232
Views
3
CrossRef citations to date
0
Altmetric
Oncology

KRAS/NRAS Mutations Associated with Distant Metastasis and BRAF/PIK3CA Mutations Associated with Poor Tumor Differentiation in Colorectal Cancer

, , , & ORCID Icon
Pages 4109-4120 | Received 02 Jul 2023, Accepted 30 Aug 2023, Published online: 11 Sep 2023

Abstract

Background

The occurrence, progression, and prognosis of colorectal cancer (CRC) are regulated by EGFR-mediated signaling pathways. However, the relationship between the core genes (KRAS/NRAS/BRAF/PIK3CA) status in the signaling pathways and clinicopathological characteristics of CRC patients in Hakka population remains controversial.

Methods

Patients were genotyped for KRAS (codons 12, 13, 61, 117, and 146), NRAS (codons 12, 61, 117, and 146), BRAF (codons 600), and PIK3CA (codons 542, 545 and 1047) mutations. Clinical records were collected, and clinicopathological characteristic associations were analyzed together with mutations of studied genes.

Results

Four hundred and eight patients (256 men and 152 women) were included in the analysis. At least one mutation in the four genes was detected in 216 (52.9%) patients, while none was detected in 192 (47.1%) patients. KRAS, NRAS, BRAF, and PIK3CA mutation status were detected in 190 (46.6%), 11 (2.7%), 10 (2.5%), 34 (8.3%) samples, respectively. KRAS exon 2 had the highest proportion (62.5%). Age, tumor site, tumor size, lymphovascular invasion, and perineural invasion were not associated with gene mutations. KRAS mutations (adjusted OR 1.675, 95% CI 1.017–2.760, P=0.043) and NRAS mutations (adjusted OR 5.183, 95% CI 1.239–21.687, P=0.024) appeared more frequently in patients with distant metastasis. BRAF mutations (adjusted OR 7.224, 95% CI 1.356–38.488, P=0.021) and PIK3CA mutations (adjusted OR 3.811, 95% CI 1.268–11.455, P=0.017) associated with poorly differentiated tumor.

Conclusion

KRAS/NRAS mutations are associated with distant metastasis and BRAF/PIK3CA mutations are associated with poor tumor differentiation in CRC. And the results provided a better understanding between clinicopathological characteristics and gene mutations in CRC patients.

Introduction

Colorectal cancer (CRC) is a common malignancy of the gastrointestinal tract. According to the latest estimates of global cancer incidence and mortality, CRC (10.0%) ranks third in incidence after breast cancer and lung cancer (11.7% and 11.4%, respectively), and ranks second in mortality (9.4%) after lung cancer (18%).Citation1 In China, from 2015 to 2020, the incidence of CRC increased rapidly, while gastrointestinal cancer (stomach, colorectal, liver and esophageal cancer) had the highest mortality rate, and CRC ranked fifth in mortality after lung, liver, stomach and esophageal cancer.Citation2 With the clinical application of anti-epidermal growth factor receptor (EGFR) drug cetuximab and anti-angiogenic drug bevacizumab, targeted therapy has become the first-line treatment for CRC.Citation3

The occurrence, progression, and prognosis of CRC are regulated by some molecular signal transduction pathways, such as EGFR-mediated signaling pathways. As a transmembrane tyrosine kinase receptor, EGFR can activate Ras/RAF/MAPK and PI3K/AKT/mTOR pathways after binding to corresponding ligands, thereby inducing tumor cell proliferation, invasion, metastasis and angiogenesis.Citation4 Target drug tyrosine kinase inhibitors (TKI) targeting the EGFR have a significant therapeutic effect in cancer patients with EGFR gene mutations.Citation5 In addition, mutations in some genes downstream of EGFR signaling pathway, such as KRAS proto-oncogene (KRAS), NRAS proto-oncogene (NRAS), B-Raf proto-oncogene (BRAF) and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), will affect the targeted therapeutic effect of anti-EGFR monoclonal antibody drugs.Citation6 Mutations in these genes result in the EGFR signaling pathway always in an active status, making EGFR-targeted drug therapy ineffective.Citation7,Citation8 In recent years, KRAS/NRAS/BRAF/PIK3CA wild-type CRC patients have significantly benefited from anti-EGFR monoclonal antibodies.

The different populations, lifestyles, and the interaction between some environmental factors and genetic polymorphisms will affect the clinicopathological characteristics of CRC patients.Citation9 In addition, the clinical characteristics of CRC are also related to age, and although study has found that older pT4 patients were more likely to develop serious postoperative complications, the prognosis of older patients may vary depending on the stage, tumor site, and pre-existing comorbidities.Citation10 Study has shown that KRAS, NRAS or BRAF mutations were associated with CRC metastasis.Citation11,Citation12 However, study has shown that distant metastasis was not related to KRAS, NRAS, BRAF, or PIK3CA mutations.Citation13 Study has shown that BRAF mutations were associated with poorer differentiation, but mutant KRAS was associated with greater differentiation,Citation14 and PIK3CA was not associated with differentiation.Citation15–18 Therefore, in different sample sizes, different populations and different studies, the results of the correlation between the studied gene status and clinicopathological features are inconsistent.

The association of the studied gene status with clinicopathological features will provide valuable information for clinicians to better assess the disease severity and disease progression, such as colorectal cancer brain metastases. The development of brain metastases is an important factor in overall cancer mortality in patients with advanced cancer, and the prognosis is poor.Citation19 Clinicians can monitor the progress of such patients with a combination of screening techniques to achieve early detection and treatment. Electroencephalogram (EEG) biomarkers differ significantly in patients with consciousness after severe acquired brain injury, which can be used as diagnosis and prediction for clinical evaluation of patients.Citation20 EEG alpha band indices can be used by clinicians to diagnose, select and evaluate treatment for main neuropsychiatric disorders of the brain.Citation21

The Hakka is a Han ethnic group with a unique genetic background formed by the Hakka ancestors from the Han nationality in central China, who migrated southward for many times and fused with the ancient Yue residents in Guangdong, Fujian, and Jiangxi.Citation22 At present, there is no study on the relationship between the mutation status of KRAS, NRAS, BRAF, PIK3CA genes in EGFR signaling pathway and the clinicopathological features of CRC in Hakka population. Therefore, this study retrospectively analyzed mutations of these genes in CRC tissues, and explored their relationship with clinicopathological features, aiming to provide valuable data for precise treatment of CRC individuals.

Materials and Methods

Participants

After approval from the Ethics Committee of the Meizhou People’s Hospital (Clearance No.: 2020-A-57), 408 CRC patients who received KRAS/NRAS/BRAF/PIK3CA molecular testing were retrospectively investigated between January 2019 and October 2020. The inclusion criteria for the study subjects were: (1) colorectal cancer was confirmed by hematoxylin and eosin (HE) staining and histological analysis; (2) at the time of enrolment, patients presenting in stage I to IV (according to the American Joint Committee on Cancer (AJCC) guidelinesCitation23) were enrolled; (3) there was complete medical records; (4) aged more than 18 years. The exclusion criteria for the study subjects as follows: (1) patients with other tumors disease other than colorectal cancer; (2) other circumstances inconsistent with the inclusion criteria mentioned above.

Detection of Mismatch Repair (MMR) Protein by Immunohistochemistry

All specimens were immobilized with 4% neutral methanol solution and embedded with paraffin. Max Vision two-step method was used for immunohistochemistry. Paraffin sections were dewaxed and hydrated, then washed with PBS. Peroxidase blocker was used to block endogenous peroxidase activity. Primary antibody was added and incubated at room temperature, biotin-labeled secondary antibody was added and streptomycin antibiotin peroxidase solution was added. After incubation, Hematoxylin-3,3’-Diaminobenzidine (DAB) was added for coloration, restained with hematoxylin, dehydration, transparent, and tablets were sealed. The expression of mismatch repair proteins (MutS homolog 2 (MSH2), MutS homolog 6 (MSH6), MutL homolog 1 (MLH1), and PMS1 homolog 2 (PMS2)) was observed under the microscope. When samples from patients were positive for all four of these mismatch repair proteins (>30%) were identified as mismatch repair-proficient (pMMR), and mismatch repair-deficient (dMMR) was identified when one or more proteins were missing.

Genotyping for KRAS, NRAS, BRAF and PIK3CA Genes

Formalin-fixed and paraffin-embedded (FFPE) tissue was cut into 5μm thick slices, and 10 slices were loaded into EP tubes for DNA extraction. DNA was extracted using the AmoyDx® Tissue DNA Kit (Spin Column) (Amoy Diagnostics, Xiamen, China) according to the manufacturers’ instructions. NanoDrop 2000 (Thermo Scientific) was used to measure DNA concentration. The DNA samples should be diluted to 2 ng/μL, and the OD260/OD280 should be 1.8–2.0.

Genetic tests were performed using amplification refractory mutation system polymerase-chain reaction (ARMS-PCR) with corresponding mutation detection kits, respectively (Amoy Diagnostics Co. Ltd, Xiamen, China). The DNA of the sample to be tested, positive quality control DNA, and negative quality control DNA were added to the PCR reaction solution, respectively. PCR amplification was performed according to the following conditions: initial denaturation at 95°C for 5 minutes; 95°C 25 seconds, 64°C 20 seconds, 72°C 20 seconds run 15 cycles; 95°C 25 seconds, 60°C 35 seconds, 72°C 20 seconds run 31 cycles; Amplification was performed in a LightCycler 480 real-time PCR system (Roche Diagnostics, Germany). According to the amplification curve of the experimental results, the mutation cycle threshold (Ct) values of each reaction tube and external control of the sample were determined, and the sample to be tested was determined as mutation positive or negative according to the obtained Ct values. The genetic sites tested mainly included common mutations in exons 2, 3 and 4 of KRAS (codons 12, 13, 61, 117, and 146), exons 2, 3 and 4 of NRAS (codons 12, 61, 117, and 146), exons 9 and 20 of PIK3CA (codons 542, 545 and 1047), and exon 15 of BRAF (codons 600).

Data Collection and Statistical Analysis

Clinical records, including age, sex, tumor site, maximum diameter of tumor, histological type, tumor differentiation, lymphovascular invasion, perineural invasion, and TNM stage were collected. SPSS statistical software version 21.0 (IBM Inc., State of New York, USA) was used for data analysis. The patients’ clinicopathological features were summarized with descriptive statistics. Categorical variables were compared using χ2 test and Fisher’s exact test. Gender, age, tumor site, maximum diameter of tumor, tumor differentiation, lymphovascular invasion, perineural invasion, and distant metastasis were selected as covariates in the multivariate logistic regression analysis for KRAS, NRAS, BRAF, PIK3CA mutations, based on estimating the odds ratios (OR) and their 95% confidence intervals (CIs). The significance test was two-sided, and a P value <0.05 was considered statistically significant.

Results

Characteristics of Subjects

Four hundred and eight patients were included in the analysis, including 256 (62.7%) men and 152 (37.3%) women. The age of all patients was ranging from 25 to 86 years old. There were 185 (45.3%) patients with <60 years old, and 223 (54.7%) patients with ≥60 years old. The number of colon, rectum and cecum tumors was 227 (55.6%), 175 (42.9%) and 6 (1.5%), respectively. There were 255 patients (62.5%) with tumor maximum diameter less than 5 cm, and 138 patients (33.8%) with tumor maximum diameter ≥5 cm. Well differentiation, moderate differentiation and poor differentiation tumor detected in 4 (1.0%), 370 (90.7%) and 34 (8.3%) cases, respectively. There were 80 (19.6%) and 54 (13.2%) patients with lymphatic vascular space invasion and perineural invasion, respectively. And 317 (77.7%) cases, and 91 (22.3%) cases were classified as stage I-III, and stage IV, respectively. There were 15 (3.7%) cases with dMMR ().

Table 1 Baseline Characteristics of CRC Patients

Frequency and Composition Ratio of KRAS/NRAS/BRAF/PIK3CA Mutations

All patients were genotyped for KRAS (codons 12, 13, 61, 117, and 146), NRAS (codons 12, 61, 117, and 146), BRAF (codons 600), and PIK3CA (codons 542, 545 and 1047) mutations. At least one mutation in these genes was detected in 216 (52.9%) patients, while none was detected in 192 (47.1%) patients. Mutation in KRAS, NRAS, BRAF, and PIK3CA was detected in 190 (46.6%), 11 (2.7%), 10 (2.5%), 34 (8.3%) samples, respectively. Among them, mutations in KRAS and NRAS, KRAS and BRAF, KRAS and PIK3CA were detected simultaneously in 1 (0.2%), 1 (0.2%), and 27 (6.6%) samples, respectively.

Ranking the mutations in order of composition, mutations in KRAS exon 2 had the highest proportion (62.5%), followed by mutations in both KRAS exon 2 and PIK3CA exon 9 simultaneously (7.9%), KRAS exon 4 (6.9%), KRAS exon 3 (5.1%), BRAF exon 15 (4.2%), both KRAS exon 2 and PIK3CA exon 20 simultaneously (3.7%), NRAS exon 2 (3.2%), and PIK3CA exon 9 (2.8%) ().

Table 2 Frequency and Composition Ratio of KRAS/NRAS/BRAF/PIK3CA Mutation in CRC Patients

Relationship of Clinicopathological Characteristics and KRAS/NRAS/BRAF/PIK3CA Gene Mutation Status

Fisher’s exact test was used to compare the clinicopathological differences between the KRAS/NRAS/BRAF/PIK3CA mutant and wild-type patients. KRAS, NRAS, BRAF, and PIK3CA mutations were not related to gender, age, tumor site, tumor size (maximum diameter of tumor), perineural invasion, lymph node metastasis, and MMR status (all P>0.05). The percentage of mutations in KRAS gene in stage IV patients was significantly higher than that in stage I-III patients (56.0% (51/91) vs 43.8% (139/317), P=0.043), and that in patients with distant metastasis was higher than that in patients without distant metastasis (57.1% (52/91) vs 43.5% (138/317), P=0.024). The mutation percentage of BRAF gene was higher in the tumor with poor differentiation than that in the tumor with moderate and well differentiation (13.3% (4/30) vs 1.6% (6/364) and 0 (0/4), P=0.012), and it was also significantly higher in the patients with lymphovascular invasion than without it (6.3% (5/80) vs 1.5% (5/328), P=0.029) ().

Table 3 Clinicopathological Characteristics According to KRAS/NRAS/BRAF/PIK3CA Gene Mutation Status in CRC Patients

The regression analysis was performed with KRAS mutations, NRAS mutations, BRAF mutations and PIK3CA mutations as the response variables, and gender (X1), age (X2), tumor site (X3), maximum diameter of tumor (X4), tumor differentiation (X5), lymphovascular invasion (X6), perineural invasion (X7), and distant metastasis (X8) as independent variables. And multicollinearity and model goodness-of-fit was checked. The multivariate logistic regression analysis demonstrated that KRAS mutations (adjusted OR 1.675, 95% CI 1.017–2.760, P=0.043) and NRAS mutations (adjusted OR 5.183, 95% CI 1.239–21.687, P=0.024) appeared more frequently in patients with distant metastasis. BRAF mutations (adjusted OR 7.224, 95% CI 1.356–38.488, P=0.021) and PIK3CA mutations (adjusted OR 3.811, 95% CI 1.268–11.455, P=0.017) appeared more frequently in tumor with poor differentiation. Moreover, a relationship still existed between PIK3CA mutations and female patients (male/female adjusted OR 0.443, 95% CI 0.210–0.934, P=0.032) ().

Table 4 Multivariate Logistic Regression in CRC Patients Between Gene Mutations and Clinicopathological Characteristics

Discussion

CRC is a common malignant tumor in human digestive tracts.Citation24 There is evidence that the occurrence of tumors is related to inflammation, and blood inflammatory markers can be used as markers for risk prediction and prognosis assessment of CRC. In addition, the significance of microRNAs (miRNAs) in tumor diagnosis, prognosis and prediction is constantly being explored.Citation25 In the past decades, a large number of research data have emerged in the studies on the molecular basis,Citation26,Citation27 drug investigation and usage,Citation28,Citation29 and gene testingCitation30 of colorectal cancer, leading to the rapid development of gene testing and targeted therapy for CRC. The targeted therapy of CRC patients needs to be based on the genes’ status, and molecular testing has been paid more and more attention.Citation30 Studies have shown that genes downstream of the EGFR signaling pathway can affect the efficacy of targeted therapy with monoclonal antibodies.Citation31

KRAS is one of the most important oncogenic genes in human genome, KRAS gene activating mutations are found in >80% of pancreatic cancer, >30% of CRC, cholangiocarcinoma and lung adenocarcinoma.Citation32 The frequency of mutations may vary from region to region and population to population. Approximately 41.5% of Danish CRC patients have KRAS mutations,Citation33 45.5% in Slovene patients,Citation34 49.6% in Saudi Arabian patients,Citation35 34.7% in sigmoid colon and 58.2% in cecum of patients in the United States,Citation36 29% in Iranian patients,Citation37 54.84% in Turks patients,Citation38 42% in Japanese patients,Citation39 41.0% in Vietnamese patients,Citation40 and 23% in Indian patients.Citation41 The frequency of KRAS mutations was 35.0%–50.0% in Chinese CRC patients.Citation42–46 The frequency of NRAS mutations was 4.2% in Danish CRC patients,Citation33 7.3% in Tunisian patients,Citation47 2.0% in Saudi Arabian patients,Citation35 5% in Japanese patients,Citation39 9.6% in Vietnamese patients,Citation40 2.0% in Indian patients,Citation41 and 7% in Mexican patients.Citation48 The mutation percentage of NRAS was 1.2%–3.85% in Chinese CRC patients.Citation45,Citation46,Citation49 In this study, at least one mutation in KRAS, NRAS, BRAF and PIK3CA was detected in 219 (52.6%) CRC patients, while none was detected in 197 (47.4%) CRC patients. KRAS and NRAS mutation was detected in 190 (46.6%) and 11 (2.7%) samples, respectively. The mutation percentage of KRAS and NRAS gene of CRC patients in this study is basically the same as that in other populations.

The frequency of BRAF was 18.0% in Danish CRC patients,Citation33 0.4% in Saudi Arabian patients,Citation35 7% in Iranian patients,Citation37 12.9% in Turks patients,Citation38 7% in Japanese patients,Citation39 8.3% in Vietnamese patients,Citation40 17% in Indian patients,Citation41 and 12.44% in Greek and Romanian patients.Citation50 The frequency of BRAF mutations was 2.3%–8% in Chinese CRC patients.Citation45,Citation49,Citation51–53 The frequency of PIK3CA mutations was 18.8% in Danish CRC patients,Citation33 13.3% in Arab patients,Citation54 and 8% in Italian patients.Citation55 The frequency of PIK3CA mutations was about 9.4%–18.9% in Chinese CRC patients.Citation42,Citation51,Citation56,Citation57 In this study, BRAF and PIK3CA mutation status were detected in 10 (2.5%) and 34 (8.3%) samples, respectively. Compared with other populations, the mutation percentage of BRAF and PIK3CA genes of CRC patients in this study was relatively low.

In this study, KRAS or NRAS mutations appeared more frequently in CRC patients with distant metastasis. Studies have shown that KRAS, NRAS or BRAF mutations were associated with lung metastasis in CRC.Citation11,Citation12 KRAS/NRAS/BRAF mutations may predict late distant metastasis.Citation58 KRAS mutations were related to poor tumour differentiation and liver metastasis.Citation59 Distant metastatic tumors had a higher mutation percentage of NRAS mutation but not KRAS.Citation15 BRAF mutations were more common in peritoneal metastasis patients.Citation16 However, another study has shown that distant metastasis was not related to KRAS, NRAS, BRAF, or PIK3CA mutations.Citation13 In addition, BRAF and PIK3CA mutations appeared more frequently in tumor with poor differentiation in this study. Study has shown that BRAF mutations were associated with poorer differentiation, but mutant KRAS was associated with greater differentiation.Citation14 BRAF mutations were more common in poorly differentiated tumors, but PIK3CA is not.Citation15–18 PIK3CA mutations showed null associations with tumor differentiation.Citation60 In summary, the correlation of KRAS, NRAS, BRAF, PIK3CA mutations with distant metastasis, tumor differentiation was inconsistent. Different sample sizes, different populations, and different detection methods for genetic mutations can partly explain the results. In addition, the prognosis of CRC is also associated with DNA mismatch repair, and CRC patients with microsatellite instability-low (MSI-L) may experience shorter survival; however, there are challenges in the availability of data for MSI testing.Citation61

In terms of molecular mechanisms, mutation-activated KRAS in tumor cells reprograms macrophages with tumor-associated macrophage (TAM)-like phenotypes, which not only promotes tumor progression but also induces resistance of tumor cells to targeted therapy.Citation62 MiR-450b-5p can activate Wnt/β-catenin signaling to promote cell proliferation, tumor growth, and inhibit the apoptosis of CRC cells, while miR-450b-5p can be up-regulated by KRAS/AP-1 signaling.Citation63 The differential expression of some RNAs in exosomes of CRC cells with BRAF V600E mutation is closely related to proliferation, metabolism of tumor cells and tumor microenvironment changes.Citation64 B-Raf/MEK/ERK pathway was related to inhibition of tumor cell differentiation.Citation65 The possible mechanisms of the relationship between distant metastasis and KRAS and NRAS mutations, as well as the relationship between poor tumor differentiation and BRAF and PIK3CA mutations, are still not completely clear and need further study.

The results of this study suggest that for CRC patients with KRAS and NRAS mutations, more attention should be paid to the possibility of distant metastases, such as liver metastasis, lung metastasis, and brain metastasis. Some techniques can be applied to the diagnosis of liver metastases in CRC.Citation66 Tumor brain metastases are the most common tumors of the adult central nervous system,Citation67 and the rate of brain metastases in CRC is the highest among gastrointestinal tumors. Brain metastases usually occur in the advanced stages of the disease and the prognosis is poor in most patients. The quality of life and prognosis of patients with colorectal cancer with neurological symptoms can be improved by early detection and treatment of metastatic lesions. CRC patients with KRAS and NRAS mutations with neurological symptoms, through the application of some new technology such as non-invasive brain stimulation techniques (NIBS),Citation68–71 can be used to identify the risk factors for brain metastases, so that early detection and treatment.

The study has some limitations that are worth noting. First of all, the number of research objects in this study is relatively small, which leads to some deviations in the results. Second, we only studied the common mutation sites of KRAS/NRAS/BRAF/PIK3CA genes; the status of the other mutation sites in these genes is unknown. Third, this study was limited to the correlation between gene mutations and clinicopathological features, and did not analyze the correlation between gene mutations and clinical outcomes and treatment responses in CRC patients receiving chemotherapy or radiotherapy. Therefore, the next step is to conduct a multi-center study with a larger sample size and a comprehensive analysis of KRAS/NRAS/BRAF/PIK3CA gene.

Conclusions

The relationship between clinicopathological features and KRAS, NRAS, BRAF, PIK3CA genes status in CRC patients were studied, and found that there was a significant relationship between distant metastasis and KRAS or NRAS mutations, while poor tumor differentiation and BRAF or PIK3CA mutations. Importantly, more attention should be paid to the possibility of distant metastasis in colorectal cancer patients with KRAS and NRAS mutations. Monitoring these patients for early diagnosis and treatment of distant metastases will have the potential to improve their survival and prognosis.

Ethics Approval and Consent to Participate

The study was approved by the Ethics Committee of Medicine, Meizhou People’s Hospital. All participants signed informed consent in accordance with the Declaration of Helsinki.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Disclosure

The authors declare that they have no competing interests in this work.

Acknowledgments

The author would like to thank other colleagues whom were not listed in the authorship of Department of Medical Oncology, Meizhou People’s Hospital for their helpful comments on the manuscript.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Additional information

Funding

This study was supported by the Science and Technology Program of Meizhou [Grant No.: 2019B0202001], and the Social Development Science and Technology Program Project of Meizhou [2022B21].

References

  • Sung H, Ferlay J, Siegel RL. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209–249. doi:10.3322/caac.21660
  • Cao W, Chen HD, Yu YW, Li N, Chen WQ. Changing profiles of cancer burden worldwide and in China: a secondary analysis of the global cancer statistics 2020. Chin Med J. 2021;134(7):783–791. doi:10.1097/CM9.0000000000001474
  • Xie YH, Chen YX, Fang JY. Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct Target Ther. 2020;5(1):22. doi:10.1038/s41392-020-0116-z
  • Martinelli E, Ciardiello D, Martini G, et al. Implementing anti-epidermal growth factor receptor (EGFR) therapy in metastatic colorectal cancer: challenges and future perspectives. Ann Oncol. 2020;31(1):30–40. doi:10.1016/j.annonc.2019.10.007
  • Chan DLH, Segelov E, Wong RS, et al. Epidermal growth factor receptor (EGFR) inhibitors for metastatic colorectal cancer. Cochrane Database Syst Rev. 2017;6(6):Cd007047. doi:10.1002/14651858.CD007047.pub2
  • Sforza V, Martinelli E, Ciardiello F, et al. Mechanisms of resistance to anti-epidermal growth factor receptor inhibitors in metastatic colorectal cancer. World J Gastroenterol. 2016;22(28):6345–6361. doi:10.3748/wjg.v22.i28.6345
  • Miyamoto Y, Suyama K, Baba H. Recent Advances in Targeting the EGFR Signaling Pathway for the Treatment of Metastatic Colorectal Cancer. Int J Mol Sci. 2017;18(4):752. doi:10.3390/ijms18040752
  • Khan K, Valeri N, Dearman C, et al. Targeting EGFR pathway in metastatic colorectal cancer- tumour heterogeneity and convergent evolution. Crit Rev Oncol Hematol. 2019;143:153–163. doi:10.1016/j.critrevonc.2019.09.001
  • Fitzmaurice C, Abate D, Abbasi N, et al. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived With Disability, and Disability-Adjusted Life-Years for 29 Cancer Groups, 1990 to 2017: a Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol. 2019;5(12):1749–1768. doi:10.1001/jamaoncol.2019.2996
  • Osseis M, Nehmeh WA, Rassy N, Derienne J. Surgery for T4 colorectal cancer in older patients: determinants of outcomes. J Pers Med. 2022;12(9):1534. doi:10.3390/jpm12091534
  • Michl M, Taverna F, Kumbrink J, et al. Biomarker alterations associated with distinct patterns of metastatic spread in colorectal cancer. Virchows Arch. 2021;478(4):695–705. doi:10.1007/s00428-020-02983-6
  • Lipsyc M, Yaeger R. Impact of somatic mutations on patterns of metastasis in colorectal cancer. J Gastrointest Oncol. 2015;6(6):645–649. doi:10.3978/j.issn.2078-6891.2015.045
  • Kleist B, Meurer T, Poetsch M. Mitochondrial DNA alteration in primary and metastatic colorectal cancer: different frequency and association with selected clinicopathological and molecular markers. Tumour Biol. 2017;39(3):1010428317692246. doi:10.1177/1010428317692246
  • Zhang J, Zheng J, Yang Y, et al. Molecular spectrum of KRAS, NRAS, BRAF and PIK3CA mutations in Chinese colorectal cancer patients: analysis of 1110 cases. Sci Rep. 2015;5(1):18678. doi:10.1038/srep18678
  • Shen Y, Wang J, Han X, et al. Effectors of epidermal growth factor receptor pathway: the genetic profiling ofKRAS, BRAF, PIK3CA, NRAS mutations in colorectal cancer characteristics and personalized medicine. PLoS One. 2013;8(12):e81628. doi:10.1371/journal.pone.0081628
  • Kawazoe A, Shitara K, Fukuoka S, et al. A retrospective observational study of clinicopathological features of KRAS, NRAS, BRAF and PIK3CA mutations in Japanese patients with metastatic colorectal cancer. BMC Cancer. 2015;15(1):258. doi:10.1186/s12885-015-1276-z
  • Yaeger R, Cercek A, Chou JF, et al. BRAF mutation predicts for poor outcomes after metastasectomy in patients with metastatic colorectal cancer. Cancer. 2014;120(15):2316–2324. doi:10.1002/cncr.28729
  • Chang SC, Lin PC, Lin JK, et al. Mutation spectra of common cancer-associated genes in different phenotypes of colorectal carcinoma without distant metastasis. Ann Surg Oncol. 2016;23(3):849–855. doi:10.1245/s10434-015-4899-z
  • Achrol AS, Rennert RC, Anders C, et al. Brain metastases. Nat Rev Dis Primers. 2019;5(1):5. doi:10.1038/s41572-018-0055-y
  • Di Gregorio F, La Porta F, Petrone V, Battaglia S. Accuracy of EEG Biomarkers in the Detection of Clinical Outcome in Disorders of Consciousness after Severe Acquired Brain Injury: preliminary Results of a Pilot Study Using a Machine Learning Approach. Biomedicines. 2022;10(8):1897. doi:10.3390/biomedicines10081897
  • Ippolito G, Bertaccini R, Tarasi L. The role of alpha oscillations among the main neuropsychiatric disorders in the adult and developing human brain: evidence from the last 10 years of research. Biomedicines. 2022;10(12):3189. doi:10.3390/biomedicines10123189
  • Wang WZ, Wang CY, Cheng YT, et al. Tracing the origins of Hakka and Chaoshanese by mitochondrial DNA analysis. Am J Phys Anthropol. 2010;141(1):124–130. doi:10.1002/ajpa.21124
  • Bruni D, Angell HK. The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy. Nat Rev Cancer. 2020;20(11):662–680. doi:10.1038/s41568-020-0285-7
  • Dekker E, Tanis PJ, Vleugels JLA, Kasi PM, Wallace MB. Colorectal cancer. Lancet. 2019;394(10207):1467–1480. doi:10.1016/S0140-6736(19)32319-0
  • Boussios S, Ozturk MA. The developing story of predictive biomarkers in colorectal cancer. J Pers Med. 2019;9(1):12. doi:10.3390/jpm9010012
  • Markowitz SD, Bertagnolli MM. Molecular origins of cancer: molecular basis of colorectal cancer. N Engl J Med. 2009;361(25):2449–2460. doi:10.1056/NEJMra0804588
  • Wang MY, Qiu YH, Cai ML, et al. Role and molecular mechanism of stem cells in colorectal cancer initiation. J Drug Target. 2020;28(1):1–10. doi:10.1080/1061186X.2019.1632317
  • Garcia J, Hurwitz HI, Sandler AB, et al. Bevacizumab (Avastin®) in cancer treatment: a review of 15 years of clinical experience and future outlook. Cancer Treat Rev. 2020;86:102017. doi:10.1016/j.ctrv.2020.102017
  • Piawah S, Venook AP. Targeted therapy for colorectal cancer metastases: a review of current methods of molecularly targeted therapy and the use of tumor biomarkers in the treatment of metastatic colorectal cancer. Cancer. 2019;125(23):4139–4147. doi:10.1002/cncr.32163
  • Biller LH, Schrag D. Diagnosis and treatment of metastatic colorectal cancer: a review. JAMA. 2021;325(7):669–685. doi:10.1001/jama.2021.0106
  • Aleksakhina SN, Imyanitov EN. Cancer therapy guided by mutation tests: current status and perspectives. Int J Mol Sci. 2021;22(20):10931. doi:10.3390/ijms222010931
  • Timar J, Kashofer K. Molecular epidemiology and diagnostics of KRAS mutations in human cancer. Cancer Metastasis Rev. 2020;39(4):1029–1038. doi:10.1007/s10555-020-09915-5
  • Poulsen TS, de Oliveira D, Espersen MLM, Klarskov LL, Skovrider-Ruminski W, Hogdall E. Frequency and coexistence of KRAS, NRAS, BRAF and PIK3CA mutations and occurrence of MMR deficiency in Danish colorectal cancer patients. APMIS. 2021;129(2):61–69. doi:10.1111/apm.13091
  • Licar A, Cerkovnik P, Ocvirk J, Novakovic S. KRAS mutations in Slovene patients with colorectal cancer: frequency, distribution and correlation with the response to treatment. Int J Oncol. 2010;36(5):1137–1144. doi:10.3892/ijo_00000596
  • Alharbi A, Bin Dokhi H. Prevalence of colorectal cancer biomarkers and their impact on clinical outcomes in Riyadh, Saudi Arabia. PLoS One. 2021;16(5):e0249590. doi:10.1371/journal.pone.0249590
  • Uhlig J, Cecchini M, Sheth A, Stein S, Lacy J, Kim HS. Microsatellite Instability and KRAS Mutation in Stage IV Colorectal Cancer: prevalence, Geographic Discrepancies, and Outcomes From the National Cancer Database. J Natl Compr Canc Netw. 2021;19(3):307–318. doi:10.6004/jnccn.2020.7619
  • Yari A, Samoudi A. Mutation Status and Prognostic Value of KRAS and BRAF in Southeast Iranian Colorectal Cancer Patients: first Report from Southeast of Iran. J Gastrointest Cancer. 2021;52(2):557–568. doi:10.1007/s12029-020-00426-8
  • Bagci B, Sari M, Karadayi K, Turan M, Ozdemir O. BRAF oncogene mutations and tissue specific promoter hypermethylation of tumor suppressor SFRP2, DAPK1, MGMT, HIC1 and p16 genes in colorectal cancer patients. Cancer Biomark. 2016;17(2):133–143. doi:10.3233/CBM-160624
  • Ikoma T, Shimokawa M, Kotaka M, et al. Clinical and prognostic features of patients with detailed RAS/BRAF-mutant colorectal cancer in Japan. BMC Cancer. 2021;21(1):518. doi:10.1186/s12885-021-08271-z
  • Ta TV, Nguyen QN, Chu HH, Truong VL, Vuong LD. RAS/RAF mutations and their associations with epigenetic alterations for distinct pathways in Vietnamese colorectal cancer. Pathol Res Pract. 2020;216(4):152898. doi:10.1016/j.prp.2020.152898
  • Bagadi SB, Sanghvi M, Nair SB, Das BR. Combined mutational analysis of KRAS, NRAS and BRAF genes in Indian patients with colorectal carcinoma. Int J Biol Markers. 2012;27(1):27–33. doi:10.5301/JBM.2012.9108
  • Huang W, Li H, Shi X, et al. Characterization of genomic alterations in Chinese colorectal cancer patients. Jpn J Clin Oncol. 2021;51(1):120–129. doi:10.1093/jjco
  • Yuan Y, Liu Y, Wu Y, et al. Clinical characteristics and prognostic value of the KRAS mutation in Chinese colorectal cancer patients. Int J Biol Markers. 2021;36(2):33–39. doi:10.1177/17246008211017152
  • Chang XN, Shang FM, Jiang HY, et al. Clinicopathological Features and Prognostic Value of KRAS/NRAS/BRAF Mutations in Colorectal Cancer Patients of Central China. Curr Med Sci. 2021;41(1):118–126. doi:10.1007/s11596-021-2326-1
  • Song Y, Wang L, Ran W, et al. Effect of tumor location on clinicopathological and molecular markers in colorectal cancer in eastern china patients: an analysis of 2356 cases. Front Genet. 2020;11:96. doi:10.3389/fgene.2020.00096
  • Guo F, Gong H, Zhao H, et al. Mutation status and prognostic values of KRAS, NRAS, BRAF and PIK3CA in 353 Chinese colorectal cancer patients. Sci Rep. 2018;8(1):6076. doi:10.1038/s41598-018-24306-1
  • Ounissi D, Weslati M. Clinicopathological characteristics and mutational profile of KRAS and NRAS in Tunisian patients with sporadic colorectal cancer. Turk J Med Sci. 2021;51(1):148–158. doi:10.3906/sag-2003-42
  • Sanchez-Ibarra HE, Jiang X, Gallegos-Gonzalez EY, et al. KRAS, NRAS, and BRAF mutation prevalence, clinicopathological association, and their application in a predictive model in Mexican patients with metastatic colorectal cancer: a retrospective cohort study. PLoS One. 2020;15(7):e0235490. doi:10.1371/journal.pone.0235490
  • Yang Q, Huo S, Sui Y, et al. Mutation Status and Immunohistochemical Correlation of KRAS, NRAS, and BRAF in 260 Chinese Colorectal and Gastric Cancers. Front Oncol. 2018;8:487. doi:10.3389/fonc.2018.00487
  • Negru S, Papadopoulou E, Apessos A, et al. KRAS, NRAS and BRAF mutations in Greek and Romanian patients with colorectal cancer: a cohort study. BMJ Open. 2014;4(5):e004652. doi:10.1136/bmjopen-2013-004652
  • Wang Y, Liu H, Hou Y, et al. Performance validation of an amplicon-based targeted next-generation sequencing assay and mutation profiling of 648 Chinese colorectal cancer patients. Virchows Arch. 2018;472(6):959–968. doi:10.1007/s00428-018-2359-4
  • Zhang X, Wu J, Wang L, Zhao H, Li H. HER2 and BRAF mutation in colorectal cancer patients: a retrospective study in Eastern China. Peer J. 2020;8:e8602. doi:10.7717/peerj.8602
  • Jia X, Li B, Wang H, Yan Z. Clinical features, molecular alterations and prognosis of colorectal adenocarcinoma with mucinous component in Chinese patients. Appl Immunohistochem Mol Morphol. 2021;29(10):765–772. doi:10.1097/PAI.0000000000000950
  • Al-Shamsi HO, Jones J, Fahmawi Y, et al. Molecular spectrum of KRAS, NRAS, BRAF, PIK3CA, TP53, and APC somatic gene mutations in Arab patients with colorectal cancer: determination of frequency and distribution pattern. J Gastrointest Oncol. 2016;7(6):882–902. doi:10.21037/jgo.2016.11.02
  • Reggiani Bonetti L, Barresi V. Clinical Impact and Prognostic Role of KRAS/BRAF/PIK3CA Mutations in Stage I Colorectal Cancer. Dis Markers. 2018;2018:2959801. doi:10.1155/2018/2959801
  • Fu X, Lin H, Fan X, et al. The Spectrum, Tendency and Predictive Value of PIK3CA Mutation in Chinese Colorectal Cancer Patients. Front Oncol. 2021;11:595675. doi:10.3389/fonc.2021.595675
  • Chang YS, Lee CC, Ke TW, et al. Molecular characterization of colorectal cancer using whole-exome sequencing in a Taiwanese population. Cancer Med. 2019;8(8):3738–3747. doi:10.1002/cam4.2282
  • Liu J, Zeng W, Huang C, Wang J, Xu L, Ma D. Upregulation of c-mesenchymal epithelial transition expression and RAS mutations are associated with late lung metastasis and poor prognosis in colorectal carcinoma. Exp Ther Med. 2018;15(5):4229–4242. doi:10.3892/etm.2018.5966
  • Li Z, Chen Y, Wang D, Wang G, He L, Suo J. Detection of KRAS mutations and their associations with clinicopathological features and survival in Chinese colorectal cancer patients. J Int Med Res. 2012;40(4):1589–1598. doi:10.1177/147323001204000439
  • Jin J, Shi Y, Zhang S, Yang S. PIK3CA mutation and clinicopathological features of colorectal cancer: a systematic review and Meta-Analysis. Acta Oncol. 2020;59(1):66–74. doi:10.1080/0284186X.2019.1664764
  • Adeleke S, Haslam A, Choy A, Diaz-Cano S, Galante JR. Microsatellite instability testing in colorectal patients with Lynch syndrome: lessons learned from a case report and how to avoid such pitfalls. Per Med. 2022;19(4):277–286. doi:10.2217/pme-2021-0128
  • Liu H, Liang Z, Zhou C, et al. Mutant KRAS triggers functional reprogramming of tumor-associated macrophages in colorectal cancer. Signal Transduct Target Ther. 2021;6(1):144. doi:10.1038/s41392-021-00534-2
  • Ye YP, Wu P, Gu CC, et al. miR-450b-5p induced by oncogenic KRAS is required for colorectal cancer progression. Oncotarget. 2016;7(38):61312–61324. doi:10.18632/oncotarget.11016
  • Zhi J, Jia XJ, Yan J, et al. BRAF(V600E) mutant colorectal cancer cells mediate local immunosuppressive microenvironment through exosomal long noncoding RNAs. World J Gastrointest Oncol. 2021;13(12):2129–2148. doi:10.4251/wjgo.v13.i12.2129
  • Herr R, Köhler M, Andrlová H, et al. B-Raf inhibitors induce epithelial differentiation in BRAF-mutant colorectal cancer cells. Cancer Res. 2015;75(1):216–229. doi:10.1158/0008-5472.CAN-13-3686
  • Boraschi P, Donati F, Cervelli R, et al. Colorectal liver metastases: ADC as an imaging biomarker of tumor behavior and therapeutic response. Eur J Radiol. 2021;137:109609. doi:10.1016/j.ejrad.2021.109609
  • Lah TT, Novak M, Breznik B. Brain malignancies: glioblastoma and brain metastases. Semin Cancer Biol. 2020;60:262–273. doi:10.1016/j.semcancer.2019.10.010
  • Borgomaneri S, Battaglia S, Sciamanna G, Tortora F, Laricchiuta D. Memories are not written in stone: re-writing fear memories by means of non-invasive brain stimulation and optogenetic manipulations. Neurosci Biobehav Rev. 2021;127:334–352. doi:10.1016/j.neubiorev.2021.04.036
  • Borgomaneri S, Battaglia S, Avenanti A, Pellegrino GD. Don’t Hurt Me No More: state-dependent Transcranial Magnetic Stimulation for the treatment of specific phobia. J Affect Disord. 2021;286:78–79. doi:10.1016/j.jad.2021.02.076
  • Tanaka M, Vécsei L. Editorial of Special Issue “Crosstalk between Depression, Anxiety, and Dementia: comorbidity in Behavioral Neurology and Neuropsychiatry”. Biomedicines. 2021;9(5):517. doi:10.3390/biomedicines9050517
  • Spekker E, Tanaka M, Szabó Á, Vécsei L. Neurogenic Inflammation: the Participant in Migraine and Recent Advancements in Translational Research. Biomedicines. 2021;10(1):76. doi:10.3390/biomedicines10010076