colon or rectum, initial stage at the time of diagnosis (according to the TNM (tumour T, nodes N, and metastases M) classification sys- tem), the treatment option used for the primary tumour, (radiotherapy or surgery, followed or not by adjuvant chemotherapy, with the duration time treatment and the type of the adjuvant chemo- therapy (capecitabine, 5-FU or oxaliplatin). The date of metastatic diagnosis was also included in the analysis, the sites of metastatic spread and the treatment option used, as surgery and systemic treatment, with systemic treatment comprising the treatment goals – curative, potentially curative or palliative, liver and/or lung metastases potentially respectable, depending upon tumour burden. The systemic first line treatment options included vari- Radiol Oncol 2023; 57(1): 103-110. Mesti T et al. / Metastatic colorectal cancer and RAS and BRAF biomarkers106 ous combinations of irinotecan, oxaliplatin based therapy, 5-FU, capecitabine, FOLFOXIRI protocol, cetuximab in a weekly or biweekly manner, beva- cizumab, panitumumab or other. The tumour characteristics were assessed re- garding with KRAS/RAS and BRAF status. The date of the request for the biomolecular analysis was included, and the date of the analysis receipt. DNA for molecular analysis was extracted from formalin-fixed, paraffin-embedded tumour tissue of primary tumours or metastases with at least 70% of tumour cells. Molecular testing was per- formed with RT-PCR KRAS Mutation Analysis Kit (EntroGen, Inc.), RT-PCR NRAS Mutation Analysis Kit (EntroGen, Inc.) and RT-PCR BRAF Mutation Analysis Kit (EntroGen, Inc.), according to man- ufacturer’s instructions. The KRAS and NRAS analyses were assessed on exons 2, 3 and 4, and for BRAF on exon 15. Statistical methods In this study 95% confidence intervals were calcu- lated to indicate the degree of certainty of KRAS, NRAS and BRAF status frequency as being wild type or mutant type. A sample size of 551-634 patients was neces- sary for a two-sided 95% confidence interval with a width of 4% anticipating a BRAF mutant rate of 5.5% to 6.5%. The planned sample size was in- creased to 650 patients, to consider non-evaluable biomarker test results and missing data. Results In total, 650 patients of the whole mCRC patients treated at Oncology Institute of Ljubljana were en- rolled in this prospective clinical study. Patient’s characteristics and treatment The demographic data about each patient were recorded, such as date of birth, gender, perfor- mance status (WHO) and smoking habits. Two thirds of the patients were male and most (84.9%) of the patients initially had a very good (ECOG 0–1) performance status. The main localization of the primary tumour was colon in 60.8% of the pa- tients. All of included patients in this study had pathological confirmation of the carcinoma of the colon or rectum. Almost one third (27.9%) of the patients had clinical signs for CRC, before being pathologically confirmed, including occult bleed- ing or were asymptomatic and detected in the Slovenian national program for the early detec- tion of the colorectal cancer named SVIT. For more than half (56.7%) of patients the diagnosis was con- firmed pathologically, after biopsy was performed at colonoscopy, and for the others after the surgery. TABLE 1. Patients baseline characteristics Patients baseline characteristics Number (%) Gender female male 233 (36.6) 404 (63.4) WHO clasiffication: 0 1 2 241 (37.8) 300 (47.1) 72 (11.3) Tumor location: colon rectum 387 (60.8) 250 (39.2) Clinical signs of the primary colorectal cancer present before pathological confirmation 178 (20.9) Primary metastatic 361 (56.7) Liver metastases 218 (34.3) * Some pathohistological reports from external hospital had unclear description, about the TNM status and risk factors TABLE 2. Disease characteristics Disease characteristics Number (%) pT4 of primary tumor 186 (29.2) Affected regional lymph nodes (N): N0 (no affected regional lymph nodes) N1 (1 to 3 affected regional lymph nodes) N2 (more than 3 affected regional lymph nodes) Missing data* 110 (17.3) 209 (32.8) 239 (37.5) 62 (11.3) Vascular invasion 114 (17.9) Perineural invasion 95 (27.9) Lymphangiosis 115 (18.1) Grade of differentiation: G1 (well) G2 (medium) G3 (poorly) G4 (no differentiated) Missing data* 19 (3) 162 (25.4) 53 (8.3) 1 (0.2) 402 (63.2) Resection of primary tumour R0 R1 R2 63 (9.9) 10 (1.6) 16 (2.5) * Other metastatic locations: brain, ovaries, suprarenal glands, local recurrence, muscular dissemination Radiol Oncol 2023; 57(1): 103-110. Mesti T et al. / Metastatic colorectal cancer and RAS and BRAF biomarkers 107 Complete resection (R0) of operated primary tu- mour was achieved in 9.9%. Most of the included patients were non-smokers (62.6%). According to the TNM characteristics, initially at the first presentation of the primary CRC, in the external hospitals, 53.7% of patients had T3 tu- mours, 31.6% of patients had N1 (31,6%) and 34.5% of patients had N2. One third of patients had ini- tially no metastatic disease (33.4%). Mostly, the primary CRC had pathohistological characteristics as follows: grade 2, with one third having vascu- lar invasion, perineural invasion or lymphangi- osis. Primary metastatic disease was confirmed in 56.7% of patients. The most common sites of metastases were liver and lung. Some of patients with mCRC in liver and lungs had surgery of the metastasis before the mCRC treatment, 6.8% and 2.0% respectively. In 10% of the mCRC patients R0 resection (70.8% of the total number of resections) was achieved. The metastatic sites were included in the analysis, and the main sites of CRC dis- semination were liver (34.2%), lungs (11.0%), lymph nodes (5.3%), peritoneum (5.0%). Sixty-two patients were no previous smokers, and only 9% of patients were smoking at the time of our analysis. Patients’ baseline characteristics and are shown in Table 1, disease characteristics are shown in Table 2, the most frequent metastatic site are shown in Table 3, smoking habits of included patients are shown in Table 4. The treatment goals were in oligometastatic disease as follows: respectable and potentially re- sectable, or palliative for patients with high bur- den of the disease. More than a half (58.7%) of the patients were initially candidates for the palliative treatment, and 24.2% were potentially resectable at presentation. Treatment goal options at presenta- tion of the mCRC patients are shown in Figure 1. More than half (58.7%) of patients were initially candidates for palliative systemic treatment, and 24.2% were potentially resectable at presentation. Of all 637 mCRC patients had the following possible first line systemic treatment options: chemotherapy - fluoropyrimidine based systemic therapy combined with oxaliplatin and/or irinote- can plus, for subjects with KRAS/RAS wild type tumours, the EGFR inhibitors cetuximab or pani- tumumab, or the VEGF inhibitor bevacizumab for those with KRAS/RAS mutated tumours. Most TABLE 3. The most frequent metastatic sites The most frequent metastatic sites Number (%) Total 637 (100) Liver 218 (34.3) Lungs 70 (11.0) Lymph nodes 34 (5.3) Peritoneum 115 (18.1) Bones 4 (0.6) Other* 18 (2.8) Multiple locations 261 (41.0) Palliative/sequential treatment 374 pts (58.7%) Early tumour shrinkage (ETS) 109 pts (17.1 %) Liver±Lung- potentially resectable 154 pts (24.2%) FIGURE 1. Treatment goal options at presentation of the metastatic colorectal cancer (mCRC) patients. TABLE 4. Treatment regimen decision for the first line treatment of the metastatic colorectal cancer (mCRC) Systemic therapy options Number (%) Fluoropirimidines/Irinotecan/Cetuximab* 73 (11.4) Fluoropirimidines/Oxaliplatin/Cetuximab* 49 (7.7) Fluoropirimidines/Irinotecan/Panitumumab* 17 (2.7) Fluoropirimidines/Oxaliplatin/Panitumumab* 12 (1.9) EGFR inhibitors other** 46 (7.2) Fluoropirimidines/Irinotecan/Bevacizumab 126 (19.8) Fluoropirimidines/Oxaliplatin/Bevacizumab 191 (30.0) VEGF inhibitor other*** 21 (3.3) Chemotherapy only 102 (16.0) *EGFR inhibitors for RAS (NRAS and KRAS) wild type only **EGFR inhibitors monotherapy ± one chemotherapy ***VEGF inhibitor monotherapy ± one chemotherapy Radiol Oncol 2023; 57(1): 103-110. Mesti T et al. / Metastatic colorectal cancer and RAS and BRAF biomarkers108 frequently used was bevacizumab-based therapy (53.1%), either in combination with doublet chemo- therapy or with one cytostatic or as monotherapy. EGFR inhibitors cetuximab and panitumumab were used in RAS (KRAS/NRAS) wild type mCRC subjects (30.9%). Data presented in the Table 9. Fifty-one subjects with RAS (KRAS/NRAS) wild type tumours received chemotherapy alone or in combination with Bevacizumab, as first line treat- ment. The first line treatment decision is made by the oncologist in accordance with NCCN and ESMO guidelines and consider the patient’s pref- erences. Some patients found skin side effects and higher parenteral application frequency important parameters for a possible decrease of their quality of life. Most frequently prescribed treatment pro- tocols are shown in Table 4. Biomarker status The assessment of the KRAS/RAS and BRAF status was performed for all 650 patients. The RAS and BRAF status analysis were performed simultane- ously, and for the group of subjects from the first Amendment, after the Amendment II was added to the protocol, the NRAS and BRAF data were added to the database. Data of the RAS/KRAS and BRAF status distribution are shown in Table 5. The distribution of patients with KRAS mutated and wild-type tumours, was almost equal, 48.8% and 47.9% respectively. For 3.3% of patients of the as- sessed population the KRAS status analysis was unsuccessful, due to unrepresentative tumour samples (Table 6). For 2.0% and 0.7% of the subjects the NRAS and BRAF genotyping was not possi- ble due to unrepresentative tumour samples. The KRAS and NRAS analyses were assessed on ex- ons 2, 3 and 4, and for BRAF on exon 15. The most frequent mutation was in KRAS in codon 12 and codon 13, and the same was true for NRAS and for the BRAF V600E mutation. The most frequent mutations, as follows were: pGly12Asp (c35 G>A) 7.8%, pGly 12Val (c35G>T) 6.6%, pGly13Asp (c38 G>A) 4.7% and pGly12Cys (c34 G>T) 2.4%. The median time for biomarkers status assess- ment was 14 days. Mostly direct sequencing was used for KRAS/ RAS and BRAF status assessment, but also oth- er methods, as pyrosequencing and qPCR. The Entrogen RT PCR KRAS/RAS and BRAF mutation Kit was used. Methods used for KRAS/RAS and BRAF analysis are shown in Table 6. Discussion The aim of the observational mCRC population- based study was to determine the biomarker sta- tus in the RAS and BRAF genes, to determine their distribution in the mCRC patient Slovenian popu- lation, and to determine the time from receiving the histological sample to the determination of the biomarker status and its impact on the initiation of systemic oncological therapy. This study was performed in order to achieve a real perspective of the bio-markers status of the patients with metastatic colorectal cancer in comparison with worldwide data. The analysis of KRAS/NRAS and BRAF status was a relatively new approach in the treatment of the patients with metastatic colorectal cancer at the time of conduct- ing of our clinical study, so we also wanted to get real insight of the time frame needed for such anal- ysis and its possible effect on the initiation of the first-line treatment. The status of mutations in the RAS gene is a mo- lecular predictive factor for response to treatment with EGFR inhibitors inmCRC. Determination of mutational status in KRAS gene has been stand- ard clinical practice since 2008. Additional muta- tions in the codons of 61 and 146 of KRAS gene, and in codons 12, 13, 61 and 146 of NRAS gene are determined at our Institute of Oncology Ljubljana TABLE 5. The RAS/KRAS and BRAF status distribution The RAS/KRAS and BRAF status distribution Number (%) KRAS testing KRAS mutated KRAS wild type Not possible 637 (100) 311 (48.8) 305 (47.9) 21 (3.3) NRAS testing NRAS mutated NRAS wild type Not possible 637 (100) 57 (9.0) 567 (89.0) 13 (2.0) BRAF testing BRAF mutated BRAF wild type Not possible 637 (100) 84 (13.2) 548 (86.1) 5 (0.7) TABLE 6. Methods used for KRAS/RAS and BRAF analysis Methods used for KRAS/RAS and BRAF analysis Number (%) Direct sequencing 223 (35.0) Pyrosequencing 322 (50.5) qPCR 92 (14.5) Radiol Oncol 2023; 57(1): 103-110. Mesti T et al. / Metastatic colorectal cancer and RAS and BRAF biomarkers 109 since autumn 2013 and are being standard in inter- national clinical practice since 2014. According to the literature data, are about 15%. In our prospective cohort study KRAS muta- tions were found in 48.8% of patients, and addi- tionally 9% of NRAS mutations were determined. So approximately 60% of patients had RAS mutat- ed mCRC, which is consistent with reports from previous literature.2,3,22,45 In our retrospective anal- ysis we found 17% of additional mutations in RAS gene, which is a higher percentage than in our pro- spective cohort study, probably due to characteris- tics of patient population included.45 Also, higher, 13.2% BRAF mutations, were de- termined in our cohort study. According to the lit- erature, the frequency of this mutation is from 5 to 10%. 2,3,24 According to our previous retrospective analysis, the V600E mutation in the BRAF gene was also present at a similar percentage of patients, in 7.4%.46 It is assumed that this difference in the per- centage of BRAF mutations in clinical studies is due to the characteristics of the patients included in the analysis. So, according to our study analysis data, the distribution between wild type and mutated type tumours of mCRC patients was approximately the same, as worldwide, so the Slovenian popula- tion with mCRC has the same ratio distribution of KRAS, NRAS and BRAF wild and mutated genes. The two-week time period from the initial pres- entation of the patient until the biomarker status analysis report did not affect the starting of the systemic treatment or the treatment decision, as usually mCRC patients are given a brief period of time for psychical and physical preparation for the systemic treatment, at which time supportive care – nutritional and symptomatic support, is given. As for the time spent on biomarkers analysis, we concluded that the two-week period was not influ- encing the first line treatment decision, because patients started with systemic chemotherapy im- mediately and after 2 weeks have already received a biologic drug at the 2nd cycle of chemotherapy ac- cording to molecular genetic testing, And also in Slovenia we do not have waiting lists and cancer patients, who need treatment are referred to the oncology treatment facility shortly after the diag- nosis, so this brief period of time is usual for psy- chical and physical preparation for the systemic treatment, at which time supportive care – nutri- tional and symptomatic support, is given. In conclusion, we have proven that the bio- markers distribution in Slovenian population with mCRC was approximately the same as worldwide, so the decision treatment approach should be in the accordance with the worldwide treatment guidelines based on evidence-based medicine. Secondly, the laboratory for the molecular analysis that we have in the Institute of Oncololgy Ljubljana was performing the needed biomarkers analysis in an acceptable time that didn’t affect the treatment decision or delay the needed cancer treatment. Acknowledgement The authors acknowledge the financial support from the state budget by the Slovenian Research Agency (ARRS), program No. P3-0321. References 1. Cancer in Slovenia 2018. Ljubljana: Institute of Oncology Ljubljana, Epidemiology and Cancer Registry, Slovenian Cancer Registry; 2021. 2. Schmoll HJ, Van Cutsem E, Stein A, Valentini V, Glimelius B, Haustermans K, et al. ESMO Consensus Guidelines for management of patients with colon and rectal cancer. A personalized approach to clinical decision making. Ann Oncol 2012; 23: 2479-516. doi:10.1093/annonc/mds236 3. van Cutsem E, Cervantes A, Adam R, Sobrero S, J. 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. doi:10.1093/ annonc/mdw235 4. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: Colon Cancer. V.1.2022. [cited 2022 Aug 25]. Avaible at: http:// www.nccn.org/profesionals/phisician_gls/PDF/colon.pdf 5. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: Rectal Cancer. V.1.2022. [cited 2022 Aug 25]. Avaible at: http:// www.nccn.org/profesionals/phisician_gls/PDF/colon.pdf 6. Tournigand C, André T, Achille E, Lledo G, Flesh M, Mery-Mignard D, et a. FOLFIRI followed by FOLFOX6 or the reverse sequence in advanced colorec- tal cancer: a randomized GERCOR study. J Clin Oncol 2004; 22: 229-37. doi: 10.1200/JCO.2004.05.113. Epub 2003 Dec 2. PMID: 14657227 7. Grothey A, Sargent D, Goldberg RM, Schmoll HJ. Survival of patients with advanced colorectal cancer improves with the availability of fluorouracil- leucovorin, irinotecan, and oxaliplatin in the course of treatment. J Clin Oncol 2004; 22: 1209-14. doi: 10.1200/JCO.2004.11.037. PMID: 15051767 8. Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for meta- static colorectal cancer. N Engl J Med 2004; 350: 2335-42. doi: 10.1056/ NEJMoa032691 9. Saltz LB, Clarke S, Díaz-Rubio E, Scheithauer W, Figer A, Wong R, et al. Bevacizumab in combination with oxaliplatin-based chemotherapy as first- line therapy in metastatic colorectal cancer: a randomized phase III study. J Clin Oncol 2008; 26: 2013-9. doi: 10.1200/JCO.2007.14.9930. Erratum in: J Clin Oncol 2008; 26: 3110. Erratum in: J Clin Oncol 2009; 27: 653. PMID: 18421054 10. Van Cutsem E, Köhne CH, Hitre E, Zaluski J, Chang Chien CR, Makhson A, et al. Cetuximab and chemotherapy as initial treatment for metastatic colorec- tal cancer. N Engl J Med 2009; 360: 1408-17. doi: 10.1056/NEJMoa0805019 11. Bokemeyer C, Bondarenko I, Makhson A, Hartmann JT, Aparicio J, de Braud F, et al. Fluorouracil, leucovorin, and oxaliplatin with and without cetuximab in the first-line treatment of metastatic colorectal cancer. J Clin Oncol 2009; 27: 663-71. doi: 10.1200/JCO.2008.20.8397 12. Bokemeyer C, Bondarenko I, Hartmann JT, de Braud F, Schuch G, Zubel A, et al. Efficacy according to biomarker status of cetuximab plus FOLFOX-4 as first-line treatment for metastatic colorectal cancer: the OPUS study. Ann Oncol 2011; 22: 1535-46. doi: 10.1093/annonc/mdq632 Radiol Oncol 2023; 57(1): 103-110. Mesti T et al. / Metastatic colorectal cancer and RAS and BRAF biomarkers110 13. Folprecht G, Grothey A, Alberts S, Raab HR, Köhne CH. Neoadjuvant treat- ment of unresectable colorectal liver metastases: correlation between tumour response and resection rates. Ann Oncol 2005; 16: 1311-9. doi: 10.1093/annonc/mdi246 14. Adam R, Delvart V, Pascal G, Valeanu A, Castaing D, Azoulay D, et al. Rescue surgery for unresectable colorectal liver metastases downstaged by chemo- therapy: a model to predict long-term survival. Ann Surg 2004; 240: 644-57; discussion 657-8. doi: 10.1097/01.sla.0000141198.92114.f6 15. Bellio H, Fumet JD, Ghiringhelli F. Targeting BRAF and RAS in colorectal cancer. Cancers 2021; 13: 2201. doi: 10.3390/cancers13092201 16. Gong J, Cho M, Fakih M. RAS and BRAF in metastatic colorectal cancer management. J Gastrointest Oncol 2016; 7: 687-704. doi: 10.21037/ jgo.2016.06.12 17. Bos JL, Fearon ER, Hamilton SR, Verlaan-de Vries M, van Boom JH, van der Eb AJ, et al. Prevalence of ras gene mutations in human colorectal cancers. Nature 1987; 327: 293-7. doi: 10.1038/327293a0. PMID: 3587348 18. Vogelstein B, Fearon ER, Hamilton SR, Kern SE, Preisinger AC, Leppert M, et al. Genetic alterations during colorectal-tumor development. N Engl J Med 1988; 319: 525-32. doi: 10.1056/NEJM198809013190901 19. Zhu D, Keohavong P, Finkelstein SD, Swalsky P, Bakker A, Weissfeld J, et al. K-ras gene mutations in normal colorectal tissues from K-ras mutation- positive colorectal cancer patients. Cancer Res 1997; 57: 2485-92. PMID: 9192830 20. Markowitz SD, Bertagnolli MM. Molecular basis of colorectal cancer. N Engl J Med 2009; 361: 2449-60. doi: 10.1056/NEJMra0804588 21. Sebolt-Leopold JS. Advances in the development of cancer therapeutics directed against the RAS-mitogen-activated protein kinase pathway. Clin Cancer Res 2008; 14: 3651-6. doi: 10.1158/1078-0432.CCR-08-0333 22. Ličar A, Cerkovnik P, Novaković S. Distribution of some activating KRAS and BRAF mutations in Slovene patients with colorectal cancer. Med Oncol 2011; 28: 1048-53. doi: 10.1007/s12032-010-9631-z 23. De Roock W, Jonker DJ, De Nicolantonio F, Sartore-Bianchi A, Tu D, Siena S, et al. Association of KRAS p.G13D mutation with outcome in patients with out- come with chemotherapy-refractory metastatic colorectal cancer treated with cetuximab. JAMA 2010; 304: 1812-20. doi: 10.1001/jama.2010.1535 24. De Roock W, Claes B, Bernasconi D, De Schutter J, Biesmans B, Fountzilas G, et al. Effect of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol 2010; 11: 753-62. doi: 10.1016/S1470-2045(10)70130-3 25. Artale S, Sartore- Bianchi A, Veronese SM, Gambi V, Sarnataro CS, Gambacorta M, et al. Mutations of KRAS and BRAF in primary and matched metastatic sites of colorectal cancer. J Clin Oncol 2008; 26: 4217-9. doi:10.1200/JCO.2008.18.7266 26. Artale S, Sartore-Bianchi A, Veronese SM, Gambi V, Sarnataro CS, Gambacorta Met al. Mutations of KRAS and BRAF in primary and matched metastatic sites of colorectal cancer. J Clin Oncol 2008; 26: 4217-9. doi: 10.1200/JCO.2008.18.7286 27. Lamprechts D, De Roock W, Prenen H, Schutter JD, Jacobs B, Biesmans B, et al. The role of KRAS, BRAF, NRAS and PIK3CA mutations as a markers of resistence of cetuximab in chemorefractory metastatic colorectal cancer. J Clin Oncol 2009; 27: 9020. doi: 10.1158/1078-0432.CCR-08-2961 28. Tejpar S, De Roock W, Claes B, Fountzilas G. PIKI3CA, BRAF and KRAS muta- tions and outcome prediction in chemorefractory metastatic colorectal cancer (mCRC) patients treated with EGFR targeting monoclonal antibodies (MoAbs): results of a European Consortium. 8th Annual Meeting of the Japanese-Society-of-Medical-Oncology Ann Oncol 2010; 21(Suppl 9): 6. 29. Loupakis F, Ruzzo A, Cremolini C, Vincenzi B, Salvatore L, Santini D, et al. KRAS codon 61, 146 and BRAF mutations predict resistence to cetuximab + irinotecan in KRAS codon 12 and 13 wild-type metastatic colorectal cancer. Br J Cancer 2009; 101: 715-21. doi: 10.18632/oncotarget.13697 30. Peters M, Douillard JY, Van Cutsem E, Siena S, Zhang K, Williams R et al. Mutant (MT) KRAS codon 12 and 13 alleles in patients (pts) with metastatic colorectal cancer (mCRC): assessment as prognostic and predictive bio- markers of response to panitumumab (pmab). [abstract]. J Clin Oncol 2012; 30(Suppl 4): abstr 383. 31. Tejpar S, Celic I, Schlichting M, Bokemeyer C, Van Cutsem E. Association of KRAS G13D tumor mutations with outcome in patients with metastatic colorectal cancer treated with first-line chemotherapy with or without ce- tuximab. J Clin Oncol 2012; 30: 3570-7. doi: 10.1200/JCO.2012.42.2592 32. Fujiyoshi K, Yamamoto G, Takahashi A, Arai Y, Yamada M, Kakuta M, et al. High concordance rate of KRAS/BRAF mutations and MSI-H between pri- mary colorectal cancer and corresponding metastases. Oncol Rep 2017; 37: 785-92. doi: 10.3892/or.2016.5323 33. Gaedcke J, Grade M, Jung K, Schirmer M, Jo P, Obermeyer C, et al. KRAS and BRAF mutations in patients with rectal cancer treated with preopera- tive chemoradiotherapy. Radiother Oncol 2010; 94: 76-81. doi: 10.1016/j. radonc.2009.10.001 34. Pietrantonio F, Cremolini C, Petrelli F, Di Bartolomeo M, Loupakis F, Maggi C, et al. First-line anti-EGFR monoclonal antibodies in panRAS wild-type metastatic colorectal cancer: a systematic review and meta-analysis. Crit Rev Oncol Hematol 2015; 96: 156-66. doi: 10.1016/j.critrevonc.2015.05.016 35. Sorich MJ, Wiese MD, Rowland A, Kichenadasse G, McKinnon RA, Karapetis CS, et al. Extended RAS mutations and anti-EGFR monoclonal antibody sur- vival benefit in metastatic colorectal cancer: a meta-analysis of randomized, controlled trials. Ann Oncol 2015; 26: 13-21. doi: 10.1093/annonc/mdu378 36. Therkildsen C, Bergmann TK, Henrichsen-Schnack T, Ladelund S, Nilbert M. The predictive value of KRAS, NRAS, BRAF, PIK3CA and PTEN for anti-EGFR treatment in metastatic colorectal cancer: a systematic review and meta- analysis. Acta Oncol 2014; 53: 852-64. doi: 10.3109/0284186X.2014.895036 37. Karapetis CS, Khambata-Ford S, Jonker DJ, O’Callaghan CJ, Tu D, Tebbutt NC, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med 2008; 359: 1757-65. doi: 10.1056/NEJMoa0804385 38. Samowitz WS, Curtin K, Schaffer D, Robertson M, Leppert M, Slattery ML. Relationship of Ki-ras mutations in colon cancers to tumor location, stage, and survival: a population-based study. Cancer Epidemiol Biomarkers Prev 2000; 9: 1193-7. PMID: 11097226 39. Van Cutsem E Köhne CH, La´ng I, Folprecht G, Nowacki MP, Cascinu S, et al. Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treat- ment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J Clin Oncol 2011; 29: 2011-9. doi: 10.1200/JCO.2010.33.5091 40. Bokemeyer C, Bondarenko I, Hartmann JT, de Braud F, Schuch G, Zubel A, et al. Efficacy according to biomarker status of cetuximab plus FOLFOX-4 as first-line treatment for metastatic colorectal cancer: the OPUS study. Ann Oncol 2011; 22: 1535-46. doi: 10.1093/annonc/mdq632 41. Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med 2013; 369: 1023-34. doi: 10.1056/NEJMoa1305275 42. Cremolini C, Loupakis F, Antoniotti C, Lupi C, Sensi E, Lonardi S, et al. FOLFOXIRI plus bevacizumab versus FOLFIRI plus bevacizumab as first-line treatment of patients with metastatic colorectal cancer: updated overall survival and molecular subgroup analyses of the open-label, phase 3 TRIBE study. Lancet Oncol 2015; 16: 1306-15. doi: 10.1016/S1470-2045(15)00122- 9. 43. Tol J, Nagtegaal ID, Punt CJA. BRAF mutation in metastatic colorectal cancer. N Engl J Med 2009; 361: 98-9. doi: 10.3978/j.issn.2078-6891.2015.077 44. Samowitz WS, Sweeney C, Herrick J, Albertsen H, Levin TR, Murtaugh MA, et al. Poor survival associatedwith the BRAF V600E mutation in microsat- ellite-stable colon cancer. Caner Res 2005; 65: 6063-9. doi: 10.1158/0008- 5472.CAN-05-0404 45. Rebersek M, Mesti T, Boc M, Ocvirk J. Molecular biomarkers and histological parameters impact on survival and response to first- line systemic therapy of metastatic colorectal cancer patients. Radiol Oncol 2019; 53: 85-95. doi: 10.2478/raon-2019-0013 46. Rebersek M, Boc M, Škerl P, Benedik J, Hlebanja Z, Volk N, et al. Efficacy of First- line systemic treatment in correlation with BRAF V600E and differ- ent KRAS mutations colorectal cancer - a single institution retrospective analysis. Radiol Oncol 2011; 45: 285-91. doi: 10.2478/v10019-011-0039-y