Skip to main content
PLOS One logoLink to PLOS One
. 2023 Oct 5;18(10):e0292087. doi: 10.1371/journal.pone.0292087

Multigene panel next generation sequencing in metastatic colorectal cancer in an Australian population

Udit Nindra 1,2,3,4,5,*, Abhijit Pal 1,3,4, Vivienne Lea 6, Stephanie Hui-Su Lim 2,4,5, Kate Wilkinson 1, Ray Asghari 3, Tara L Roberts 4,5,7, Therese M Becker 4,5,7, Mahtab Farzin 6, Tristan Rutland 5,6, Mark Lee 7, Scott MacKenzie 5, Weng Ng 1,4,5,7, Bin Wang 6, C Soon Lee 3,5,6,7,, Wei Chua 1,4,5,7,
Editor: Avaniyapuram Kannan Murugan8
PMCID: PMC10553362  PMID: 37796807

Abstract

Background

Next generation sequencing (NGS) is increasingly used in standard clinical practice to identify patients with potentially actionable mutations. Stratification of NGS mutation tiers is currently based on the European Society of Medical Oncology (ESMO) Scale for Clinical Actionability of Molecular Targets (ESCAT[E]) Tier I–V & X. Allele frequency is also increasingly recognised as an important prognostic tool in advanced cancer. The aim of this study was to determine the genomic mutations in metastatic colorectal cancer (CRC) in an Australian multicultural population and their influence on survival outcomes.

Methods

Next generation sequencing with the 50-gene panel Oncomine Precision Assay was used on 180 CRC tissue samples obtained across six Sydney hospitals between June 2021 and March 2022.

Results

From 180 samples, 147 (82%) had at least one gene mutation identified with 68 (38%) having two or more concurrent mutations. Tier I variants included RAS wild-type [EI] in 73 (41%) and BRAF V600E [EIA] in 27 (15%). Non-tier I variants include 2 (1%) ERBB2 amplification [EIIB], 26 (15%) PIK3CA hotspot mutations [EIIIA] and 9 (5%) MET focal amplifications [EIIIA]. NGS testing revealed an additional 22% of cases with Tier II & III mutations. 43% of patients also presented with potentially actionable Tier III & IV mutations. Patients with concurrent TP53 and RAS mutations had significantly reduced overall survival (6.1 months versus 21.1 months, p <0.01). High KRAS allele frequency, as defined by those with over 20% variant allele frequency (VAF), also demonstrated reduced overall survival (12.1 months versus 42.9 months, p = 0.04).

Conclusions

In addition to identifying patients with genomic alterations suitable for clinically proven standard of care therapeutic options, the 50 gene NGS panel has significant potential in identifying potentially actionable non-tier 1 mutations and therefore may become future standard clinical practice.

1. Introduction

Colorectal Cancer (CRC) continues to remain a significant cause of cancer death worldwide [1]. Initial treatment of metastatic CRC has been largely reliant on chemotherapy and targeted therapy but recent advances in technology have introduced the role of genomic testing to identify druggable targets, either established or currently being tested in clinical trial settings. One example of actionable and targetable mutations that has influenced treatment of CRC is the BRAF V600E mutation. Recent data has shown efficacy of mutant BRAF ATP-competitive RAF kinase inhibitors, such as encorafenib, in combination with cetuximab, a monoclonal antibody targeting the epidermal growth factor receptor (EGFR) receptor, for the treatment of BRAF V600E mutated CRC [2]. The ability to detect mutations in tumours not only provides clinicians and researchers with possible therapeutic targets but also provides an understanding of potential resistance mechanisms to particular treatments. For example, KRAS mutations in CRC are known to cause resistance to EGFR monoclonal antibody therapies such as cetuximab and panitumumab [3, 4]. Additionally, the presence of certain mutations can be associated with higher risk histological features at diagnosis. For example, PIK3CA hotspot mutations are reported to occur in approximately 16% of CRC (5). These mutations have been shown to correlate with poor histological grade, late clinical stage at presentation, worse prognosis and first line chemotherapy resistance [5].

Next Generation Sequencing (NGS) provides rapid testing of multiple genetic molecular variants to inform clinicians of therapeutic options. Targeted NGS technology relies on known mutational molecular variants with the ability to streamline detection for a panel of variants of clinical interest. NGS technology can identify hotspot mutations and copy number variant mutations in tumour DNA as well as fusion transcript variants in tumour RNA. This can lead to the ability to not only identify specific variant mutations of interest but also the variant allele frequency. Allele frequency is currently not used in clinical practice to influence treatment decisions but early evidence, especially in the EGFR mutated NSCLC setting, suggests it could play a role in predicting therapeutic efficacy [6, 7]. NGS testing in patients with stage IV CRC has been increasingly utilised to help determine the genetic profile of tumours and possible resistance mechanisms through concomitant mutations. NGS using the Oncomine Precision Assay provides a means of testing for mutations across 50 genes, namely: KRAS, NRTK, BRAF, NRAS, ERBB2, RET, MET, ROS1, PIK3CA, NRG1, AKT1, AKT2, AKT3, CDK4, CTNNB1, ERBB3, FGFR2, FGFR3, FLT3, GNA11, GNAQ, GNAS, HRAS, IDH1, IDH2, MAP2K1, MAP2K2, PDGFRA, PTEN, SMO, TP53, EGFR, ALK, AR, ARAF, RAF1, MTOR, KIT, FGFR, ERBB4, ESR1, FGFR1, CDKN2A and CHEK2. Although not all these mutations currently guide therapeutic options, emerging understanding of the roles of these genes and novel drug development may provide further information into prognostication of patients, treatment options, or predict therapy resistance.

Currently, there is no standardised method for classifying mutations identified with NGS technology into tiers of importance. The European Society of Medical Oncology (ESMO) Precision Medicine Working Group has proposed a set of guidelines, defining mutations from Tier 1 to Tier X actionability which are summarised in Table 1 [8]. Here we used the Oncomine Precision Assay for sequencing of stage IV CRC from large patient numbers to obtain frequencies of mutations in CRC for our real-world, multicultural Australian patient cohort, as stratified by the ESMO Precision Medicine Working Group.

Table 1. ESMO tiers of mutational actionability.

Tier Definition Evidence required
I Ready for routine clinical use. Access to treatment should be considered standard of care. I-A: Prospective RCT demonstrating improvement of survival end point
I-B: Prospective non-randomised trials demonstrating meaningful benefit
II Investigational mutation. The treatment is considered preferrable in context of evidence collected Retrospective studies demonstrate a clinically meaningful survival benefit or prospective studies demonstrate increase response without survival data available
III-IV Hypothetical Target Clinical benefit seen in patients but not in the same tumour stream.
V Mutation is associated with objective response but without clinically meaningful benefit Prospective studies show that targeted therapy is associated with objective responses but not improved survival
X Lack of evidence for actionability No evidence. The genomic alteration is therapeutically actionable.

2. Methods

2.1 NGS mutation analysis

Next generation sequencing was performed with the Ion Torrent Genexus integrated next generation sequencer using the Oncomine Precision Assay, which is 50-gene NGS panel that was introduced to our laboratory in June 2021. Only DNA hotspot mutations and copy number variations (CNVs) were assessed. RNA sequencing for fusions was not performed. The Ion Torrent Genexus System fully automates the specimen-to-report workflow. It automates all steps of the targeted NGS workflow starting from extraction, purification and quantification of nucleic acid. The Genexus Purification System replaces sample prep by extracting and quantifying nucleic acids. This is followed by automation of the library preparation (including DNA synthesis), template preparation and sequencing. Lastly, primary data analysis is also automated and variant reporting for DNA applications are performed using the Oncomine Reporter software, a Thermo Fisher supplied bioinformatics analytic tools. The reported results from the 50-gene Oncomine Precision Assay are filtered to provide a summary of the variants and copy number variation as well as the variant allele frequency. Historically, NGS testing using Oncomine panels from surgical samples has a reported success rate of approximately 80% for certain solid organ malignancies [9]. However, this accuracy is variable depending on the NGS system and software used.

2.2 Data collection and correlation

This is a retrospective observational cohort study utilising CRC patient NGS and clinical records. We included all patients with histologically confirmed metastatic CRC that had Oncomine NGS testing between June 2021 and March 2022. In total, records from 180 de-identified patients with CRC were available from 5 hospitals across greater Sydney, Australia. We collected patient demographic data including age at diagnosis, sex and ethnicity (as defined by country of birth) in addition to NGS mutation reports. Data regarding tumour differentiation and mismatch repair (MMR) status was collected where possible. The project was approved by the Sydney South West Local Health District Human Research Ethics Committee (2019/ETH04187). Survival outcomes were also collected with data censored as of the end of February 2023. We stratified identified mutations according to the ESMO guidelines as documented in Table 1. We correlated mutation frequency with clinicopathological variables and survival outcomes using cox-proportional hazard ratios and Kaplan-Meier curve analyses A p-value of 0.05 was defined as statistically significant. All data analysis was done using R 4.1.1.

3. Results

3.1 Clinicopathological characteristics and survival outcomes

One hundred and eighty patients with CRC were included in the analysis. The median age was 67 with 59% of the patients being male. All patients had histologically confirmed adenocarcinoma. Sixety-nine (38%) had moderately differentiated disease, with majority being unknown at the time of testing (50%). 86 (47%) had mismatch repair (MMR) testing completed with 7 (4%) demonstrating deficient MMR status. Baseline patient demographics are summarised in Table 2.

Table 2. Patient demographics.

N 180
Age (median, range) [years] 67 (22–89)
Sex
 Male 104 (58%)
 Female 76 (42%)
Region of Birth
 Australia / New Zealand 112 (62%)
 Pacific Islands 2 (1%)
 South East Asia 23 (13%)
 Eastern Europe 28 (16%)
 Middle East 11 (6%)
 Other 3 (2%)
 Unknown 1 (1%)
Differentiation
 Well differentiated 5 (3%)
 Moderately differentiated 69 (38%)
 Poorly differentiated 16 (9%)
 Unknown 90 (50%)
Mismatch Repair Status
 Proficient 79 (44%)
 Deficient 7 (4%)
 Unknown 93 (52%)

3.1.1 Genomic profiling for actionable mutations and potentially actionable targets

Out of 180 patients, 147 (82%) had at least one gene mutation identified via NGS with 68 (38%) having two or more mutations identified. Based on stratification of NGS mutation tiers as per the ESMO Precision Medicine Working Group (9), 105 (58%) patients had Tier 1 mutations, 59 (33%) of which were patients with tier 1 mutations alone. Overall, 33 (18%) of patients had no mutation identified. Tier I variants included RAS wild-type [EI] in 73 (41%) and BRAF V600E [EIA] in 27 (15%). Non-tier I included 2 (1%) ERBB2 amplification [EIIB], 26 (15%) PIK3CA hotspot mutations [EIIIA] and 9 (5%) MET focal amplifications [EIIIA]. These results are summarised in in Table 3 and Fig 1.

Table 3. Mutations identified using next generation sequencing as stratified by the European society of medical oncology scale for clinical actionability of molecular targets guidelines.
Mutation Tier Frequency
All RAS / BRAF WT 1 73 (41%)
BRAF V600E 1 27 (15%)
ERBB2 Amplifications 2 2 (1%)
MET Focal Amplifications 3 9 (5%)
PIK3CA 3 26 (15%)
AKT1 3 2 (1%)
Fig 1. Distribution of concurrent mutations identified by next generation sequencing in colorectal cancer.

Fig 1

Individual patients with concurrent mutations in each column. Tiers 1–3 as outlined by the European Society of Medical Oncology Scale for Clinical Actionability of Molecular Targets.

In our cohort, 71 (43%) had at least one of 22 potentially actionable Tier 3–4 mutations. We classified these 27 mutations as Tier 3–4 as there are currently active early phase clinical trials being run to investigate the therapeutic role of targeted agents against these mutations in either CRC or any solid organ malignancy. These 22 genes are summarised in Table 4. We also classified 12 genes as Tier X as these have no current published evidence suggesting that these genes play a role in tumourigenesis, treatment efficacy or resistance. Furthermore, there were no ongoing trials investigating the roles of these genes in CRC or any solid organ malignancy. These were AR, CDKN2A, ARAF, RAF1, MTOR, KIT, FGFR4, ERBB4, ERS1, FGFR1, GNAS and CHEK2.

Table 4. Potentially actionable Tier 3–4 mutations.
Mutation Frequency (CRC) CRC specific clinical trial Any solid organ malignancy clinical trial
AKT2 0 (0%) NCT01333475 -
AKT3 0 (0%) NCT01333475 -
BRAF (non-V600E) 2 (2%) NCT03087071 -
CDK4 0 (0%) - NCT01037790
CTNNB1 3 (2%) NCT04851119 NCT04851119
EGFR 1 (1%) NCT00326495 -
ERBB3 1 (1%) NCT04479436 -
FGFR2 0 (0%) NCT04096417 -
FGFR3 0 (0%) NCT04096417 -
FLT3 0 (0%) NCT01762293 -
GNA11 0 (0%) - NCT03947385
GNAQ 0 (0%) - NCT03947385
HRAS 0 (0%) NCT04853043 -
IDH1 0 (0%) - NCT04521686
IDH2 0 (0%) - NCT04521686
KRAS (non-G12C) 0 (0%) NCT03948763 NCT03948763
MAP2K1 1 (1%) NCT03087071 NCT04488003
MAP2K2 0 (0%) - NCT04488003
PDGFRA 0 (0%) - NCT03693326
PTEN 2 (3%) - NCT01458067
SMO 0 (0%) - NCT02002689
TP53 60 (33%) NCT03144804 NCT00393029

3.1.2 Concurrent mutations and their impact on overall survival

68 (38%) patients had 2 or more concurrent mutations and these have been summarised in Fig 2. Patients with RAS mutations had a median overall survival of 16.9 months from time of metastatic disease diagnosis. In patients with RAS mutated CRC, concurrent presence of TP53 mutations conferred significantly reduced overall survival (6.1 months versus 21.1 months, HR 4.2, 95% CI 1.7–10.6, p <0.01) as demonstrated in Fig 2. In patients with RAS mutated CRC, concurrent presence of PIK3CA mutation did not influence survival.

Fig 2. Overall survival of RAS-mutated metastatic colorectal cancer with and without concurrent P53 mutations.

Fig 2

Kaplan-Meier curve comparing TP53 wildtype (yellow) vs TP53 mutated (blue) patient outcomes. Dotted line represents median (6.1 months versus 21.1 months, HR 4.2, 95% CI 1.7–10.6, p <0.01).

3.1.3 Allele frequency and its impact on survival

For 44 (24%) patients with RAS mutations, allele frequency and survival data was available. High RAS allele frequency, as defined by those with over 20%, demonstrated reduced overall survival (12.1 months versus not reached, HR 2.2, 95%CI 1.3–6.7, p = 0.04). This is demonstrated in Fig 3. RAS allele frequency did not correlate with upfront chemotherapy responsiveness as defined as stable disease or better on first imaging done approximately 3 months after commencement of first line chemotherapy in the metastatic setting. Median progression free survival in high RAS allele frequency patients was 6.8 months versus 10.3 months in low RAS allele frequency patients but this result was not statistically significant (p = 0.22). In 12 patients with BRAF V600E mutations (7%), allele frequency and survival data were available. High BRAF V600E mutation allele frequency, as defined by those with over 15%, trended towards shorter overall survival without statistical significance (9.8 months versus 12.0 months, p = 0.7).

Fig 3. Overall survival of RAS-mutated metastatic colorectal cancer based on allele frequency.

Fig 3

Kaplan-Meier curve comparing KRAS high variant allele frequent as defined by over 20% (yellow) vs KRAS low variant allele frequent as defined by below 20% (blue). Dotted line represents median (12.1 months versus not reached, HR 2.2, 95%CI 1.3–6.7, p = 0.04).

3.2 Ethnicity and mutational landscape

Culturally and linguistically diverse (CALD) patients are those who are either born in non-English speaking counties and/or who do not speak English at home [3]. 64/180 (36%) patients in our cohort were CALD background. There was a slightly higher incidence of any tier mutation identified in the CALD cohort compared with non-CALD populations (84% versus 80%) but this result was not statistically significant. RAS mutations were noted in 42% of patients (38% with KRAS mutations and 4% with NRAS mutations). In our cohort, patients from Asia had the highest rate of RAS mutation (52%) compared with any other ethnic subgroup with those of Middle Eastern ethnicity having the lowest rate of RAS mutation (36%) in our cohort. BRAF mutations were noted in 29 (17%) patients with 27/29 having the V600E alteration. The highest incidence of BRAF mutations was noted in the Australian Caucasian population compared with those from Asia or the Middle East (21% versus 18% and 13% respectively). In our cohort, PIK3CA mutations were present in 14% of Australian ethnicity patients, 14% of Eastern European patients, 17% of South East Asian ethnicity patients and 18% of Middle Eastern ethnicity patients. Additionally, one third of patients had some variant of TP53. TP53 mutations appeared to be fairly evenly present across the various ethnic subgroups with 33% of Australian ethnicity patients, 28% of Eastern European patients, 26% of South East Asian ethnicity patients and 45% of Middle Eastern ethnicity patients exhibiting mutations. An overall summary of mutations based on region of birth is presented in Table 5.

Table 5. Genetic variations based on region of birth in colorectal cancer.

Region of Birth N KRAS BRAF TP53 PIK3CA MET HER2
Australia / New Zealand 112 42 (38%) 23 (21%) 37 (33%) 16 (14%) 6 (5%) 3 (3%)
South East Asia 23 11 (48%) 3 (13%) 6 (26%) 4 (17%) 1 (4%) 0 (0%)
Pacific Islands 2 1 (50%) 0 (0%) 2 (100%) 0 (0%) 0 (0%) 0 (0%)
Eastern Europe 28 11 (40%) 0 (0%) 8 (29%) 3 (11%) 1 (4%) 1 (4%)
Middle East 11 4 (36%) 2 (18%) 5 (45%) 2 (18%) 0 (0%) 0 (0%)
Other 4 2 (50%) 1 (25%) 2 (50%) 0 (0%) 0 (0%) 0 (0%)

4. Discussion

NGS testing provides a wealth of information regarding a patient’s tumour biology, and informs mechanisms of treatment response and clinical prognosis. Currently, the majority of the information generated is not used in clinical decision making. Our study highlights important hypothesis generating questions including the influence of allele frequency and presence of non-traditional co-mutations on clinical outcomes. Currently, there are no published studies in the CRC space that investigate the impact of RAS allele frequency on clinical outcomes. Friedlaender et al., reported the role of allele frequency on survival outcomes in NSCLC [7]. Their retrospective study demonstrated that allele frequency was an independent prognostic marker of both progression free and overall survival in advanced EGFR-mutated NSCLC who were treated with EGFR targeting TKIs. This was also supported by Ding et al. who similarly demonstrated a possible prognostic role of allele frequency in EGFR-mutated NSCLC [6]. Furthermore, BRAF allele frequency has been shown to be of prognostic relevance in melanoma [10].

To our knowledge, our study is the first to suggest that NGS may provide clinically relevant information for prognostication of patients with metastatic CRC and high RAS allele mutation frequency. Our data suggests that patients with high RAS allele mutation frequency trended towards reduced overall survival as measured from diagnosis of metastatic disease. This raises questions in clinical decision making. Should clinicians be referring patients with high RAS allele frequency for clinical trials given poor prognosis or is there a role for increased surveillance imaging in these patients to detect earlier treatment failure? Our data set did not demonstrate a lack of upfront response to chemotherapy but further, larger studies are required to answer these questions. These same questions can be posed for patients with CRC tissue that carries the combination of TP53 and RAS mutation who show worse prognosis in our cohort (Fig 2). Only recently has the combined presence of both TP53 and RAS mutation been shown to be associated with reduced survival outcomes and our NGS data adds to the existing literature [11]. This data would not be available using more limited traditional mutation testing methods of immunohistochemistry or PCR thereby demonstrating the benefit of routine targeted NGS in metastatic CRC. Recently, early data has also proposed prognostic utility of BRAF allele frequency in metastatic CRC [12], thus further demonstrating the potential utility of routine NGS testing. Our data did not demonstrate any differences in survival based on BRAF allele frequency but findings may be limited to small numbers of BRAF-V600E mutated CRC in our cohort.

Our retrospective cohort study demonstrates the utility of routine NGS testing in stage IV CRC patients and describes the tumour mutational landscape in the Australian population. NGS is currently recommended as an alternative to mutation testing by PCR in CRC. Although the use of multigene sequencing panels is recommended in large research centres in the context of screening for access to novel potentially therapeutic targets, this needs to be balanced with cost and the detection of mutations of unknown clinical significance. In Table 4 we have listed examples of clinical trials that have been or are being conducted to investigate the role of these mutations either specifically in CRC or in all solid organ malignancies. All of the trials in this table are early phase I to II studies. The results of these studies may shift these mutations from potential tier 3 to 4 to a higher tier if clinical utility is determined but may also downstage these variants to Tier 5 if they are determined to be bystander mutations. Hence, we can see that extended panel testing currently remains investigational but is critical in a research or clinical trial context in identifying genes that are potential targets for directed therapy. It is unclear if larger gene panels may identify more genes with actionable targets in early phase studies. Furthermore, identification of resistance genes and those that confer worse prognosis is possible by targeted NGS. Ongoing studies are required to further clarify the role of specific mutations as either therapeutic targets, resistance pathways or prognostic markers. In addition, specific NGS panels for individual cancers is also important to consider should NGS testing be incorporated into routine practice. Of note, APC (Adenomatous Polyposis Coli) is recommended as part of young CRC and hereditary CRC work-ups but is not included as part of the Oncomine panel. This is a limitation with pan-cancer NGS testing and more targeted selection is required to ensure optimal testing and resource allocation.

In the future, NGS testing is likely to become increasingly common in the diagnostic setting given the increased therapeutic targets in cancer treatment. Although NGS provides a wealth of biometric data, the clinical utility of much of this information is currently unknown. Other potential challenges of routine NGS testing is a lack of standardization across testing centres and funding for NGS testing. NGS testing costs can range into thousands of dollars depending on the number of genes tested and whether both DNA and RNA are sequenced.

5. Conclusion

Our study adds to the literature by describing the tumour mutational landscape of patients with stage IV CRC in a diverse Australian population. In addition to identifying patients with genomic alterations suitable for clinically proven therapeutic options, standard of care NGS testing revealed an additional 22% of cases with Tier II & III ESCAT mutations in CRC. 43% of patients also demonstrated potentially targetable Tier III & IV mutations. Our data also highlighted that patients with concurrent TP53 and RAS mutations have significant worse prognosis than those with RAS mutation alone. Furthermore, we were able to demonstrate that allele frequency has a potential clinical role in prognostication as well as therapeutic decision making. This supports the routine use of extended gene panels in identifying potentially actionable mutations in standard clinical practice. Future research is directed towards investigating the utility and cost effectiveness of NGS panels, especially with the advent of larger multigene panels, as well as standardisation of NGS testing in metastatic CRC.

Supporting information

S1 Data

(XLSX)

Data Availability

All relevant data are within the paper and its Supporting information files.

Funding Statement

The author(s) received no specific funding for this work.

References

  • 1.Rawla P, Sunkara T, Barsouk A. Epidemiology of colorectal cancer: incidence, mortality, survival, and risk factors. Prz Gastroenterol. 2019;14(2):89–103. doi: 10.5114/pg.2018.81072 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Kopetz S, Grothey A, Yaeger R, Van Cutsem E, Desai J, Yoshino T, et al. Encorafenib, Binimetinib, and Cetuximab in BRAF V600E–Mutated Colorectal Cancer. New England Journal of Medicine. 2019;381(17):1632–43. doi: 10.1056/NEJMoa1908075 [DOI] [PubMed] [Google Scholar]
  • 3.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(11):1023–34. doi: 10.1056/NEJMoa1305275 [DOI] [PubMed] [Google Scholar]
  • 4.Douillard JY, Siena S, Cassidy J, Tabernero J, Burkes R, Barugel M, et al. Randomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: the PRIME study. J Clin Oncol. 2010;28(31):4697–705. doi: 10.1200/JCO.2009.27.4860 [DOI] [PubMed] [Google Scholar]
  • 5.Phipps AI, Ahnen DJ, Cheng I, Newcomb PA, Win AK, Burnett T. PIK3CA Somatic Mutation Status in Relation to Patient and Tumor Factors in Racial/Ethnic Minorities with Colorectal Cancer. Cancer Epidemiol Biomarkers Prev. 2015;24(7):1046–51. doi: 10.1158/1055-9965.EPI-15-0204 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ding PN, Roberts TL, Chua W, Becker TM, Caixeiro N, de Souza P, et al. Plasma pre-treatment T790M relative allelic frequency in patients with advanced EGFR-mutated non-small cell lung cancer predicts treatment response to subsequent-line osimertinib. Transl Lung Cancer Res. 2021;10(4):1623–34. doi: 10.21037/tlcr-20-1125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Friedlaender A, Tsantoulis P, Chevallier M, De Vito C, Addeo A. The Impact of Variant Allele Frequency in EGFR Mutated NSCLC Patients on Targeted Therapy. Front Oncol. 2021;11:644472. doi: 10.3389/fonc.2021.644472 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Mosele F, Remon J, Mateo J, Westphalen CB, Barlesi F, Lolkema MP, et al. Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO Precision Medicine Working Group. Annals of Oncology. 2020;31(11):1491–505. doi: 10.1016/j.annonc.2020.07.014 [DOI] [PubMed] [Google Scholar]
  • 9.Sakata S, Otsubo K, Yoshida H, Ito K, Nakamura A, Teraoka S, et al. Real-world data on NGS using the Oncomine DxTT for detecting genetic alterations in non-small-cell lung cancer: WJOG13019L. Cancer Sci. 2022;113(1):221–8. doi: 10.1111/cas.15176 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Berrino E, Balsamo A, Pisacane A, Gallo S, Becco P, Miglio U, et al. High BRAF variant allele frequencies are associated with distinct pathological features and responsiveness to target therapy in melanoma patients. ESMO Open. 2021;6(3):100133. doi: 10.1016/j.esmoop.2021.100133 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lillemoe HA, Passot G, Kawaguchi Y, DeBellis M, Glehen O, Chun YS, et al. RAS/TP53 Co-mutation is Associated With Worse Survival After Concurrent Resection of Colorectal Liver Metastases and Extrahepatic Disease. Ann Surg. 2022;276(2):357–62. doi: 10.1097/SLA.0000000000004672 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ros J, Matito J, Villacampa G, Comas R, Garcia A, Martini G, et al. Plasmatic BRAF-V600E allele fraction as a prognostic factor in metastatic colorectal cancer treated with BRAF combinatorial treatments. Annals of Oncology. doi: 10.1016/j.annonc.2023.02.016 [DOI] [PubMed] [Google Scholar]

Decision Letter 0

Avaniyapuram Kannan Murugan

18 Jul 2023

PONE-D-23-14740Multigene panel next generation sequencing in metastatic colorectal cancer in an Australian populationPLOS ONE

Dear Dr. Nindra,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process. Please submit your revised manuscript by Sep 01 2023 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols.

We look forward to receiving your revised manuscript.

Kind regards,

Avaniyapuram Kannan Murugan, M.Phil., Ph.D.

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and

https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. In your Data Availability statement, you have not specified where the minimal data set underlying the results described in your manuscript can be found. PLOS defines a study's minimal data set as the underlying data used to reach the conclusions drawn in the manuscript and any additional data required to replicate the reported study findings in their entirety. All PLOS journals require that the minimal data set be made fully available. For more information about our data policy, please see http://journals.plos.org/plosone/s/data-availability.

Upon re-submitting your revised manuscript, please upload your study’s minimal underlying data set as either Supporting Information files or to a stable, public repository and include the relevant URLs, DOIs, or accession numbers within your revised cover letter. For a list of acceptable repositories, please see http://journals.plos.org/plosone/s/data-availability#loc-recommended-repositories. Any potentially identifying patient information must be fully anonymized.

Important: If there are ethical or legal restrictions to sharing your data publicly, please explain these restrictions in detail. Please see our guidelines for more information on what we consider unacceptable restrictions to publicly sharing data: http://journals.plos.org/plosone/s/data-availability#loc-unacceptable-data-access-restrictions. Note that it is not acceptable for the authors to be the sole named individuals responsible for ensuring data access.

We will update your Data Availability statement to reflect the information you provide in your cover letter.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: No

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: The authors present a mutational landscape analysis of metastatic CRC patients using a targeted sequencing panel. The results suggest the importance of RAS /RAS-P53 mutations on survival in stage IV patients. More importantly, this analysis shows the value of NGS sequencing panels for stratifying patients into clinically actionable sub-groups.

1. Overall findings:

The work focuses on the importance of mutational profiling of patients to determine clinically actionable groups. It uses the survival dependence of KRAS-P53 mutations in an ethnically diverse cohort to show the utility of targeted NGS panels. It is important to expose this work and present the analytical strategy for other researchers/clinicians to consider. This multi-ethnic cohort represents the addition of new/novel data to the field.

The data/analysis presented does support the conclusions from the paper. The sample sizes could be larger, but this is frequently observed with human subject data. Generally, the manuscript lacks some key and relevant references, and discussion of the trends in context of the current similar findings in the field. For example, the KRAS-P53 effect on survival is documented in CRC and other cancers. The analysis results could be probed in more detail and it is not stated whether this is the only mutation pattern that bore any effect on survival trends.

2. Statistical analysis:

The sample size is small for statistical validity using Kaplain Meier statistics. For example, 14 patients with both RAS-P53 mutations in Fig 2a. However CI values are given which helps support the analysis trend. This trend is not novel and has been observed in multiple other studies/spontaneous CRC subtypes. This should be discussed in the manuscript.

See:

https://journals.lww.com/amjclinicaloncology/fulltext/2004/02000/simultaneous_mutations_in_k_ras_and_tp53_are.8.aspx and https://pubmed.ncbi.nlm.nih.gov/33351476/

Were these the only patterns identified that correlated with survival from the current study/oncomine profiles ?

What was the breakdown of sex/ethnic groupings in the KRAS-P53 mutated sets ? vs random sampling.

Other technical points include sharing a code/script/repo for the analysis code.

3. Legibility:

The manuscript was well written and easy to understand. The study goals were well described and the impact of the work clearly put into context with the current field.

4. Data Availability:

The research involves human subjects and consequently is exempt from the PLoS rules around making all datasets publicly accessible. The authors suggest that these data could be available on request. It is unclear why the data from this study might not be deposited in a protected-data site like dbGap or BioProject archive.

Reviewer #2: The manuscript entitled “Multigene panel next generation sequencing in metastatic colorectal cancer in an Australian population” by Nindra et al. reported a 50-gene panel analysis of a large cohort of CRC samples by next generation sequencing (NSG) and concluded this approach is beneficial as it is likely to identify actionable genomic alterations. However, additional analyses are needed to support the conclusions.

1. APC is the most frequently mutated gene in colon cancer, but it is not included in the panel. Do the authors have any information about APC mutational status of these samples? If yes, taking into account the APC mutations, whether the conclusion is different from the current one?

2. Is there any correlation of mutation loads/amount of mutated genes detected with the MMR status in the samples tested?

3. Whether there are any correlation of the different mutations with the responses to any therapies the patients received?

4. What kinds of mutations were detected by this method? Do these mutations include non-sense mutations or only include the ones that have been reported to alter the function of the genes? If non-sense mutations were included, the authors should consider separate them from the other damaging mutations.

5. Related to the Introduction section, as far as I know, BRAF is a S/T kinase, and therefore the encorafenib is not a tyrosine kinase inhibitor.

6. It should be CDKN2A instead of CDK2NA in Results 3.1.1. Also, it is confusing that the authors claimed there was no ongoing trials testing the roles of the 12 genes. At least inhibitors for MTOR and KIT are readily available.

7. Whether the difference of RAS mutations among diverse ethnic groups is statistically significant?

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: Yes: Anna Lobley

Reviewer #2: No

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2023 Oct 5;18(10):e0292087. doi: 10.1371/journal.pone.0292087.r002

Author response to Decision Letter 0


20 Jul 2023

Response to Reviewers:

Comment 1:

The authors present a mutational landscape analysis of metastatic CRC patients using a targeted sequencing panel. The results suggest the importance of RAS /RAS-P53 mutations on survival in stage IV patients. More importantly, this analysis shows the value of NGS sequencing panels for stratifying patients into clinically actionable sub-groups.

Answer 1:

We thank the reviewer for this positive feedback. There is a paucity of data in the Australian context on NGS testing in the colorectal space and thus our manuscript is novel in adding to that information. We feel that our cohort has a significant degree of multi-cultural variability, reflective of the modern Australian cohort, and thus is a relevant literature manuscript

Comment 2:

The work focuses on the importance of mutational profiling of patients to determine clinically actionable groups. It uses the survival dependence of KRAS-P53 mutations in an ethnically diverse cohort to show the utility of targeted NGS panels. It is important to expose this work and present the analytical strategy for other researchers/clinicians to consider. This multi-ethnic cohort represents the addition of new/novel data to the field.

Answer 2:

Again, we thank the reviewer for this positive feedback

Comment 3:

The data/analysis presented does support the conclusions from the paper. The sample sizes could be larger, but this is frequently observed with human subject data. Generally, the manuscript lacks some key and relevant references, and discussion of the trends in context of the current similar findings in the field. For example, the KRAS-P53 effect on survival is documented in CRC and other cancers. The analysis results could be probed in more detail and it is not stated whether this is the only mutation pattern that bore any effect on survival trends.

Answer 3:

Again, we thank the reviewer for this positive feedback. This cohort of 180 patients is one of the largest NGS testing samples of CRC done in Australia. We did not identify any other mutation patterns that affected survival. Of note, PIC3KA correlated with no difference in clinical outcomes and this has been mentioned in our manuscript.

Comment 4:

The sample size is small for statistical validity using Kaplain Meier statistics. For example, 14 patients with both RAS-P53 mutations in Fig 2a. However, CI values are given which helps support the analysis trend. This trend is not novel and has been observed in multiple other studies/spontaneous CRC subtypes. This should be discussed in the manuscript.

See:

https://journals.lww.com/amjclinicaloncology/fulltext/2004/02000/simultaneous_mutations_in_k_ras_and_tp53_are.8.aspx and https://pubmed.ncbi.nlm.nih.gov/33351476/

Answer 3:

Again, we thank the reviewer for this positive feedback. The reference “https://pubmed.ncbi.nlm.nih.gov/33351476/” mentioned is already cited in our discussion and we have made reference to previous manuscripts highlighint the poor prognostic trnd in RAS and P53 mutated CRC.

We did not include “https://journals.lww.com/amjclinicaloncology/fulltext/2004/02000/ simultaneous_mutations_in_k_ras_and_tp53_are.8.aspx” as the results are from 2004 and treatment outcomes from that period are dated and hard to correlate with current standard of care regimens.

Comment 4:

Were these the only patterns identified that correlated with survival from the current study/oncomine profiles ?

Answer 4:

We did not identify any other mutation patterns that affected survival. Of note, PIC3KA correlated with no difference in clinical outcomes and this has been mentioned in our manuscript.

Comment 5:

What was the breakdown of sex/ethnic groupings in the KRAS-P53 mutated sets ? vs random sampling.

Answer 5:

There was no grouping or random sampling applied in the cohort. We used 180 sequential samples of CRC.

Comment 6:

The manuscript was well written and easy to understand. The study goals were well described and the impact of the work clearly put into context with the current field.

Answer 6:

Again, we thank the reviewer for this positive feedback

Comment 7:

The research involves human subjects and consequently is exempt from the PLoS rules around making all datasets publicly accessible. The authors suggest that these data could be available on request. It is unclear why the data from this study might not be deposited in a protected-data site like dbGap or BioProject archive.

Answer 7:

We have uploaded a de-identified minimum data set as part of the supplemental material

Comment 8:

APC is the most frequently mutated gene in colon cancer, but it is not included in the panel. Do the authors have any information about APC mutational status of these samples? If yes, taking into account the APC mutations, whether the conclusion is different from the current one?

Answer 8:

APC genes are not mutated in colon cancer but are germline mutations that predispose to increased risk of colon cancer. NGS testing is done on somatic tumour tissue to look for tumour specific rather than germline specific mutations. Thus this comment is not relevant to our project with assesses the role of NGS testing in somatic tissue mutation identification in CRC.

Comment 9:

Is there any correlation of mutation loads/amount of mutated genes detected with the MMR status in the samples tested?

Answer 9:

No specific correlation is noted between tumour mutational burden and MMR – although this is not a key focus or specific to NGS testing and thus outside the scope of our paper

Comment 10:

Whether there are any correlation of the different mutations with the responses to any therapies the patients received?

Answer 10:

Upfront response rate to first line chemotherapy was not different between mutations but given the retrospective nature of the project this is a difficult question to answer. Prospective cohort studies are required to accurately assess tumour response using RECIST1.1 criteria to assess this and cannot be inferenced from our data.

Comment 11:

What kinds of mutations were detected by this method? Do these mutations include non-sense mutations or only include the ones that have been reported to alter the function of the genes? If non-sense mutations were included, the authors should consider separate them from the other damaging mutations.

Answer 11:

We have described the types and nature of mutation testing in our manuscript as follows: -“Next generation sequencing was performed with the Ion Torrent Genexus integrated next generation sequencer using the Oncomine Precision AssayTM, which is 50-gene NGS panel that was introduced to our laboratory in June 2021. Only DNA hotspot mutations and copy number variations (CNVs) were assessed. RNA sequencing for fusions was not performed. The Ion Torrent Genexus System fully automates the specimen-to-report workflow. It automates all steps of the targeted NGS workflow starting from extraction, purification and quantification of nucleic acid. The Genexus Purification System replaces sample prep by extracting and quantifying nucleic acids. This is followed by automation of the library preparation (including DNA synthesis), template preparation and sequencing. Lastly, primary data analysis is also automated and variant reporting for DNA applications are performed using the OncomineTM Reporter software, a Thermo Fisher supplied bioinformatics analytic tools. The reported results from the 50-gene OncomineTM Precision Assay are filtered to provide a summary of the variants and copy number variation as well as the variant allele frequency.”

Comment 12:

Related to the Introduction section, as far as I know, BRAF is a S/T kinase, and therefore the encorafenib is not a tyrosine kinase inhibitor.

Answer 12:

Amended; Encorafenib acts as an ATP-competitive RAF kinase inhibitor – we have updated the manuscript to reflect this.

Comment 13:

It should be CDKN2A instead of CDK2NA in Results 3.1.1.

Answer 13:

Amended; Thank you.

Comment 14:

Whether the difference of RAS mutations among diverse ethnic groups is statistically significant?

Answer 14:

There was a slightly higher proportion of RAS mutations in patients born in Australia compared with overseas but the results were not statistically significant

Comment 15:

PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

Answer 15:

Yes, happy for publication of peer review history

Attachment

Submitted filename: Response to Reviewers.docx

Decision Letter 1

Avaniyapuram Kannan Murugan

3 Sep 2023

PONE-D-23-14740R1Multigene panel next generation sequencing in metastatic colorectal cancer in an Australian populationPLOS ONE

Dear Dr. Nindra,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

Please submit your revised manuscript by Oct 18 2023 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols.

We look forward to receiving your revised manuscript.

Kind regards,

Avaniyapuram Kannan Murugan, M.Phil., Ph.D.

Academic Editor

PLOS ONE

Journal Requirements:

Please review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the rebuttal letter that accompanies your revised manuscript. If you need to cite a retracted article, indicate the article’s retracted status in the References list and also include a citation and full reference for the retraction notice.

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #1: All comments have been addressed

Reviewer #2: (No Response)

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: I Don't Know

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: No

Reviewer #2: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: The authors have addressed the outstanding queries in their responses, however, I'm not sure the minimal data added as supplementary materials is sufficient to support the findings in the manuscript. Were there any sample qc results for the patients samples ie. Depth and read numbers or similar ? The only other query I have is that the confidence intervals appear to be shown only for one of the groups on the test... could the authors clarify if the baseline kras data a single sample or the CI interval not shown ? Following clarity in this aspect I would recommend the work be published.

Reviewer #2: In this recently revised manuscript, the authors have made significant strides in addressing the concerns I had previously raised. However, there remains an issue pertaining to APC mutations that I strongly disagree with the authors on. In their rebuttal letter, the authors assert that APC is exclusively subject to germline mutations and not somatic mutations in the context of colorectal cancer (CRC). I find this assertion problematic due to the well-established knowledge spanning over two decades, which affirms that somatic APC mutations are indeed among the earliest events in the development of non-hereditary colorectal carcinogenesis. These somatic mutations are pivotal in driving the formation of early adenoma/dysplastic crypt structures.

While it is reasonable to acknowledge that the commercial 50-gene panel used in the Oncomine Precision AssayTM might not have initially incorporated APC, I believe that as professionals and investigators focused on CRC, the authors should have at least initiated a scientific discussion concerning the potential limitations of omitting this widely recognized driver gene from their study. It seems unlikely that the authors were unaware of prior genetic research pertaining to APC's role in CRC.

With the exception of this concern, I do not have any further queries or reservations regarding the content of this manuscript.

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

**********

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2023 Oct 5;18(10):e0292087. doi: 10.1371/journal.pone.0292087.r004

Author response to Decision Letter 1


6 Sep 2023

Response to Reviewers

Comment 1:

The authors have addressed the outstanding queries in their responses, however, I'm not sure the minimal data added as supplementary materials is sufficient to support the findings in the manuscript. Were there any sample qc results for the patients samples ie. Depth and read numbers or similar?

Answer 1:

Depth and read numbers were not available on next generation sequencing reports. For example, I have attached a redacted copy of one example of the output next generation sequencing obtained. As you can see below, such information is not present.

Comment 2:

The only other query I have is that the confidence intervals appear to be shown only for one of the groups on the test... could the authors clarify if the baseline kras data a single sample or the CI interval not shown ?

Answer 2:

Apologies, that is an omission on our end. The confidence intervals are now included in the main text and figure.

Comment 3:

Following clarity in this aspect I would recommend the work be published.

Answer 3:

We thank the authors for their feedback and hope PLOS ONE agrees for publication.

Comment 4: In this recently revised manuscript, the authors have made significant strides in addressing the concerns I had previously raised. However, there remains an issue pertaining to APC mutations that I strongly disagree with the authors on. In their rebuttal letter, the authors assert that APC is exclusively subject to germline mutations and not somatic mutations in the context of colorectal cancer (CRC). I find this assertion problematic due to the well-established knowledge spanning over two decades, which affirms that somatic APC mutations are indeed among the earliest events in the development of non-hereditary colorectal carcinogenesis. These somatic mutations are pivotal in driving the formation of early adenoma/dysplastic crypt structures. While it is reasonable to acknowledge that the commercial 50-gene panel used in the Oncomine Precision AssayTM might not have initially incorporated APC, I believe that as professionals and investigators focused on CRC, the authors should have at least initiated a scientific discussion concerning the potential limitations of omitting this widely recognized driver gene from their study. It seems unlikely that the authors were unaware of prior genetic research pertaining to APC's role in CRC. With the exception of this concern, I do not have any further queries or reservations regarding the content of this manuscript.

Answer 4:

We thank the authors for their feedback. We agree with the reviewer that somatic APC mutations are among the earliest in the development of colorectal cancer. APC mutations are typically screened using specific PCR or NGS in certain subgroups.

Based on Australian guidelines, testing for APC using NGS is only indicated in patients who have isolated colorectal cancer under the age of 40, those with KRAS G12C mutations or those with family histories suggestive of hereditary cancer. This gene testing is not part of the Oncomine 50-gene panel, which in itself if an investigational tool and not part of standard government funded practice in Australia.

We have added a discussion regarding the APC gene in our revised manuscript, but it’s relevance and utility is outside the scope of our manuscript.

Attachment

Submitted filename: Response to Reviewers v2.docx

Decision Letter 2

Avaniyapuram Kannan Murugan

12 Sep 2023

Multigene panel next generation sequencing in metastatic colorectal cancer in an Australian population

PONE-D-23-14740R2

Dear Dr. Nindra,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice for payment will follow shortly after the formal acceptance. To ensure an efficient process, please log into Editorial Manager at http://www.editorialmanager.com/pone/, click the 'Update My Information' link at the top of the page, and double check that your user information is up-to-date. If you have any billing related questions, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Avaniyapuram Kannan Murugan, M.Phil., Ph.D.

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Acceptance letter

Avaniyapuram Kannan Murugan

19 Sep 2023

PONE-D-23-14740R2

Multigene panel next generation sequencing in metastatic colorectal cancer in an Australian population

Dear Dr. Nindra:

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now with our production department.

If your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information please contact onepress@plos.org.

If we can help with anything else, please email us at plosone@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Avaniyapuram Kannan Murugan

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Data

    (XLSX)

    Attachment

    Submitted filename: Response to Reviewers.docx

    Attachment

    Submitted filename: Response to Reviewers v2.docx

    Data Availability Statement

    All relevant data are within the paper and its Supporting information files.


    Articles from PLOS ONE are provided here courtesy of PLOS

    RESOURCES