Next Article in Journal
ERβ Isoforms Have Differential Clinical Significance in Breast Cancer Subtypes and Subgroups
Next Article in Special Issue
Whole-Transcriptome Profiling on Small FFPE Samples: Which Sequencing Kit Should Be Used?
Previous Article in Journal
Laminin 511-E8 Fragment Offers Superior Adhesion Properties for Gastric Cancer Cells Compared with Full-Length Laminin 511
Previous Article in Special Issue
Next-Generation Sequencing of Connective Tissue Genes in Patients with Classical Ehlers-Danlos Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Comparison of Next-Generation Sequencing and Polymerase Chain Reaction for Personalized Treatment-Related Genomic Status in Patients with Metastatic Colorectal Cancer

1
Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
2
Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
3
Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
4
Department of Surgery, Kaohsiung Municipal Tatung Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
5
Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
6
Joint Biobank, Office of Human Research, Taipei Medical University, Taipei 10675, Taiwan
7
Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
8
Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
9
Cohort Research Center, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
10
Pingtung Hospital, Ministry of Health and Welfare, Pingtung 90054, Taiwan
11
Clinical Pharmacogenomics and Pharmacoproteomics, School of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
*
Author to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2022, 44(4), 1552-1563; https://doi.org/10.3390/cimb44040106
Submission received: 14 March 2022 / Revised: 1 April 2022 / Accepted: 3 April 2022 / Published: 5 April 2022
(This article belongs to the Special Issue Next-Generation Sequencing (NGS) Technique and Personalized Medicine)

Abstract

:
Personalized treatments based on the genetic profiles of tumors can simultaneously optimize efficacy and minimize toxicity, which is beneficial for improving patient outcomes. This study aimed to integrate gene alterations associated with predictive and prognostic outcomes in patients with metastatic colorectal cancer (mCRC) with polymerase chain reaction (PCR) and in-house next-generation sequencing (NGS) to detect KRAS, NRAS, and BRAF mutations. In the present study, 41 patients with mCRC were assessed between August 2017 and June 2019 at a single institution. The overall concordance between NGS and PCR results for detecting KRAS, NRAS, and BRAF mutations was considerably high (87.8–92.7%), with only 15 discrepant results between PCR and NGS. Our companion diagnostic test analyzes KRAS, NRAS, and BRAF as a panel of CRC molecular targets; therefore, it has the advantages of requiring fewer specimens and being more time and cost efficient than conventional testing for separate analyses, allowing for the simultaneous analysis of multiple genes.

1. Introduction

With approximately 1.93 million newly diagnosed cases and 935,000 deaths occurring in 2020, colorectal cancer (CRC) is the third most diagnosed cancer and the second leading cause of cancer death worldwide [1]. In Taiwan, CRC is among the most frequently diagnosed malignancies and the third leading cause of cancer death [2]. Fluorouracil (5-FU) administration is the established standard of care for patients with CRC, but the treatment landscape for metastatic cancer has evolved quickly after the approval of several targeted therapies, leading to improved tumor response rates and patient survival [3].
Molecular testing has provided new insights and guidance regarding oncological management in CRC due to the increased availability of new targeted therapies [4]. Therefore, such tests are routinely incorporated into CRC resection specimen reports [4]. Treatments for human cancer are increasingly incorporating precision medicine. Targeting the genomic status of the tumors with molecularly targeted therapies has become a common modality [5]. Drugs that target molecular pathways are available for patients who have CRC and exhibit relevant gene variations.
The introduction and approval of targeted therapies for metastatic CRC (mCRC) have led to improved patient outcomes [6]. Although various treatments are available, the outcomes and toxicity associated with each regimen can vary from patient to patient [7]. Personalized treatments based on genetic profiles of tumors can simultaneously optimize efficacy and minimize toxicity, which improves CRC treatment outcomes [6].
Established data from clinical trials have demonstrated that rat sarcoma virus (RAS) mutations in codons 12 and 13 predict a poor response to monoclonal antibodies (panitumumab/cetuximab) that target the epidermal growth factor receptors (EGFRs), which is detrimental to therapies that combine monoclonal antibodies with oxaliplatin- or irinotecan-based chemotherapy [6,8,9,10,11,12,13,14]. Numerous studies have shown that the proto-oncogene B-Raf (BRAF) mutation is associated with considerably poor clinical outcomes in patients with mCRC [15,16,17], regardless of the treatment modality that was adopted [18].
If a traditional analysis method such as polymerase chain reaction (PCR) is used to detect all the aforementioned molecular alterations, a larger amount of specimens is required, and the sample quality is likely to be low or variable; however, these drawbacks do not apply to next-generation sequencing (NGS) [5]. For example, NGS has advantages in the screening of targetable fusions that may trigger genomic events in 8–20% of advanced melanomas that lack distinctive driver mutations [19]. Similarly, NGS germline multigene panels including breast cancer gene 1 (BRCA1) and breast cancer gene 2 (BRCA2) genes can identify driver mutations and molecular targets to allow a personalized treatment of prostate cancer [20].
The present study aimed to compare PCR and in-house NGS companion diagnostic test results for gene alterations associated with predictive and prognostic outcomes in patients with mCRC.

2. Materials and Methods

2.1. Patients

In the present study, patients with mCRC were assessed between August 2017 and June 2019 at a single institution, and 50 patients were enrolled initially. The final sample comprised 41 patients with mCRC, after 9 patients who had insufficient tumor samples for both PCR and NGS analyses were excluded. The Consolidated Standards of Reporting Trials (CONSORT) diagram is shown in Figure 1.
Tumor, node, and metastasis (TNM) staging was performed per the eighth edition of the Cancer Staging Manual of the American Joint Commission on Cancer and the Union for International Cancer Control [21]. The patient data that were collected comprised age, sex, tumor location, TNM classification, presence of lymphovascular invasion, and presence of perineural invasion.
The genetic profiles of the 41 patients (including their KRAS, NRAS, and BRAF) were accessed before a suitable regimen was selected for each patient. Patients with the RAS wild-type status were prescribed first-line regimens that were reimbursed by Taiwan’s government; the regimens were selected after discussions between the physician and patients, regarding available medical insurance reimbursement schemes. The regimens were as follows: (1) irinotecan plus fluorouracil/leucovorin (FOLFIRI) with bevacizumab, (2) FOLFIRI with cetuximab, and (3) FOLFOX with panitumumab. For patients with RAS mutations, FOLFIRI with bevacizumab was used as the first-line regimen.
The evaluation of tumor response was performed per the Response Evaluation Criteria in Solid Tumors Version 1.1 guidelines [22,23] and typically conducted after every sixth cycle. When a BRAF mutation was detected, the treatment response was evaluated after the first four to six cycles of first-line therapy; when patients exhibited poor treatment response, their regimens were switched. Radiotherapy was administered to patients with a clinical diagnosis of mCRC with cT3–cT4, and/or clinical nodal stage N1–N2 rectal cancer if they were eligible for the treatment.

2.2. Ethics Approval

The present study was approved by the Ethical and Research Committee of Kaohsiung Medical University Hospital (approval no.: KMUHIRB-G(I)-20180049; obtained on 17 May 2019). The protocol was conducted per the Good Clinical Practice guidelines and institutional review board regulations that conformed to the 1983 revision of the Helsinki Declaration of 1975.

2.3. Analyses of KRAS, NRAS, BRAF

DNA was extracted by the QIAamp DNA Mini Kit (Qiagen, Hilden, Germany) according to the manufacturer’s instructions. All mutations are detected by using Sanger sequencing. The program for the PCR amplification in KRAS, NRAS, and BRAF involves 5 min of initial denaturation at 94 °C, 40 cycles of amplification consisting of 30 s at 94 °C, 30 s at 58 °C, and 30 s at 72 °C, with a final additional elongation at 72 °C for 7 min. PCR was performed using a LightCycler 480 (Roche Diagnostics, Wien, Austria). After amplification, the products were purified and directly sequenced by using the BigDye Terminator V.3.1 chemistries (Applied Biosystems, Foster City, CA, USA). Sequences were run on an ABI3130XL automated sequencer (Applied Biosystems). The primers used are listed in Supplementary Table S1.

2.4. Sample Preparation for Genotyping of KRAS, NRAS, and BRAF Hotspot Mutations through In-House NGS

Tumor samples were obtained from the tissue sections of optimal cutting temperature compound (OCT compound)-embedded frozen tissue samples. DNA was extracted using a QI-Aamp DNA Mini Kit (Qiagen, Hilden, Germany). DNA quantification was performed using a Qubit dsDNA HS Assay Kit (Invitrogen, Waltham, MA, USA).

2.5. Detection of KRAS, NRAS, and BRAF Hotspot Mutations through In-House NGS

The detection of hotspot mutations was performed using an AmpliSeq Assay Kit. DNA was amplified using customized primers (IDT) covering hotspot mutations for KRAS codons 12, 13, 61, and 146; NRAS codons 12, 13, 59, 61, 117, and 146; and BRAF Val600Glu. The libraries were amplified using a KAPA HiFi HotStart ReadyMixPCR Kit (Roche) and Nextera XT Index Primers (Illumina, San Diego, CA, USA), and subsequently purified using Agencourt AMPure XP (Beckman). Sequencing was performed using a MiSeq System and MiSeq Reagent Kit v3 (Illumina). All library preparations were performed at the translational core facility of Taipei Medical University. After sequencing was completed, read files (fastq) were mapped to the Hg19 reference using BWA [24]. The BAM files of all samples were sorted and then converted to the mpileup file format by using SAMtools [25]. Subsequently, the hotspot mutations were called using Varscan (v2.4.3) [26] and the following criteria were applied: minimal coverage > 200× and minimal variant frequency > 5%. The amplicon sequence of the mutational hotspots in KRAS, NRAS, and BRAF are listed in Supplementary Table S2. The figures that display the NGS results were visualized using Integrative Genomic Viewer (IGV) (version 2.12.2; © 2013–2021 Broad Institute and the Regents of the University of California).

2.6. Statistical Analysis

Analyses were performed using Microsoft Excel 2010. Continuous variables were ex-pressed as medians and interquartile ranges (IQRs) and analyzed using a one-way analysis of variance.

3. Results

3.1. Patient Demographics

The median (IQR) age of the 41 patients with mCRC in the present study was 65 (34−88) years and their male–female ratio was 1.73. The left side of the colon (defined as the area from the splenic flexure to the sigmoid colon) was the most common tumor site (85.3%). Vascular and perineural invasions were present in 10 (24.4%) and 12 (29.3%) patients, respectively (Table 1).

Comparison of PCR and NGS for Testing of Targeted Genes

Table 2 presents a comparison of the PCR and NGS genetic analysis results obtained from 41 patients with mCRC. The Kirsten rat sarcoma virus (KRAS), neuroblastoma RAS viral oncogene homolog (NRAS), and BRAF genes were subjected to both PCR and NGS tests. The discrepancies between the PCR and NGS results (Table 2) are presented in Supplementary Figures S1–S15. Table 3 reveals the high concordance (87.8–92.7%) between the detection results (KRAS, NRAS, and BRAF mutations) obtained through NGS and those obtained through PCR. Among the 41 patients with mCRC, three KRAS mutations were detected by PCR but not by NGS (Table 3). Similarly, among 39 patients with mCRC who were tested for NRAS mutations, three NRAS mutations were detected by NGS but not by PCR (Table 3). BRAF mutations in five patients were detected by NGS but not by PCR (Table 3). Table 4 summarizes the KRAS, NRAS, and BRAF mutations detected by both PCR and NGS in the present study. Examples of all gene mutations presented in Table 4 are provided in Supplementary Figures S16–S21.

4. Discussion

We compared PCR and in-house NGS test results for KRAS, NRAS, and BRAF mutation detection in patients with mCRC. Overall, the concordance between the PCR and in-house NGS results was high. Only 15 discrepant results were detected between PCR and in-house NGS (Table 2). Three variants of the KRAS gene were detected only by PCR (Table 3). Three variants of the NRAS gene and five variants of the BRAF gene were detected only by NGS (Table 3). This suggests that NGS is more sensitive to detecting genetic mutations than real-time PCR is.
Multistep processes associated with histological, morphological, and genetic modifications that emerge with age contribute to the development of CRC [27]. Activated RAS–RAF–MAPK and PI3K–PTEN–AKT signaling pathways play a crucial role in modulating cell proliferation, angiogenesis, cell motility, and apoptosis [28,29]. The cumulative mutations in tumor suppressor genes and proto-oncogenes (including KRAS, NRAS, BRAF, and PIK3CA) in RAS–RAF–MAPK and PI3K–PTEN–AKT signaling pathways lead to the development of CRC [30,31].
Cetuximab and panitumumab, which are monoclonal antibodies against EGFR, have been used since 2004 to treat mCRC [32]. KRAS mutation is present in nearly 30–45% of CRC tumors, and mutant KRAS is associated with resistance to anti-EGFR therapies [33,34,35,36]. KRAS codons 12 and 13 are the two most common hotspots, accounting for approximately 95% of all KRAS mutation types (approximately 80% and 15% of such mutations occur in codons 12 and 13, respectively) [34,35]. Patients with KRAS-mutated metastatic colorectal tumors are resistant to cetuximab and have shorter progression-free survival and overall survival durations when compared with patients without such mutations [37,38,39].
The downstream effectors of the EGFR signaling pathway include BRAF and NRAS, and the components of the PI3K signaling pathway can trigger negative responses to anti-EGFR treatments [33,40]. Notably, right-sided CRC has a significantly poorer prognosis relative to left-sided CRC, and it is often unresponsive to EGFR inhibition [41]. Compared with left-sided mCRC, right-sided mCRC with microsatellite stability is more likely to present either a hotspot RAS mutation or BRAF V600E (80% for right-sided mRC vs. 46% for left-sided mCRC [41,42]. Conversely, mutations in Adenomatous polyposis coli (APC) and tumor protein p53 (TP53), as well as amplifications of Erb-B2 Receptor Tyrosine Kinase 2 (ERBB2) and EGFR, are more frequently observed in left-sided CRC [43]. The expressions of EGFR ligands epiregulin (EREG) and amphiregulin (AREG) are also significantly higher in left-sided CRC [43].
Another member of the RAS family is NRAS, which has effector-binding domains that are identical to those in KRAS and yield similar mutational effects [44,45]. The Panitumumab Randomized Trial in Combination with Chemotherapy for Metastatic Colorectal Cancer to Determine Efficacy (PRIME) trial reported the detrimental effect of adding panitumumab to first-line FOLFOX in patients with RAS mutations [10]. Patients with NRAS mutations respond poorly and are resistant to anti-EGFR monoclonal antibody therapy [16].
Studies have shown that the BRAF mutation is associated with aggressive tumors and poor prognosis in patients with mCRC [15,16,17,18]. BRAF mutations are present almost exclusively in wild-type KRAS CRC, and they are present in 8.1% of patients with mCRC [36,46]. Among the eight mCRC patients with the BRAF codon 600 mutation in the present study, all except one patient presented KRAS wild-type results. The V600E mutation of BRAF represents a significant, independent, and negative prognostic factor for mCRC [36,47].
BRAF may be a negative prognostic factor for patients who have CRC and have received hepatic or pulmonary metastasectomy [48]. Poor efficacy of BRAF inhibitors in monotherapy for patients with mCRC and BRAF mutation was reported [49]. Therefore, detecting the BRAF mutation can help to predict poor treatment response. Evaluations of treatment response in patients with BRAF-mutated CRC are conducted after the first four to six cycles of first-line therapy. When a poor response is noted in patients with BRAF-mutated CRC, a second-line regimen is implemented.
In the present study, the genetic profiles of patients with mCRC aided the personalization of regimens to enhance treatment efficacy and minimize treatment toxicity, which led to improved overall and disease-free survival among patients with CRC. Based on our experience in the clinical setting, initial treatment considerations for patients with mCRC are generally determined by first testing the RAS gene status of such patients [36]. We published results in February 2022 on the personalized regimens of patients with wild-type RAS mCRC, and we reported that they could tolerate escalated doses of irinotecan based on the results of UGT1A1 testing and potentially achieve a more favorable clinical outcome without a significantly increased exposure to toxicity [50].
The NGS-based companion diagnostic test conducted in the present study analyzed KRAS, NRAS, and BRAF together as a panel of CRC molecular targets; therefore, it has the advantages of requiring fewer specimens and being more time and cost efficient in determining the optimal regimen for each patient. Implementing individualized treatment plans for patients with mCRC on the basis of their molecular testing results is essential for achieving optimal clinical outcomes.
The discrepancies between PCR and NGS results may be due to the low tumor content of the specimens and prolonged nucleic acid conversion, which may have led to false negative PCR results. The false negative rates of NGS in the discrepant results may have been influenced by pipeline parameters and read coverage [51]. Discrepancies can be confirmed using Sanger sequencing, but this was not performed in the present study and represents one of the study’s limitations. However, the discrepant results were generated from multiple mutation sites, indicating the absence of bias for any mutation.

5. Conclusions

Our results suggest that NGS, which produced results that shared high concordance with those produced by PCR, can be used to investigate the status of popular druggable genes associated with CRC and be expanded to include more genes. Given the limited sample size and data (collected from a single institution) used in the present study, future studies could conduct similar research using larger sample sizes.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cimb44040106/s1, Figures S1–S21: Experimental data on decrepencies are displayed in Figures S1–S15; Example of various gene mutations detected by NGS visualized by Integrative Genomic Viewer (IGV) are displayed in Figures S16–S20; Electropherogram examples of various gene mutations are displayed in Figure S21. Table S1: The primers used in PCR amplification of KRAS, NRAS, and BRAF, Table S2: Amplicon sequence of mutational hotspots in KRAS, NRAS, and BRAF.

Author Contributions

Conceptualization, W.-C.S. and J.-Y.W.; methodology, W.-C.S., Y.-C.T., Y.-R.L. and T.-H.H.; software, Y.-C.T.; Y.-R.L. and T.-H.H., validation, T.-K.C. and T.-C.Y.; formal analysis, H.-L.T. and C.-W.H.; investigation, Y.-C.C. and C.-C.L.; resources, P.-J.C.; data curation, W.-C.S. and Y.-C.T.; writing—original draft preparation, W.-C.S. and Y.-C.T.; writing—review and editing, J.-Y.W.; visualization, W.-C.S. and Y.-C.T.; supervision, J.-Y.W.; project administration, W.-C.S. and J.-Y.W.; funding acquisition, J.-Y.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants through funding from the Ministry of Science and Technology (MOST 109-2314-B-037-035, MOST 109-2314-B-037-040, MOST 109-2314-B-037-046-MY3, MOST110-2314-B-037-097) and the Ministry of Health and Welfare (MOHW109-TDU-B-212-134026, MOHW109-TDU-B-212-114006, MOHW110-TDU-B-212-1140026), and funded by the health and welfare surcharge on tobacco products, and the Kaohsiung Medical University Hospital (KMUH110-0R37, KMUH110-0R38, KMUH110-0M34, KMUH110-0M35, KMUH110-0M36, KMUH109-9M32, KMUH108-8M35, KMUH106-6M30, KMUH105-5M21, KMUH104-4M25, KMUHSA11013, KMUH-DK(C)110010, KMUH-DK(B)110004-3), KMU Center for Cancer Research (KMU-TC111A04-1), KMU Center for Liquid Biopsy and Cohort Research Center Grant (KMU-TC109B05), and KMU Office for Industry–Academic Collaboration (S109036), Kaohsiung Medical University. In addition, this study was supported by the Grant of Taiwan Precision Medicine Initiative, Academia Sinica, Taiwan, R.O.C.

Institutional Review Board Statement

The study was conducted in accordance with the guidelines of the Declaration of Helsinki, and approved by the Institutional Review Board of Kaohsiung Medical University Hospital (KMU-HIRB-G(I)-20180049; approval was obtained on 17 May 2019).

Informed Consent Statement

Written informed consent was obtained from all patients, who consented to their specimens being collected, used for scientific research purposes, and added to a specimen bank. The anonymization of patient information was completed prior to the analysis.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

The authors thank the members of the Joint Biobank of Office of Human Research at Taipei Medical University for their assistance with the NGS analysis.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cancer. Available online: http://www.who.int/news-room/fact-sheets/detail/cancer (accessed on 21 May 2018).
  2. 2019 Causes of Death Statistics. Available online: https://www.mohw.gov.tw/lp-4964-2.html (accessed on 21 May 2021).
  3. Patil, H.; Saxena, S.G.; Barrow, C.J.; Kanwar, J.R.; Kapat, A.; Kanwar, R.K. Chasing the personalized medicine dream through biomarker validation in colorectal cancer. Drug Discov. Today 2017, 22, 111–119. [Google Scholar] [CrossRef]
  4. Tsang, A.H.-F.; Cheng, K.H.; Wong, A.S.; Ng, S.S.; Ma, B.B.; Chan, C.M.; Tsui, N.B.; Chan, L.W.; Yung, B.Y.; Wong, S.C. Current and future molecular diagnostics in colorectal cancer and colorectal adenoma. World J. Gastroenterol. 2014, 20, 3847–3857. [Google Scholar] [CrossRef] [PubMed]
  5. Nong, L.; Zhang, Z.; Xiong, Y.; Zheng, Y.; Li, X.; Li, D.; He, Q.; Li, T. Comparison of next-generation sequencing and immunohistochemistry analysis for targeted therapy-related genomic status in lung cancer patients. J. Thorac. Dis. 2019, 11, 4992–5003. [Google Scholar] [CrossRef] [PubMed]
  6. Taieb, J.; Jung, A.; Sartore-Bianchi, A.; Peeters, M.; Seligmann, J.; Zaanan, A.; Burdon, P.; Montagut, C.; Laurent-Puig, P. The Evolving Biomarker Landscape for Treatment Selection in Metastatic Colorectal Cancer. Drugs 2019, 79, 1375–1394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Van Cutsem, E.; Cervantes, A.; Adam, R.; Sobrero, A.; van Krieken, J.H.; Aderka, D.; Aguilar, E.A.; Bardelli, A.; Benson, A.; Bodoky, G.; et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann. Oncol. 2016, 27, 1386–1422. [Google Scholar] [CrossRef]
  8. Shen, Y.; Wang, J.; Han, X.; Yang, H.; Wang, S.; Lin, N.; Shi, Y. Effectors of Epidermal Growth Factor Receptor Pathway: The Genetic Profiling of KRAS, BRAF, PIK3CA, NRAS Mutations in Colorectal Cancer Characteristics and Personalized Medicine. PLoS ONE 2013, 8, e81628. [Google Scholar] [CrossRef]
  9. Peeters, M.; Price, T.; Cervantes, A.; Sobrero, A.F.; Ducreux, M.; Hotko, Y.; André, T.; Chan, E.; Lordick, F.; Punt, C.J.; et al. Randomized Phase III Study of Panitumumab With Fluorouracil, Leucovorin, and Irinotecan (FOLFIRI) Compared With FOLFIRI Alone As Second-Line Treatment in Patients With Metastatic Colorectal Cancer. J. Clin. Oncol. 2010, 28, 4706–4713. [Google Scholar] [CrossRef]
  10. Douillard, J.-Y.; Oliner, K.S.; Siena, S.; Tabernero, J.; Burkes, R.; Barugel, M.; Humblet, Y.; Bodoky, G.; Cunningham, D.; Jassem, J.; et al. Panitumumab–FOLFOX4 Treatment and RAS Mutations in Colorectal Cancer. N. Engl. J. Med. 2013, 369, 1023–1034. [Google Scholar] [CrossRef] [Green Version]
  11. Amado, R.G.; Wolf, M.; Peeters, M.; Van Cutsem, E.; Siena, S.; Freeman, D.J.; Juan, T.; Sikorski, R.; Suggs, S.; Radinsky, R.; et al. Wild-TypeKRASIs Required for Panitumumab Efficacy in Patients With Metastatic Colorectal Cancer. J. Clin. Oncol. 2008, 26, 1626–1634. [Google Scholar] [CrossRef]
  12. Karapetis, C.S.; Khambata-Ford, S.; Jonker, D.J.; O’Callaghan, C.J.; Tu, D.; Tebbutt, N.C.; Simes, R.J.; Chalchal, H.; Shapiro, J.D.; Robitaille, S.; et al. K-rasMutations and Benefit from Cetuximab in Advanced Colorectal Cancer. N. Engl. J. Med. 2008, 359, 1757–1765. [Google Scholar] [CrossRef] [Green Version]
  13. Sorich, M.J.; Wiese, M.; Rowland, A.; Kichenadasse, G.; McKinnon, R.; Karapetis, C. Extended RAS mutations and anti-EGFR monoclonal antibody survival benefit in metastatic colorectal cancer: A meta-analysis of randomized, controlled trials. Ann. Oncol. 2014, 26, 13–21. [Google Scholar] [CrossRef] [PubMed]
  14. Bokemeyer, C.; Bondarenko, I.; Makhson, A.; Hartmann, J.T.; Aparicio, J.; de Braud, F.; Donea, S.; Ludwig, H.; Schuch, G.; Stroh, C.; et al. Fluorouracil, Leucovorin, and Oxaliplatin With and Without Cetuximab in the First-Line Treatment of Metastatic Colorectal Cancer. J. Clin. Oncol. 2009, 27, 663–671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Laurent-Puig, P.; Cayre, A.; Manceau, G.; Buc, E.; Bachet, J.-B.; LeComte, T.; Rougier, P.; Lievre, A.; Landi, B.; Boige, V.; et al. Analysis of PTEN, BRAF, and EGFR Status in Determining Benefit From Cetuximab Therapy in Wild-Type KRAS Metastatic Colon Cancer. J. Clin. Oncol. 2009, 27, 5924–5930. [Google Scholar] [CrossRef]
  16. De Roock, W.; Claes, B.; Bernasconi, D.; De Schutter, J.; Biesmans, B.; Fountzilas, G.; Kalogeras, K.T.; Kotoula, V.; Papamichael, D.; Laurent-Puig, P.; et al. Effects 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, 753–762. [Google Scholar] [CrossRef]
  17. Di Nicolantonio, F.; Martini, M.; Molinari, F.; Sartore-Bianchi, A.; Arena, S.; Saletti, P.; De Dosso, S.; Mazzucchelli, L.; Frattini, M.; Siena, S.; et al. Wild-Type BRAF Is Required for Response to Panitumumab or Cetuximab in Metastatic Colorectal Cancer. J. Clin. Oncol. 2008, 26, 5705–5712. [Google Scholar] [CrossRef] [PubMed]
  18. Hsieh, Y.-C.; Chang, T.-K.; Su, W.-C.; Huang, C.-W.; Tsai, H.-L.; Chen, Y.-C.; Li, C.-C.; Chen, P.-J.; Yin, T.-C.; Ma, C.-J.; et al. UGT1A1 Polymorphism for Irinotecan Dose Escalation in Patients with BRAF-Mutated Metastatic Colorectal Cancer Treated with First-Line Bevacizumab and FOLFIRI. J. Oncol. 2021, 2021, 6686517. [Google Scholar] [CrossRef]
  19. Revythis, A.; Shah, S.; Kutka, M.; Moschetta, M.; Ozturk, M.; Pappas-Gogos, G.; Ioannidou, E.; Sheriff, M.; Rassy, E.; Boussios, S. Unraveling the Wide Spectrum of Melanoma Biomarkers. Diagnostics 2021, 11, 1341. [Google Scholar] [CrossRef]
  20. Ghose, A.; Moschetta, M.; Pappas-Gogos, G.; Sheriff, M.; Boussios, S. Genetic Aberrations of DNA Repair Pathways in Prostate Cancer: Translation to the Clinic. Int. J. Mol. Sci. 2021, 22, 9783. [Google Scholar] [CrossRef]
  21. Amin, M.B.; Edge, S.; Greene, F.; Byrd, D.R.; Brookland, R.K.; Washington, M.K.; Gershenwald, J.E.; Compton, C.C.; Hess, K.R.; Sullivan, D.C. (Eds.) AJCC Cancer Staging Manual, 8th ed.; American College of Surgeons, Springer International Publishing: New York, NY, USA, 2017. [Google Scholar]
  22. Eisenhauer, E.A.; Therasse, P.; Bogaerts, J.; Schwartz, L.H.; Sargent, D.; Ford, R.; Dancey, J.; Arbuck, S.; Gwyther, S.; Mooney, M.; et al. New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur. J. Cancer 2009, 45, 228–247. [Google Scholar] [CrossRef] [PubMed]
  23. Tirkes, T.; Hollar, M.A.; Tann, M.; Kohli, M.D.; Akisik, F.; Sandrasegaran, K. Response Criteria in Oncologic Imaging: Review of Traditional and New Criteria. RadioGraphics 2013, 33, 1323–1341. [Google Scholar] [CrossRef]
  24. Li, H.; Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 2009, 25, 1754–1760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Li, H.; Handsaker, B.; Wysoker, A.; Fennell, T.; Ruan, J.; Homer, N.; Marth, G.; Abecasis, G.; Durbin, R. 1000 Genome Project Data Processing Subgroup. The Sequence Alignment/Map format and SAMtools. Bioinformatics 2009, 25, 2078–2079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Koboldt, D.C.; Larson, D.E.; Wilson, R.K. Using VarScan 2 for Germline Variant Calling and Somatic Mutation Detection. Curr. Protoc. Bioinform. 2013, 44, 15.4.1–15.4.17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Balchen, V.; Simon, K. Colorectal cancer development and advances in screening. Clin. Interv. Aging 2016, 11, 967–976. [Google Scholar] [CrossRef] [Green Version]
  28. Engelman, J.A. Targeting PI3K signalling in cancer: Opportunities, challenges and limitations. Nat. Cancer 2009, 9, 550–562. [Google Scholar] [CrossRef]
  29. Samuels, Y.; Diaz, L.; Schmidt-Kittler, O.; Cummins, J.M.; DeLong, L.; Cheong, I.; Rago, C.; Huso, D.L.; Lengauer, C.; Kinzler, K.W.; et al. Mutant PIK3CA promotes cell growth and invasion of human cancer cells. Cancer Cell 2005, 7, 561–573. [Google Scholar] [CrossRef] [Green Version]
  30. McCubrey, J.A.; Steelman, L.S.; Abrams, S.L.; Lee, J.T.; Chang, F.; Bertrand, F.E.; Navolanic, P.M.; Terrian, D.M.; Franklin, R.A.; D’Assoro, A.B.; et al. Roles of the RAF/MEK/ERK and PI3K/PTEN/AKT pathways in malignant transformation and drug resistance. Adv. Enzym. Regul. 2006, 46, 249–279. [Google Scholar] [CrossRef]
  31. Calistri, D.; Rengucci, C.; Seymour, I.; Lattuneddu, A.; Polifemo, A.M.; Monti, F.; Saragoni, L.; Amadori, D. Mutation analysis ofp53,K-ras, andBRAF genes in colorectal cancer progression. J. Cell. Physiol. 2005, 204, 484–488. [Google Scholar] [CrossRef]
  32. Zhang, J.; Zheng, J.; Yang, Y.; Lu, J.; Gao, J.; Lu, T.; Sun, J.; Jiang, H.; Zhu, Y.; Zheng, 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, 18678. [Google Scholar] [CrossRef]
  33. Wilson, P.M.; LaBonte, M.J.; Lenz, H.-J. Molecular Markers in the Treatment of Metastatic Colorectal Cancer. Cancer J. 2010, 16, 262–272. [Google Scholar] [CrossRef]
  34. Chuang, S.-C.; Huang, C.-W.; Chen, Y.-T.; Ma, C.-J.; Tsai, H.-L.; Chang, T.-K.; Su, W.-C.; Hsu, W.-H.; Kuo, C.-H.; Wang, J.-Y. Effect of KRAS and NRAS mutations on the prognosis of patients with synchronous metastatic colorectal cancer presenting with liver-only and lung-only metastases. Oncol. Lett. 2020, 20, 2119–2130. [Google Scholar] [CrossRef]
  35. Tan, C.; Du, X. KRAS mutation testing in metastatic colorectal cancer. World J. Gastroenterol. 2012, 18, 5171–5180. [Google Scholar] [CrossRef] [PubMed]
  36. Chen, H.-H.; Ke, T.-W.; Huang, C.-W.; Jiang, J.-K.; Chen, C.-C.; Hsieh, Y.-Y.; Teng, H.-W.; Lin, B.-W.; Liang, Y.-H.; Su, Y.-L.; et al. Taiwan Society of Colon and Rectal Surgeons Consensus on mCRC Treatment. Front. Oncol. 2021, 11, 764912. [Google Scholar] [CrossRef]
  37. Ma, C.-J.; Huang, C.-W.; Chang, T.-K.; Tsai, H.-L.; Su, W.-C.; Yeh, Y.-S.; Chen, P.-J.; Wang, J.-Y. Oncologic Outcomes in Metastatic Colorectal Cancer with Regorafenib with FOLFIRI as a Third- or Fourth-Line Setting. Transl. Oncol. 2018, 12, 502–512. [Google Scholar] [CrossRef] [PubMed]
  38. Yen, L.-C.; Yeh, Y.-S.; Chen, C.-W.; Wang, H.-M.; Tsai, H.-L.; Lu, C.-Y.; Chang, Y.-T.; Chu, K.-S.; Lin, S.-R.; Wang, J.-Y. Detection of KRAS Oncogene in Peripheral Blood as a Predictor of the Response to Cetuximab Plus Chemotherapy in Patients with Metastatic Colorectal Cancer. Clin. Cancer Res. 2009, 15, 4508–4513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. De Roock, W.; Piessevaux, H.; De Schutter, J.; Janssens, M.; De Hertogh, G.; Personeni, N.; Biesmans, B.; Van Laethem, J.-L.; Peeters, M.; Humblet, Y.; et al. KRAS wild-type state predicts survival and is associated to early radiological response in metastatic colorectal cancer treated with cetuximab. Ann. Oncol. 2008, 19, 508–515. [Google Scholar] [CrossRef] [PubMed]
  40. Yeh, J.-H.; Tsai, H.-L.; Chen, Y.-C.; Li, C.-C.; Huang, C.-W.; Chang, T.-K.; Su, W.-C.; Chen, P.-J.; Liu, Y.-P.; Wang, J.-Y. BRAF, MEK, and EGFR Triplet Inhibitors as Salvage Therapy in BRAF-Mutated Metastatic Colorectal Cancer—A Case Series Study Target Therapy of BRAF-Mutated mCRC. Medicina 2021, 57, 1339. [Google Scholar] [CrossRef] [PubMed]
  41. Boussios, S.; Ozturk, M.A.; Moschetta, M.; Karathanasi, A.; Zakynthinakis-Kyriakou, N.; Katsanos, K.H.; Christodoulou, D.K.; Pavlidis, N. The Developing Story of Predictive Biomarkers in Colorectal Cancer. J. Pers. Med. 2019, 9, 12. [Google Scholar] [CrossRef] [Green Version]
  42. Missiaglia, E.; Jacobs, B.; D’Ario, G.; Di Narzo, A.; Soneson, C.; Budinska, E.; Popovici, V.; Vecchione, L.; Gerster, S.; Yan, P.; et al. Distal and proximal colon cancers differ in terms of molecular, pathological, and clinical features. Ann. Oncol. 2014, 25, 1995–2001. [Google Scholar] [CrossRef]
  43. Zarkavelis, G.; Boussios, S.; Papadaki, A.; Katsanos, K.H.; Christodoulou, D.K.; Pentheroudakis, G. Current and future biomarkers in colorectal cancer. Ann. Gastroenterol. 2017, 30, 613–621. [Google Scholar] [CrossRef] [PubMed]
  44. Takane, K.; Akagi, K.; Fukuyo, M.; Yagi, K.; Takayama, T.; Kaneda, A. DNA methylation epigenotype and clinical features ofNRAS-mutation(+) colorectal cancer. Cancer Med. 2017, 6, 1023–1035. [Google Scholar] [CrossRef] [PubMed]
  45. Downward, J. Targeting RAS signalling pathways in cancer therapy. Nat. Cancer 2003, 3, 11–22. [Google Scholar] [CrossRef] [PubMed]
  46. Peeters, M.; Kafatos, G.; Taylor, A.; Gastanaga, V.; Oliner, K.; Hechmati, G.; Terwey, J.-H.; van Krieken, J. Prevalence of RAS mutations and individual variation patterns among patients with metastatic colorectal cancer: A pooled analysis of randomised controlled trials. Eur. J. Cancer 2015, 51, 1704–1713. [Google Scholar] [CrossRef] [PubMed]
  47. Lai, E.; Pretta, A.; Impera, V.; Mariani, S.; Giampieri, R.; Casula, L.; Pusceddu, V.; Coni, P.; Fanni, D.; Puzzoni, M.; et al. BRAF-mutant colorectal cancer, a different breed evolving. Expert Rev. Mol. Diagn. 2018, 18, 499–512. [Google Scholar] [CrossRef]
  48. Afrăsânie, V.-A.; Marinca, M.V.; Alexa-Stratulat, T.; Gafton, B.; Păduraru, M.; Adavidoaiei, A.M.; Miron, L.; Rusu, C. KRAS, NRAS, BRAF, HER2 and microsatellite instability in metastatic colorectal cancer—Practical implications for the clinician. Radiol. Oncol. 2019, 53, 265–274. [Google Scholar] [CrossRef] [Green Version]
  49. Sanz-Garcia, E.; Argiles, G.; Elez, E.; Tabernero, J. BRAF mutant colorectal cancer: Prognosis, treatment, and new perspectives. Ann. Oncol. 2017, 28, 2648–2657. [Google Scholar] [CrossRef]
  50. Tsai, H.-L.; Chen, Y.-C.; Yin, T.-C.; Su, W.-C.; Chen, P.-J.; Chang, T.-K.; Li, C.-C.; Huang, C.-W.; Wang, J.-Y. Comparison of UGT1A1 polymorphism as guidance of irinotecan dose escalation in RAS wild type metastatic colorectal cancer patients treated with cetuximab or bevacizumab plus FOLFIRI as the first-line therapy. Oncol. Res. Featur. Preclin. Clin. Cancer Ther. 2022. [Google Scholar] [CrossRef]
  51. Bobo, D.; Lipatov, M.; Rodriguez-Flores, J.L.; Auton, A.; Henn, B.M. False Negatives Are a Significant Feature of Next Generation Sequencing Callsets. BiorXiv 2016. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The Consolidated Standards of Reporting Trials (CONSORT) diagram of the present study.
Figure 1. The Consolidated Standards of Reporting Trials (CONSORT) diagram of the present study.
Cimb 44 00106 g001
Table 1. Demographics of study patients (n = 41).
Table 1. Demographics of study patients (n = 41).
Subjects with Metastatic CRC (n = 41)
Median age (years, range)65 (34–88)
Male 64 (34–86)
Female65 (38–88)
Gender
Male 26 (63.4%)
Female15 (36.6%)
Tumor location
R’t colon/L’t colon6 (14.6%)/35 (85.3%)
Depth of tumor invasion
T1/T2/T3/T40/2 (4.9%)/27 (65.8%)/12 (29.3%)
Lymph node involvement
N0/N1/N2 11 (26.8%)/17 (41.5%)/13 (31.7%)
Vascular invasion
Yes/No10 (24.4%)/31 (75.6%)
Perineural invasion
Yes/No12 (29.3%)/29 (70.7%)
CRC, Colorectal cancer; R’t colon, right-sided colon was defined as the region from the cecum to the splenic flexure; L’t colon, left-sided colon was defined as the region from the splenic flexure to the rectum.
Table 2. Comparison of PCR versus NGS for testing of targeted genes in patients with mCRC (n = 41).
Table 2. Comparison of PCR versus NGS for testing of targeted genes in patients with mCRC (n = 41).
Somatic Mutation
Case No.Tumor
Content (%)
KRAS Condon 12/13/59/61/146
Tested
by
PCR
KRAS Condon 12/13/61/146
Tested
by
NGS
NRAS Condon 12/13
Tested
by
PCR
NRAS Condon 12/13
Tested
by
NGS
BRAF Codon 600
Tested by
PCR
BRAF Codon 600
Tested by
NGS
130wild-typewild-typewild-typewild-typewild-typewild-type
2<5codon 61 mutationwild-typewild-typewild-typewild-typewild-type
390codon 12 mutationcodon 12 mutationno data *wild-typewild-typewild-type
415wild-typewild-typewild-typewild-typewild-typewild-type
570wild-typewild-typewild-typewild-typewild-typewild-type
650wild-typewild-typewild-typewild-typewild-typewild-type
710wild-typecodon 61 mutationwild-typewild-typewild-typewild-type
820codon 12 mutationcodon 12 mutationno data *codon 12 mutation wild-typewild-type
930wild-typewild-typewild-typewild-typewild-typewild-type
10<5wild-typewild-typewild-typewild-typewild-typewild-type
11<5wild-typewild-typewild-typewild-typewild-typewild-type
1220wild-typewild-typewild-typewild-typecodon 597 mutationcodon 600 mutation
1370codon 59 mutationcodon 13 mutationwild-typewild-typewild-typecodon 600 mutation
1440codon 146 mutationcodon 146 mutationwild-typewild-typewild-typewild-type
15<5wild-typewild-typewild-typewild-typewild-typewild-type
1610wild-typewild-typewild-typecodon 12 mutationwild-typecodon 600 mutation
1760wild-typewild-typewild-typecodon 12 mutationwild-typecodon 600 mutation
1820codon 61 mutationcodon 61 mutation wild-typewild-typewild-typewild-type
1960wild-typewild-typewild-typecodon 12 mutationcodon 600 mutationcodon 600 mutation
2030wild-typewild-typecodon 12 mutationcodon 12 mutationwild-typecodon 600 mutation
2115wild-typewild-typewild-typewild-typecodon 600 mutationcodon 600 mutation
2210wild-typewild-typewild-typewild-typewild-typecodon 600 mutation
2310codon 12 mutationcodon 12 mutationwild-typewild-typewild-typewild-type
2440wild-typewild-typewild-typewild-typewild-typewild-type
2510wild-typewild-typewild-typewild-typewild-typewild-type
2630codon 12 mutationcodon 12 mutationwild-typewild-typewild-typewild-type
2720wild-typewild-typewild-typewild-typewild-typewild-type
2840wild-typewild-typewild-typewild-typewild-typewild-type
2980wild-typewild-typewild-typewild-typewild-typewild-type
3010codon 13 mutationwild-typewild-typewild-typewild-typewild-type
31<5codon 12 mutationwild-typewild-typewild-typewild-typewild-type
3295codon 13 mutationcodon 13 mutationwild-typewild-typewild-typewild-type
33<5wild-typewild-typewild-typewild-typewild-typewild-type
34<5wild-typewild-typewild-typewild-typewild-typewild-type
3520codon 61 mutationwild-typewild-typewild-typewild-typewild-type
3650wild-typewild-typewild-typewild-typewild-typewild-type
3780codon 146 mutationcodon 146 mutationwild-typewild-typewild-typewild-type
3830wild-typewild-typewild-typewild-typewild-typewild-type
3930wild-typewild-typewild-typewild-typewild-typewild-type
4095codon 12 mutationcodon 12 mutationwild-typewild-typewild-typewild-type
4140wild-typewild-typewild-typewild-typewild-typewild-type
Bolded terms indicate inconsistencies in results between PCR and NGS results. * Two cases had no PCR data on NRAS.
Table 3. Detection of KRAS, NRAS, and BRAF mutation by PCR versus NGS in patients with mCRC.
Table 3. Detection of KRAS, NRAS, and BRAF mutation by PCR versus NGS in patients with mCRC.
GeneNo. of Cases ComparedWild-Type
Detected by PCR
Wild-Type
Detected by NGS
Mutation
Detected by PCR
Mutation
Detected by NGS
Percentage of
Concordance (%)
KRAS412730141192.7
NRAS *3938351492.3
BRAF4138333887.8
* Two cases had no PCR data on NRAS.
Table 4. KRAS, NRAS, and BRAF mutations detected by PCR and NGS in patients with mCRC (n = 41).
Table 4. KRAS, NRAS, and BRAF mutations detected by PCR and NGS in patients with mCRC (n = 41).
No. of Patients with mCRC
GeneSourceMutationReal-Time PCRNGS
KRASTissue DNACodon 126 (14.6%)5 (12.2%)
Codon 132 (4.9%)2 (4.9%)
Codon 591 (2.4%)0
Codon 613 (7.3%)2 (4.9%)
Codon 1462 (4.9%)2 (4.9%)
Total 14 (34.1%)11 (26.8%)
NRASTissue DNACodon 121 (2.4%)5 (12.2%)
Total 1 (2.4%)5 (12.2%)
BRAFTissue DNACodon 600
Codon 597
2 (4.9%)
1 (2.4%)
8 (19.5%)
0
Total 3 (7.3%)8 (19.5%)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Su, W.-C.; Tsai, Y.-C.; Tsai, H.-L.; Chang, T.-K.; Yin, T.-C.; Huang, C.-W.; Chen, Y.-C.; Li, C.-C.; Chen, P.-J.; Liu, Y.-R.; et al. Comparison of Next-Generation Sequencing and Polymerase Chain Reaction for Personalized Treatment-Related Genomic Status in Patients with Metastatic Colorectal Cancer. Curr. Issues Mol. Biol. 2022, 44, 1552-1563. https://doi.org/10.3390/cimb44040106

AMA Style

Su W-C, Tsai Y-C, Tsai H-L, Chang T-K, Yin T-C, Huang C-W, Chen Y-C, Li C-C, Chen P-J, Liu Y-R, et al. Comparison of Next-Generation Sequencing and Polymerase Chain Reaction for Personalized Treatment-Related Genomic Status in Patients with Metastatic Colorectal Cancer. Current Issues in Molecular Biology. 2022; 44(4):1552-1563. https://doi.org/10.3390/cimb44040106

Chicago/Turabian Style

Su, Wei-Chih, Yi-Chen Tsai, Hsiang-Lin Tsai, Tsung-Kun Chang, Tzu-Chieh Yin, Ching-Wen Huang, Yen-Cheng Chen, Ching-Chun Li, Po-Jung Chen, Yun-Ru Liu, and et al. 2022. "Comparison of Next-Generation Sequencing and Polymerase Chain Reaction for Personalized Treatment-Related Genomic Status in Patients with Metastatic Colorectal Cancer" Current Issues in Molecular Biology 44, no. 4: 1552-1563. https://doi.org/10.3390/cimb44040106

Article Metrics

Back to TopTop