Open Access

Current status and perspectives of genetic testing in gastrointestinal cancer (Review)

  • Authors:
    • Tasuku Matsuoka
    • Masakazu Yashiro
  • View Affiliations

  • Published online on: November 16, 2023     https://doi.org/10.3892/ol.2023.14155
  • Article Number: 21
  • Copyright: © Matsuoka et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Genetic testing has become widespread in daily medical care for gastrointestinal (GI) cancers. However, unlike breast cancer and non‑small cell lung cancer, in which personalized medicine targeting various driver genes is standardized, the incidence of targeted gene abnormalities in GI cancers is low. Nevertheless, such abnormalities may be linked to therapeutic agents and the further development of therapeutic agents for personalized medicine for GI cancers is desired. A liquid biopsy is of great benefit in offering clinical decision support, in applications such as GI cancer screening, surgical interventions, monitoring disease status and enhancing patient survival outcomes, all of which would contribute to personalized medicine. Germline genetic testing is required for several types of GI cancer, which shows clinical indications of hereditary predisposition. The increasing use of multigene panel testing has redefined gene‑cancer associations, and consequently the estimate of cancer risk that vary from low to high penetrance. Comprehensive genetic testing can enable the detection of novel treatment targets and the discovery of undefined multiple diagnostic/predictive markers, which may enhance the molecular‑level understanding of GI cancers. Genetic testing can also aid the design of more appropriate and adequate genomic‑driven therapies for patients who may benefit from other standardized therapeutic methods.

Introduction

Gastrointestinal (GI) cancers include several malignancies of the gastrointestinal tract and organs such as the stomach, colon, liver, intrahepatic bile duct, gallbladder, and pancreas. GI cancers have epithelial cell or stromal origin, and GI cancers account for approx. 30% of all tumor cases in 2018. Together GI cancers have been responsible for over 3.5 million deaths which corresponds to 37% of the deaths from all human malignancies (1).

While clinical assessment of GI cancer has been performed with a physical examination, blood test, imaging, and endoscopy, recent advancements in genomics have led to the development of genetic analysis for diagnosis (2,3). In the modern era of precision medicine, genetic testing has been incorporated into routine clinical practice to assist decision-making regarding appropriate genetically matched treatments for patients with GI cancers (4). To acquire genetic information in a timely and cost-effective manner, a variety of gene panel tests, which is based on next-generation sequencing (NGS), have been progressed and some have been approved by the U.S. Food and Drug Administration (FDA) as companion diagnostics for multiple molecular-targeted therapies (5). In contrast, the clinical value of genetic testing for GI cancer receptiveness is not well established. This review describes the current genomic landscape in GI cancers, and testing modalities that have prognostic, predictive, and therapeutic value. We provide an outline of the clinical use of multi-gene testing in GI cancers, and discuss the practical utility and potential of a liquid biopsy and whole genome sequencing.

Methods

Non-systematic review was conducted in a basis of an electronic search through the medical literature using PubMed and Google Scholar. The keywords ‘genetic testing’, ‘Multi-gene panel testing’, ‘Whole genome sequencing’, ‘Next-generation sequencing’, ‘gastrointestinal cancer’, ‘colorectal cancer’, ‘pancreatic cancer’, ‘gastric cancer’, ‘hepatobiliary cancer’, ‘esophageal cancer’, were searched. Guidelines and review articles from gastroenterology, oncology, endoscopy and genetics were included. When more than one guideline concerning the same subject was available, the latest one was picked up. Only full articles in the English language published in the last ten years were considered to be suitable for review. The exclusion criteria consisted duplicated articles, studies absent of diagnostic outcomes. Case reports, editorials, book chapters, correspondences, letters, and non-human research were not included. First, the titles were screened and appropriate studies were selected. Of these studies, the full text was acquired. A total of 258 articles were identified (Fig. 1).

Multi-gene panel testing

Comprehensive genomic profiling by NGS enables the detection of multiple genomic features in GI cancers. Multi-gene testing is conducted based on NGS platforms, and before sequencing, genomic regions of attention are chosen from the DNA sample (6). The sustainability and quality assurance of the molecular tumor board, named Expert Panel, examines the appropriate treatment methods such as drug treatment and participation in clinical trials, and the results were informed. Multi-gene panels are largely utilized in clinical settings for the identification of somatic and germline mutations in GI cancers, which lead to molecular classification, and prediction of therapeutic effect. It can also detect the gene which is involved in drug resistance. Additionally, microsatellite instability (MSI) and tumor mutational burden (TMB) are approved by the FDA as companion diagnostics for GI cancers.

Recently, the survival benefit of matched therapy using panel testing has been established. An encouraging impact on tumor response rates, patient outcome, and on detecting novel tools of molecularly-targeted therapy has been suggested by some clinical trials by utilizing multi-gene testing which may lead to personalized cancer treatments (79). The Know Your Tumor program testing matched therapies after multi-omics profiling provided suggestions for elucidated clinical trials and personalized therapy for patients with pancreatic cancer (PC). The outcomes of this trial revealed that the patients with BRCA mutations after Poly (ADP-Ribose) Polymerase (PARP) inhibitor treatment or those with mismatch repair deficiency after immune checkpoint blockade (ICB) treatment demonstrated 1-year survival benefit compared with patients who received unmatched therapies or those without an actionable molecular change (10). This study also showed that mutations in the DNA damage response (DDR) pathway were the most popular actionable alteration. These data would indicate a guarantee for this precision approach. Although the frequency of druggable genetic alterations in GI cancers is lower than that in breast cancer and non-small cell lung cancer, a variety of noble candidate genes have been identified over the past few years.

In this section, we summarize the current status of genetic tests and molecular-targeted therapies for GI cancer that are expected in the future (Table I).

Table I.

Key genetic alterations of gastrointestinal cancer and the corresponding targeted therapies.

Table I.

Key genetic alterations of gastrointestinal cancer and the corresponding targeted therapies.

Genomic profilingTargeted agent
Gastric cancer
  HER2Trastuzumab, Trastuzumab-
Deruxtecan
  FGFRBemarituzumab
  VEGFRN/A
  MSIPembrolizumab
Colorectal cancer
  EGFRCetuximab, Panitumumab
  BRAFV600EEncorafenib
  PIK3CAN/A
  MEKBinimetinib
  HER2Trastuzumab, Trastuzumab-
Deruxtecan
  KRAS G12CSotorasib, Adagrasib
  NTRKLarotrectinib, Entrectinib
  MSIPembrolizumab, ipilimumab
Pancreatic cancer
  KRASN/A
  TP53N/A
  CDKN2AN/A
  SMAD4N/A
  EGFRErlotinib
  NTRKLarotrectinib, Entrectinib
  ALKN/A
  BRAFEncorafenib
  PIK3CAN/A
  BRCA/HRDOlaparib
  MSIPembrolizumab
Hepatocellular carcinoma
  TERTN/A
  TP53N/A
  CTNNB1N/A
  VEGFRLenvatinib, Cabozantinib
  ARID1AN/A
  CCND1N/A
  METN/A
  PTENN/A
Biliary tract cancer
  FGFRPemigatinib, Infigratinib
  IDH1/2Ivosidenib
  BRAFN/A
  TP53N/A
  HER2Trastuzumab, Pertuzumab
  PIK3CAN/A
  NTRKLarotrectinib, Entrectinib
Colorectal cancer

For a decade, patients with KRAS/NRAS wild type are acceptable for therapy targeting the epidermal growth factor receptor (EGFR) signaling (11). BRAFV600E mutation is identified in approximately 8–10% of colorectal cancer (CRC) and generates a RAS-independent constitutional activation of the mitogen-activated protein kinases (MAPK) pathway, which leads to cell growth and survival and is a prognostic biomarker for patients with CRC (12). Even if some BRAF mutations are identified beyond the V600 hotspot in CRC, they do not present similar clinical, biological, and therapeutic results as the V600E mutation (13). These BRAF non-V600E mutated tumors tend to be well differentiated with left-sided tumor site and were correlated with improved prognosis and resistance to BRAF inhibitors, whereas some have a sensitivity to EGFR (14,15). Notably, the MSI phenotype, which can predict the efficiency of immune checkpoint blockade (ICB) therapies, was identified in approximately 20% of BRAFV600E CRC, regardless of the BRAF mutational status (12). BRAF inhibition has been said to cause a rapid feedback EGFR activation, which assists MAPK constitutive signaling. Continued proliferation and resistance of these tumors to BRAF inhibitor monotherapy may occur by EGFR-mediated stimulation of downstream signaling (16). In light of these, the combination strategy with the BRAF inhibitor, anti-EGFR agents, phosphatidylinositol-3-kinase (PI3K) inhibitors, or MEK inhibitors was investigated (1720). These studies assisted the scheme of the BEACON CRC phase III study, which elucidated encorafenib, binimetinib, and cetuximab or encorafenib and cetuximab, or other treatment options, such as cetuximab and irinotecan or cetuximab and FOLFIRI (folinic acid, fluorouracil, and irinotecan). Metastatic CRC (mCRC) patients harboring a BRAF exon 15 p.V600E point mutation, with disease progression after one or two prior treatment approaches, were randomized. Conclusively, the median overall survival (OS) was prolonged over 3 months in the triplet and the doublet experimental regimens, compared to the control. Notably, median progression-free survival (PFS) was superior in the triple-combination group and in the association of the doublet group compared with the other group. These data indicated the clinical benefit of the molecular-targeting combination therapy in previously treated patients with mCRC harboring a BRAF exon 15 p.V600E point mutation (21). Although the two experimental regimens were not compared in the study the combination of encorafenib plus cetuximab, without the MEK inhibitor, is currently positioned as the standard for second- and third-line BRAF V600E-mutated mCRC (22). KRASG12C (glycine 12 to aspartic acid) is one of the most popular KRAS mutations in CRC. A novel production of KRAS inhibitors may result in a revolutionary change in the treatment for patients with CRC (23). In a recent, convincing potencies of a direct KRASG12C inhibitor were described. AMG 510 is a new small molecule that exclusively and irretrievably impaired KRASG12C activity, by locking it in an inactive guanosine diphosphate-bound state. The initial study using AMG 510 in patients with advanced or metastatic KRASG12C mutant solid tumors (CodeBreak-100; NCT03600883) demonstrated that ORR and DCR were 12.0 and 80.0%, respectively, in KRASG12C-mutated mCRC patients (24). Although the majority of CRCs initiate through the chromosomal instability pathway, 10–15% of CRCs occur based on the MSI pathway. MSI/dMMR CRCs are characterized by a high TMB with highly immunogenic neoantigens arising from frameshift mutations that induce high infiltration through activated cytotoxic T CD8+ lymphocytes (25,26). for the therapy of CRC patients with MSI/dMMR, who progressed after first or second chemotherapy (27). HER2 gene amplification was found in approximately 1 to 8% of CRC patients (28). Although the prognostic implication of HER2 amplifications is controversial, a negative predictive value of HER2 amplifications for anti-EGFRs efficacy tends to be familiar (29). Phase II studies, named Heracles-A, and MyPathway, evaluated the combination therapy of trastuzumab plus lapatinib, and trastuzumab with pertuzumab. Convincing response rates of 30 and 32% and median PFS of 4.7 and, 2.9 months, respectively were demonstrated (30,31). Trastuzumab-Deruxtecan (T-DXd) is a new antibody drug conjugate consisting of a humanized anti-human epidermal growth factor receptor2 (HER2) antibody, a cleavable, peptide-based linker, and a potent topoisomerase I inhibitor. T-DXd showed a preliminary effect on HER2-positive mCRC refractory to standard therapy, which may lead to the increased advancement of precision treatment of HER2-positive CRC (32).

Recently, NTRK gene fusions emerged as a greatly attractive target for the treatment of patients with cancer. A remarkable clinical significance is demonstrated by TRK inhibitors (larotrectinib, entrectinib). The ALK and ROS1 genes, which encode for the homonym tyrosine kinase receptors, mediate various cellular biological activity via diverse signal transduction (33). ALK, ROS1, and NTRK fusions occur in 0.2 to 2.4% of CRCs (34). Hence, it needs to choose the population to be examined.

Pancreatic cancer

A recent study utilized whole genome sequencing (WGS) to map the genome of 100 pancreatic ductal adenocarcinoma (PDAC) specimens (35). Acknowledged common drivers of PDAC (KRAS, TP53, CDKN2A, and SMAD4) were emphasized in this study, and also various other mutations at greatly lower frequencies were shown. Activating KRAS mutations are found in more than 90% of PDAC. Besides, ALK rearrangements, NRG1 rearrangements, NTRK fusions, ROS, BRAF, PIK3CA, and a variety of cancer-associated genes identified as potential drivers have been detected (e.g., STK11, RB1, GNAS, CHEK2, and ERBB2), which may lead to potential targets. Inactivation of tumor suppressor genes, including SMAD4, CDKN2A, and, TP53 is detected in advancing pancreatic intraepithelial neoplasia progression and arises in up to 50% (36). The frequency of persistently mutated genes then diminishes to less than 10%, which accumulates into central molecular pathways, such as KRAS, wingless and int, TGF-β signaling, DDR, NOTCH, RNA processing, cell cycle regulation (37). Associations of numerous pathways with survival have been detected by analyses of pathways in PDAC patients. DNA repair-related pathways were shown to contribute to a poor outcome (38). In preclinical vivo models and the clinical setting, several of these pathways can be actionable targets for treatment. BRAF mutation and NTRK gene fusions in KRASWT, MMR-D/MSI-H, and genetic alterations in homologous recombination deficiency (HRD) are considered to be prospective actionable mutations. The American Society of Clinical Oncology (ASCO) guidelines recommended early examination for actionable genetic alterations for PC patients who can be convincing candidates for subsequent therapy following first-line therapy (39). Patients with BRCA mutations, NTRK gene fusions, and MSI-H/MMR-D are likely to be provided personalized therapies, such as PARP inhibitors, TRK fusion inhibitors, and ICB therapy, respectively (10).

In ovarian and breast cancers, discriminate defects in Homologous Recombination DNA repair genes, such as germline mutations in BRCA1, 2, and PALB2, somatic mutations in BRCA1, 2, and promoter methylation of BRCA1, have been represented (40,41). BRCA mutations also contributed to promoting the risk for PC. BRCA genes encode for proteins involved in the HR repair of DNA double-stranded breaks. PC patients with deficient HR repair are predicted to be responsible for PARP inhibition. Hence, PARP inhibitors are efficient for selective patients with HRD owning to BRCA1 or BRCA2 mutations (42). According to recent experiments of genomic profiling in large populations of PDAC, the importance of HRD in predicting the efficacy on PARP inhibitors and platinum-based therapy was accumulated (10,43). ASCO guidelines recommended treatment with PARP inhibitor or platinum-based chemotherapy for patients with a germline BRCA1, 2 mutations. In a recent randomized phase III study (POLO), the efficacy of olaparib was demonstrated in germline BRCA-mutated metastatic PDAC (44), suggesting that HRD can effectively be targeted in pancreatic cancer. Due to Lynch syndrome or somatic MMR gene mutations, about 1% of PDAC patients have MMR-D/MSI-H (45,46). ASCO guidelines reported that pembrolizumab is advocated as a second-line therapy for PDAC patients with MMR-D/MSI-H (39).

Gastric cancer

The Phase III TOGA trial assessed the trastuzumab-containing regimen compared with standard first-line chemotherapy. Trastuzumab yielded a statistical improvement in terms of median OS, median PFS, and overall response rate (ORR) (47). According to these results, trastuzumab combined therapy come to be the standard treatment for advanced HER2-positive gastric cancer (GC). Although the continuous administration of trastuzumab after progression failed to improve PFS in patients with HER2-positive GC, a phase II trial that assessed the efficacy and safety of T-DXd vs. a physician's choice of chemotherapy in patients with HER2-positive GC treated with two prior lines including trastuzumab (The DESTINY-Gastric01 trial) showed that OS, as a key secondary endpoint with T-DXd, was significantly improved (median OS 12.5 vs. 8.4 months, HR 0.59, P=0.01). According to these results of the DESTINY-Gastric01 trial, T-DXd was approved for the treatment of patients with HER2-positive unresectable GC in Japan (48,49).

Alterations in fibroblast growth factor receptor (FGFR) genes are found in gastric and gastro-esophageal junction cancers and frequency ranges between 3–7%. The most familiar alteration is amplifications, which are followed by rearrangements and mutations (50). The amplification level was revealed to be negatively associated with patients' prognosis (51). A first-in-class humanized fucosylated IgG1 monoclonal antibody directed against FGFR2b, bemarituzumab has demonstrated convincing results in a phase I study in solid tumors and FGFR2b-positive GC patients (52). The addition of bemarituzumab to chemotherapy was evaluated in first-line therapy in locally advanced, unresectable, metastatic HER2-negative and FGFR2b-positive GC patients. Notably, median PFS was prolonged to 9.5 months in the bemarituzumab group, compared to 7.4 months in the placebo group. Median OS was not reached in the bemarituzumab group vs. 12.9 months in the placebo arm (HR, 0.58, 95% confidence interval 0.35–0.95; P=0.03) and ORR was prolonged from 40 to 53% in the bemarituzumab group (53). Evaluation of Phase III trials in the near future is expected (NCT03694522).

Hepatocellular carcinoma

Although the etiology of hepatocellular carcinoma (HCC) is unsatisfactorily evaluated, recent developments in genomic studies have provided a profound understanding of HCC advancement and may result in new approaches for prevention and treatment. TP53, CTNNB1, ARID1A, ARID2, AXIN1, RB1, and NFE2L2 are the most common mutations in HCC. In a recent, catalytic telomerase reverse transcriptase (TERT) has been distinguished as a frequent driver mutation which is identified in 40–65% of HCC patients (54,55). VEGFA, MYC, CCND1, and MET are other oncogenes frequently amplified (56,57), PTEN is often suppressed (58) and p16 is commonly deleted in HCCs (59). Present guidelines recommend chemotherapy, with sorafenib being the only first-line therapy for unresectable HCC because of its approval in 2007. A recent REFLECT trial demonstrated that lenvatinib revealed OS non-inferiority to sorafenib (60). The IMbrave 150 trial displayed a combination of atezolizumab plus bevacizumab showed better PFS and OS than that associated with sorafenib (61). Therefore, atezolizumab plus bevacizumab has been positioned as a first-line HCC therapy. According to the RESORCE trial, which showed the superiority of prognosis in patients with HCC whose disease progressed during sorafenib treatment, Regorafenib has been approved as a second-line therapy (62). Additionally, in a phase III trial (CELESTIAL), cabozantinib was presented to have met clinical endpoints, compared with control, as a second-line treatment (63). However, there are no molecular-targeted drugs that match these major genetic abnormalities, and personalized medicine is rarely conducted.

Biliary tract cancers

Biliary tract cancers have poor prognoses even when cytotoxic chemotherapy is applied. Based on the phase 3 ABC-02 and BT-22 trials, combined cisplatin with gemcitabine is the recent standard treatment in unresectable, metastatic biliary tract cancers (55,64). In the second-line setting, FOLFOX (folinic acid, 5-fluorouracil, and oxaliplatin) showed a prolonged OS compared with best supportive care in the phase 3 ABC-06 trial (65). The molecular analysis of biliary tract cancers has significantly improved understanding of the underlying pathological mechanism which may lead to novel targeted therapeutic approaches. FGFR2 fusions and IDH1/2 mutations are the most ordinary and clinically important genetic aberrations in intrahepatic cholangiocarcinoma, whereas TP53 mutations, KRAS mutations, and HER2 amplifications are the most meaningful genetic aberrations in extrahepatic cholangiocarcinoma (66).

Liquid biopsy

GI cancers account for a significant proportion of mortality worldwide (1). For these tumors, staging at diagnosis persists as the most principal prognostic factor. With the advancement of tumor biology, it has become important to search for basic knowledge such as pathology as well as for biomarkers that characterize tumors for a treatment approach. Although genome-based precision medicine is convincing, tissue-based genomic sequencing for first-line therapy decision-making in GI cancer remains obstacles owning to the long turnaround time between the receipt of tissue samples and reporting results. Liquid biopsy has the potential to detect circulating tumor DNA (ctDNR) from all tumors that shed into the circulation and can be used to assess intratumor genetic heterogeneity and overcome the limitations of tissue analysis. Circulating tumor cells, ctDNR, exosome, and microRNAs exist in the blood or other body fluids and exhibit the tumor condition in real-time. More recently, methods based on NGS have enabled ctDNA profiling as a replacement for tumor tissue sequencing (67,68). So far, assays applied for ctDNA can be categorized into two classes: those targeted for a single or small number of variants including CAPP-Seq, Safe-seq, Signatera, or ArcherDX, which have a limit of identification no more than 0.01% variant allele frequency, and those aimed at a broader coverage. These comprehensive panel-based sequencing assays which integrate genomic alterations as well as methylation status, are used for genotyping or early diagnosis and achieve a detection limit of approximately 0.2% in the Guardant Health Reveal test (69). Furthermore, cancer genomic testing using ctDNA has been commercialized and approved with an insurance. Meanwhile, although cell-free DNA (cfDNA)-based liquid biopsy test has been approved by the U.S. FDA to detect EGFR mutations in the ctDNA of patients with NSCLC who are candidates for targeted therapy with erlotinib and osimertinib (70), further studies are still required to confirm the clinical usefulness of ctDNA as prescribed by ASCO (71).

Several studies evaluated the possibility of ctDNA as a screening device for tumor progression. A recent study presented that in a high-risk population of 1493 enrolled patients in a prospective cohort study, a single ctDNA methylation marker, cg10673833, revealed distinguished diagnostic accuracy, with the sensitivity of 89.7%, and specificity of 86.8% for the finding of CRC and precancerous lesions (72). The promoter methylation of APC and RASSF1A in cfDNA was illustrated as frequent epigenetic results in patients with operable GC at an early stage (73). In HCC, when using NGS technology with a panel of regularly altered genes, in a prospective cohort of 30 patients, the ctDNA detection rate reached 63% with stage A based on the Barcelona Clinic Liver Cancer score (74). 81% of concordance rate was obtained between tissue and liquid biopsy. Distinguishment of HCC specimens from control cirrhotic and not cirrhotic tissue samples was reported with a specificity of 95% by a combination of five aberrant methylation biomarkers (75). Mutations of exons 9, 11, 13, and 17 of KIT, and exons 12, 14, and 18 of PDGFRA are important drivers of oncogenesis and exist in around 85–90% of gastrointestinal stromal tumors. Hence, the main part of the studies assessing the usefulness of ctDNA in gastrointestinal stromal tumors was focused on KIT and PDGFRA alterations (76).

In GI cancers, evaluation of minimal residual disease (MRD) through the study of ctDNA is not still defined, but has already been assessed in diverse analyses. In the TRACC study (NCT04050345) designed on stage II–III CRC, 6 out of 14 (43%) MRD-positive patients recurred whereas only 8 out of 93 (9%) MRD-negative patients did.

The TRACC study (NCT04050345) demonstrated that 6 out of 14 (43%) MRD-positive patients with stage II–III CRC recurred whereas only 8 out of 93 (9%) MRD-negative patients did. The most meaningful prognostic factor related with recurrence-free survival was shown to be ctDNA status. CRC patients at high risk of recurrence and who will really receive benefit from adjuvant therapy may be identified (77). ctDNA measurements provide the capability to guide surveillance while detecting latent candidates for escalated or de-escalated adjuvant therapy approaches in resected, stage I–III CRC. A report at the conference in ESMO 2021 evaluated somatic tissue mutations using MSK-IMPACT, and ctDNA utilizing Guardant360, FoundationOne, or MSK-ACCESS. ctDNA identification predicted the risk of recurrence in resected MSI-high patients and evaluated the effect of ICI (atezolizumab) on these MRD positive patients (NCT03832569). Meanwhile, the retrospective CORRECT trial, analysis of ctDNA, could predict the clinical utility of regorafenib and evaluate the survival in mCRC patients (78). Recently, the CIRCULATE-Japan trial, which involved a prospective nationwide patient-screening registry named GALAXY using the Signatera ctDNA assay, reported preliminary findings (79). The sample size of this observational study is 5,000 and 301 patients had clinical stages I, II, and III CRC with preoperative ctDNA identified in 50 (77%), 267 (95%), and 288 (96%) patients, respectively. Interestingly, ctDNA-positive status at 4 weeks showed a negative correlation with survival despite the association with RAS, BRAF V600E, and MSI status were not demonstrated. Notably, 99% of patients with ctDNA-negative clinical stage I–III survive for the postoperative 6-months.

Liquid biopsy is nearly ready to be approved not only for diagnosis but also for monitoring the acquisition of resistance to therapy in real-time due to its minimal invasiveness and easy collection. For example, in clinical and preclinical studies, RAS mutant clones have been elucidated as drivers of acquired resistance to anti-EGFR treatment (80). The appearance of acquired RAS mutations and alterations in other genes, such as ERBB2, MET, FLT3, MEK, and EGFR was suggested by an extensive observation of ctDNA using a ctDNA assay based on NGS during anti-EGFR treatment (81). In metastatic HER2-positive gastro-esophageal cancer, a longitudinal surveillance of serial plasma samples utilizing a ctDNA assay demonstrated that to be correlated to the resistance to trastuzumab in patients treated with trastuzumab in addition to chemotherapy (82). The CRICKET phase II study, the first prospective trial evaluating the efficacy of rechallenge approach with cetuximab and irinotecan, displayed the benefit in RAS/BRAF wild-type mCRC patients with acquired resistance to cetuximab. RAS mutation was not identified in patients who partially responded to treatment (83,84). Numerous clinical studies are currently assessing the role of liquid biopsy in anti-EGFR rechallenge (CHRONOS, NCT03227926; RASINTRO, NCT03259009) (83). We summarize the clinical relevance of liquid biopsy with ctDNA in GI cancer in Fig. 2.

In the near future, liquid biopsies with ctDNA will be essential for GI cancer treatment. A recent study showed that ctDNA analysis significantly reduced the screening period and improved the study enrollment rate compared with sequencing of tumor specimens in GI cancer. Collectively, ctDNA was found in 91.4% of patients (85). Besides, liquid biopsy permits the collection of repetitive samples during the course of the patient's therapy and the collection of clones that show resistance to ongoing treatment. ctDNA as circulating biomarkers can assess the response to ongoing treatments, thus rapidly guiding the medical choice for further chemotherapy regimen and the requirement to switch treatment strategy. Thus, the application of ctDNA-based analysis may provide great benefits in supporting clinical decision-making and improving patient prognosis, which may lead to personalized medicine.

Hereditary gastrointestinal cancer

In addition to the main purpose of predicting the drug effects, genetic testing may result in findings regarding germline variants (secondary findings). Multi-gene panel testing has been increasingly required and become broadly available in the research of hereditary cancer syndromes (86). Analysis of secondary findings has been discussed, and the results will be disclosed if the patient wishes to reveal them after discussing whether they should be disclosed in the expert panel. For patients and their families identified at risk by genetic testing, strategies for rigorous screening and risk-decreasing approaches for cancer prevention are important in their outcomes. They can accept genetic counseling consisting of medical geneticists and genetic counselors. Nowadays, guidelines state that patients with suspected hereditary CRC and PC should receive genetic counseling and be offered comprehensive genetic testing (87). The National Comprehensive Cancer Network guideline (88) plans a series of clinical outlines the way to approve multi-gene panel testing: i) when personal medical and/or family cancer history meets criteria for more than one hereditary cancer syndrome ii) when family cancer history does not meet established testing guidelines, but consideration of inherited cancer risk persists iii) in individuals concerned about cancer predisposition for whom family cancer history is limited or unknown. Hereditary breast and ovarian cancer have been investigated extensively by utilizing a multi-gene panel. It consists of genes as BRCA1, BRCA2, PALB2, ATM, MLH1, MSH2, MSH6, TP53, CHEK2, STK11 and PTEN (89). A sensitivity to GI cancer was distributed among these genes, which led to personalized treatment and follow-up (88,89).

In GI cancers, the major organs involved with inherited cancer are the colon, pancreas, and stomach. Table II provides an overview of hereditary GI cancers, along with their genetic cause, cancer risks, and drug sensitivity.

Table II.

Characteristic feature of hereditary gastrointestinal cancers.

Table II.

Characteristic feature of hereditary gastrointestinal cancers.

DiseaseCausative genesInheritance traitGastrointestinal tumors (lifetime cancer risk)Other malignanciesDrug sensitivity
Lynch syndromeMLH1, MSH2,ADCRC (22–74%)Endometrium, ovary,ICI,
MSH6, PMS2, GC (11–19%)uterus, brain
EPCAM Small bowel cancer (1–4%)
PC (3–4%)
HCC and bile tract
cancer (2–7%)
FAPAPCADCRC (100 %)Thyroid, adrenalNSAID
Duodenum and ampullarygland, brain
(4–12%)
GC (<1%)
PC (2%)
JPSSMAD4,ADCRC (39%)NoneN/A
BMPR1A GC, PC, Small bowel
cancer (21%)
PJSSTK11ADCRC (39%)Breast, lung, ovary,N/A
GC (29%)uterus, testis, cervix
PC (11–36%)
Small bowel cancer (13%)Sarcoma, breast, adrenalN/A
LFSTP53ADCRC (12.5%)gland brain, lung
GC (4.8%)
MUTYH-associatedMUTYHARCRC (40–100%)Thyroid,N/A
polyposis Duodenum (4%)
Cowden syndromePTENADCRC (9–16%)Breast, thyroid,N/A
endometrium, brain,
kidney
HDGCCDH1ADGC (70–80%)BreastN/A
HBOSBRCA1, BRCA2,ADPC (1–7%)Breast, ovary,PPAP
PALB2 prostate, skininhibitor
FAMMMCDKM2AADPC (17%)Skin (melanoma),N/A
lung, larynx, breast

[i] CRC, colorectal cancer; GC, gastric cancer; PC, pancreatic cancer; FAP, familial adenomatous polyposis; JPS, Juvenile polyposis syndrome; PJS, Peutz-Jeghers syndrome; LFS, Li-Fraumeni syndrome; HDGC, Hereditary diffuse gastric cancer; HBOS, hereditary breast ovarian cancer; FAMMM, familial atypical multiple mole melanoma syndrome; AD, autosomal dominant inheritance; AR, autosomal recessive inheritance; NSAIDs, non-steroidal anti-Inflammatory drugs; PARP, poly ADP ribose polymerase; N/A, not applicable.

Colorectal cancer

Lynch syndrome (LS) is one of the most familiar hereditary cancer syndromes that is caused by germline pathogenic variants in DNA MMR, including EPCAM, MLH1, MSH2, MSH6, and PMS2 (90). Families with LS have a high risk of developing colorectal, small intestine, ureteral, urological, endometrial, ovarian, and hepatobiliary cancer, and are prone to progress cancer at a youthful age. The risk of advancing cancer in LS varies according to the causal gene (91). MSI testing is recommended by the American guidelines for all CRC patients with newly diagnosed CRC to find LS patients (92). When a pathogenic germline variant in MMR genes is detected by following genetic testing, LS is diagnosed. Microsatellite regions are involved in various genes contained in cancer initiation, and the accumulation of aberrations in these regions caused MSI-H. GI cancers with MSI-H are remarkably sensitive to ICI, suggesting that ICI should be efficient in LS (93).

Pancreatic cancer

Hereditary cancers caused by germline pathogenic variants are present in approximately 5–10% of PC. Individuals with at least one first-degree relative (FDR) with PC are at higher risk (OR 1.76). Patients with no less than one FDR with PC have elevated risk. The more FDR additionally increases this risk. If a family involves two concerned FDRs, the colleagues of this family are identified as FPC kindreds. Risk elevates promptly depending on the number of affected family members; 4.6-fold with one, 6.4-fold with two, and 32-fold with three affected FDRs (94). Well-defined genetic cancer sensitivity syndromes correlated with PC clarify a minority of this familial accumulation, as shown in Table II. In recent studies using gene-panel testing, some PCs harbor actionable BRCA1/2 pathogenic or likely pathogenic variants (0–3% for BRCA1 and 1–6% for BRCA2) were presented (9597). While screening every person for PC is expensive due to the comparatively low occurrence of this disease and the deficiency of precise, and noninvasive screening methods, screening may have significance for patients who reveal elevated risk (98,99). Genetic testing seems to be worthwhile for patients with an increased risk of carrying a pancreas-related cancer susceptibility gene.

Gastric cancer

Hereditary Diffuse Gastric Cancer (HDGC is characterized by a high prevalence of diffuse-type GC in the family lineage. HDGC is an autosomal dominant inheritance caused by a germline CDH1 mutation encoding the adhesion molecules E-cadherin. In Western countries, approximately 40% of HDGC families are shown to have germline mutations in CHD1 (100). Genetic testing is recommended for HDGC candidates because it involves multiplex ligation-dependent probe amplification is recommended for HDGC candidates. Truncating mutations in CDH1 and CTNNA1 are thought to be responsible for this syndrome. Recently, exome sequencing identified germline mutations of some related genes, such as INSR, FBXO24, MAP3K6, PALB2, RAD51C, MET, and DOT1L as other latent candidate genes for HDGC (101).

The increasing use of multigene panel testing has redefined gene-cancer associations, and consecutively, cancer risk assesses that penetrance values range from low to high. Cancer screening approvals and preventive strategies adapted by germline mutation will enable us to improve clinical prognosis for patients at greatest risk of cancer and their kindreds.

Comprehensive genetic analysis

As mentioned above, standards of cancer molecular diagnostics, including multi-gene panels have been launched and developed in clinical settings. In contrast, these tests cover only a certain number of associated genomic alterations in coding regions of the genome. Because cancer genomes evolve in a while, it is recommended to utilize comprehensive NGS techniques over restricted-gene tests. Recent advances in NGS as large-scale sequencing technology allow one to investigate the entire genome (WGS), the exons within all known genes (whole exome sequencing, WES), or total RNA (whole transcriptome sequencing) (102). WGS is theoretically straightforward. DNA is randomly fragmented by physical shearing, and 30–50× sequence depth (90–150 Gb) of the individual human whole genome is ordinarily sequenced for both cancer and normal genomes, which result in comprise 99% of the total human genome (103). WGS strategies can identify unexplored mutations, such as untranslated regions, introns, promoters, non-coding functional RNA, and mitochondrial genomes, as well as coding mutations and somatic copy number alterations. WGS also provides a range of diagnostic significance, including new detection in rare cancer mutations (104). WGS analysis will enable us to clarify the functions of these unknown genomic regions and further understand the whole landscape of cancer genomes (105). The comprehensive genetic testing for GI cancer now being examined in clinical studies is reviewed in Table III.

Table III.

Ongoing clinical trials of gastrointestinal cancer classified on comprehensive entire genetic testing.

Table III.

Ongoing clinical trials of gastrointestinal cancer classified on comprehensive entire genetic testing.

Sequencing/NCT numberType of trialClinical purposeResultsDetection methodComments
WGS
  NCT02759657 (COMPASS)CohortDiagnosticActive, not recruitingTissueComprehensive molecular characterization of PDAC for better treatment selection
  NCT03254121 (HEPCASUS)CohortDiagnosticCompletedTissueGenome studies of HCC developed in hepatitis C patients with sustained virological response
  NCT03718897 Cohort/prospectivePrognosticRecruitingTissueIdentification of prognostic gene Mutations in biliary tract cancer Using WGS
  NCT04597710 Cohort/prospective Diagnostic/predictiveRecruitingTissueUtility of WGS to aid clinical decision making in patients referred for liver resection
  NCT05242237 Cohort/prospectivePrognosticRecruitingBloodThe prognostic value of CTC isolated by a novel microfluidic platform in liver cancer patients
WES
  NCT04694391 Case-Control/prospectiveDiagnosticRecruitingTissue/bloodGenomic study of relapse EC after radiotherapy
  NCT02127359 Cohort/prospectiveDiagnosticCompletedTissueThe study is to perform WES on cancer cells and normal tissues to develop better ways to treat and prevent cancers
  NCT03486574 Family-based/prospectiveDiagnosticEnrolled by invitationBloodResearch for associated genes for developing GC in family member with first-degree relatives
  NCT03982173 (MATILDA)Single GroupTherapeuticActive, not recruitingTissueA phase II WES-based basket trial for combination therapy with durvalumab and tremelimumab in patients with metastatic solid tumors
  NCT03108885 Case-Only/prospectivePredictiveEnrolled by invitationTissue/bloodMeasuring cfDNA during the course of treatment for EC as a marker of response and recurrence
  NCT04955808 Case-Only/prospectiveDiagnosticRecruitingTissue/bloodThe utility of biospecimen collection in identifying genetic changes in patients with solid tumors or multiple myeloma undergoing surgery
  NCT02851004Single GroupTherapeuticTerminatedTissue/bloodThe efficacy and safety of BBI608 in combination with pembrolizumab in mCRC
  NCT05048524Single GroupDiagnosticRecruitingTissue/bloodThe feasibility of SLOG regimen in patients with localized PC.
  NCT03832621 (MAYA study)Single Group, open labelDiagnosticActive, not recruitingTissue/bloodThe efficacy and safety of nivolumab, ipilimumab and temozolomide combination in patients with MSS, MGMT-silenced mCRC.
  NCT03023436Single Group, open label Diagnostic/therapeuticRecruitingTissue/bloodThe survival benefit and safety of cytoreductive surgery combined with Hyperthermic Intraperitoneal Chemotherapy and chemotherapy in gastric cancer with peritoneal metastasis.
WTS
  NCT03886571 Cohort/prospectiveDiagnosticRecruitingTissue/bloodAn observational, biospecimen collection protocol to develop a bank of pancreatic cancer tissue and normal tissue.
  NCT03573791 Case-control/prospectiveDiagnosticRecruitingTissueThe purpose of this trail is to identify the biomarkers to predict resistance to neoadjuvant therapy.
  NCT03840460 Cohort/prospectiveDiagnosticRecruitingTissue/bloodA study in PDAC to enable further disease characterization and the development of predictive and prognostic biomarkers
  NCT04249739Non-RandomizedTherapeuticRecruitingTissuePembrolizumab + capecitabin/oxaliplatin or pembrolizumab + trastuzumab + capecitabine/cisplatin in GC
  NCT02015169Single GroupTherapeuticCompletedTissuePhase II study of neoadjuvant XELOX + Lapatinib in HER2- positive GC patients with liver metastasis
  NCT03841799 (COLON-IM) Cohort/prospectiveDiagnosticRecruitingTissueAssessment of colorectal tissue microenvironment (neutrophils infiltrate) of patients with benign or malignant colorectal lesion
  NCT03260712Single GroupPredictiveActive, not recruitingTissueEvaluation of pathological predictive factors for response and toxicity which are responsible for chemotherapy and pembrolizumab.
  NCT04554771 (BASALT)RandomizedTreatmentRecruitingBloodBlood-borne assessment of stromal activation in EC to guide tocilizumab therapy

[i] WGS, whole genome analysis; WES, whole exome analysis; WTS, whole transcriptome analysis; CRC, colorectal cancer; GC, gastric cancer; PC, pancreatic cancer; EC, esophageal cancer; CTC, circulating tumor cell; N/A, not applicable.

A recent genomic-based study of glioblastoma patients examined the usefulness of WGS/RNA-seq vs. targeted panels (106). WGS/RNA-seq detected more conceivably criminal clinical findings than targeted panels in 90% of cases, with an average of 16-fold more unique conceivably criminal variants identified for each patient. In PC, WES of germline DNA from whole blood of Japanese familial pancreatic cancer patients revealed novel germline susceptibility genes, FAT1 and FAT4, which encode the large transmembrane proteins protocadherins (107). Thus, WES for PC patients would offer significant information about high-risk pathogenic germline variants in hereditary cancer syndromes. A study focusing on rare genetic variants using WGS through analysis of heterozygous premature truncating variants showed that 20 significant genes, including PALD1, LRP1B, COL4A2, CYLC2, ZFYVE9, BRD3, AHDC1 were identified, which would play an important role in risk prediction of high-risk patients in families identified at risk (108).

In CRC, a novel tumor suppressor, ARID2 was detected based on WES analysis of younger patients (109). Substantial augmentation for mutations in 4 out of 23 coding and 12 out of 15 noncoding driver genes was shown in the mCRC cohort compared with primary CRC by using WES. Mutations in PIK3CA were significantly reduced in mCRC among detected putative drivers (110). Six of the newly found coding driver genes, ZFP36L2, BCL, BCL9L, ELF3, LMTK3, and TGIF1 are not detected in the CRC-specific MSK-IMPACT panel. Similarly, WGS analysis of metastatic vs. matched primary colorectal lesions, 65% of somatic mutations originate from a common progenitor, with 15% being tumor- and 19% metastasis-specific (111). Both primary- and metastasis-specific mutations maintain high levels of BRCAness. Recurrently mutated non-coding elements such as ncRNAs RP11-594N15.3, AC010091, SNHG14, 3′ UTRs of FOXP2, DACH2, TRPM3, XKR4, ANO5, CBL, CBLB and efferocytosis-/PD-L1were identified. Numerous metastasis-specific mutations were detected, including non-silent mutations of FAT1, FGF1, BRCA2, TP53, and KDR, splice site mutations of JAK2 and 3′-UTR mutations in KDR, PDGFRA, and AKT2 genes, suggesting the existence of a high degree of mutational discordance between metastatic and primary tumor (111). An original dataset containing whole genomes analysis from 60 single-cell collecting samples before therapy and after metastatic relapse resection demonstrated that three non-synonymous and one stop codon mutations specific to the recurrent lineage in four different genes, PKHD1, PCDHB15, CSF1R, and CC2D1B, were detected in CRC patients. Moreover, a distinctive mutagenic prototype distinguishing the cancer cells from the recurrent lesion illustrated by a substantial contribution of COSMIC signatures SBS35 and SBS17b was identified (112).

In esophageal cancer (EC), somatic mutations and copy number alterations in multiple chromosome segments, encoding MYC on 8q24.21, PIK3CA and SOX2 on 3q26, CCND1, SHANK2, CTTN on 11q13.3, and KRAS on 12p12 were detected using WES. Amplifications of EGFRvIII and EGFRvIVa mutants were identified, representing a novel finding in African-American EC that may lead to clinical practice (113). WGS can lead to the detection of novel treatment targets and the discovery of new genomic biomarkers, which may eventually develop the treatment modalities for patients with GI tumors.

Discussion and future perspective

In this review, we focused on the latest advances in genetic testing for the diagnosis and management of GI cancer. With the introduction of Sanger sequencing and polymerase chain reaction, laboratory genetic testing became an important instrument for the genomic profiling of cancers in clinical settings. However, although a variety of genes that are mutated in GI malignancies are known, none of the mutations has had clear actionability, and DNA analyses of GI cancers were not a part of clinical oncology until relatively recently. The concept of massively parallel sequencing led to the development of multi-gene panels, that cover the entire spectrum of all acknowledged targeted genes and assist in selecting a useful therapy (5). Studies of germline variants that contribute to cancer predisposition now help detect individuals who have a high-risk for some heritable cancers. Multi-gene panel testing has the capacity to provide significant advances in daily oncology practice. However, there are still several obstacles to be addressed before multigene panel testing can be effectively applied to patients with GI cancer.

One of the major issues regarding the use of multi-gene panel for precision medicine is the lack of appropriate treatment. The proportion of GI cancers that have clearly actionable genetic alterations is comparatively low, and there is no gene-tailored therapy for the majority of patients with GI cancer. From a translational viewpoint, the persistent success of comprehensive genetic testing will depend principally on the testing's clinical utility and ability to identify the treatable targets. To increase the number of patients whose tumors can be successfully treated, it would be indispensable to use strategies such as large-scale analyses in preclinical settings to increase our understanding of the biological processes driving cancer and to identify biomarkers for cancer diagnostics and new drugs. Comprehensive genome profiling might represent one of these strategies, and the continued progress in such profiling may lead to genetic testing as a first option in the treatment of GI cancers.

Liquid biopsies are an ideal sample source that reflects individual characteristics and the heterogeneity of GI cancers. The widespread clinical applications of ctDNA-based assays for therapy decision-making and monitoring of tumors are based on promising preliminary findings, but many challenges remain. The ctDNA levels in plasma are prone to be inconsistent and low, causing a variety of detection thresholds. Additionally, a negative ctDNA finding may be attributed to low copy number identification instead of the absence of ctDNA. Hence, the restricted sensitivity of a ctDNA examination is an essential issue in patients who have early-stage cancer as well as a lower level of plasma ctDNA. The low level of ctDNA in the plasma may require the usage of ultrahigh-depth sequencing and sophisticated statistical models for the purpose of decreasing background error rates for very low variant allele frequencies (114). False-positive ctDNA results can also be caused by DNA fragments from the clonal hematopoiesis of indeterminate potential or non-neoplastic hematopoietic stem cells can be reduced by conducting an advanced bioinformatics analysis or by comparing the results of ctDNA sequencing with the findings obtained from leukocytes and/or matched tumor tissues (115). A high-intensity cfDNA sequencing analysis based on the combination of cfDNA and white blood cell DNA analysis provides both the de novo detection of tumor-derived changes and the clarification of MSI, the TMB, mutational profiles, and the sources of somatic mutations identified in cfDNA (115). A quantitative ctDNA evaluation and methylation uncovering may increase the specificity of ctDNA identification and consequently allow to distinguish benign from cancerous GI disease, even at early tumor stages. Further explorations by a large number of clinical trials are necessary for the standardization of the detection process as well as the clinical application of liquid biopsies.

Among the multiple technical platforms that are now available, the WGS strategy is now the most effective way to construct a comprehensive image of the genomic variation in a tumor. The widespread use of WGS technologies in clinical settings seems no longer a distant dream, but the application of WGS strategy possesses tremendous challenges in light of the sequencing costs, computational processing, long-term storage, and meaningful biological interpretation. Moreover, WGS needs particular ethical and regulatory frameworks to handle accidental and secondary genomic detections in the germline. However, in light of the estimation that the costs of sequencing will result in the historic descending tendency, a more gradual approval of WGS approaches for a more improved stratification and subtyping of rare tumors may be attainable in the short period. Algorithms that can dependably support the latent significance of new genetic issues and then associate these issues to theoretical or assumed clinical activity with limited manual interference are required. The advancement of those algorithms will be essential for decreasing analysis and explanation costs and reducing the turnaround time for clinical strategy The most modern WGS platforms such as Illumina NovaSeq 6000 system can handle a great volume of specimens in comparatively short turnaround times, which makes WGS more practical (116).

Recent advances in cancer research have revealed intratumor heterogeneity at the cell levels, epigenetic profiles, and interferences with the tumor microenvironment. Hence, the incorporation of multiple layers of information for individual cancer cells is crucial for a comprehensive knowledge of the mechanisms of cancer initiation (117). The addition of ‘omics’ to a molecular word suggests a comprehensive, or worldwide evaluation of a set of molecules. A multi-omics study is a data-driven biological analysis in which the data sets are diverse individual omic analyses, such as genomics, epigenomics, transcriptomics, proteomics, metagenomics, and microbiomics that are used to investigate physiological or pathological phenomena and characterize biomolecular systems at different levels. The recent advances in high-throughput technologies for genomics and transcriptomics have resulted in a paradigm shift toward multi-omics investigations, large-scale research collaborations, and the design of computational algorithms (118). Multi-omics studies for GI cancer currently being evaluated in clinical trials are summarized in Table IV. Multi-omics investigations have been applied in a variety of clinical studies for a better detection of clinical subtypes or drug resistance, the prediction of efficient combined therapies, and the exploration of novel biomarkers. For instance, integrated proteogenomic data together with genomic and transcriptomic data of CRCs, which were illustrated by The Cancer Genome Atlas, demonstrated that a chromosome 20q amplicon was correlated with the great inclusive alterations at both messenger RNA and protein levels. In addition, the incorporation of proteomics data provides the detection of important 20q candidates, including HNF4A (hepatocyte nuclear factor 4, alpha), TOMM34 (translocase of outer mitochondrial membrane 34), and SRC (SRC proto-oncogene, nonreceptor tyrosine kinase), suggesting that incorporated proteogenomic analyses will enable novel developments in cancer diagnosis and treatment (119). A study that performed a multi-omics characterization of molecular features of GC, using WGS, WES, and RNA-seq for 35 GC patients before and after their neoadjuvant chemotherapy, showed that C10orf71 was associated with treatment resistance, whereas MYC and MDM2 amplification mutations were associated with treatment sensitivity (120).

Table IV.

Selective ongoing clinical trials of multi-omics study for gastrointestinal cancer.

Table IV.

Selective ongoing clinical trials of multi-omics study for gastrointestinal cancer.

Multi-omics studyType of trialClinical purposePhaseResultsDetection methodComments
NCT02342158 (PERMED-01 trial)Single GroupDiagnosticN/AActive, not recruitingTissue/bloodIdentification molecular alterations to guide individualized treatment in advanced solid tumor
NCT03546127 (MULTIPLI-0) Cohort/prospectiveDiagnosticN/ACompletedTissue/bloodA feasibility study in France to assess sample circuit and to perform analyzes within a limited time in CRC.
NCT03951792 Case-control/prospectiveDiagnosticN/AEnrolled by invitationTissue/stoolTime longitudinal study of the microbiome in CRC
NCT04318834 (COMPASS-B-MUHC)Single Group, open labelDiagnostic RecruitingTissueIdentification of actionable molecular alterations of biliary tract cancer through WTS.
NCT04622423 Cohort/prospectiveDiagnosticN/ARecruitingTissueAdvanced therapies for liver metastasis in CRC and PC.
NCT04871321 Cohort/prospectiveDiagnosticN/ARecruitingTissue/bloodBiomarker discovery in patients within patients with advanced biliary tract cancer who received nab-paclitaxel plus gemcitabine-cisplatin
NCT05234450 Case-only/prospectiveDiagnosticN/ARecruitingTissueIdentification of different tumor subgroups in pancreatic neuroendocrine tumors and carcinomas regardless of their grade and stage.
NCT03429816 (OPPOSITE)Single Group, open labelTherapeuticN/AActive, not recruitingTissueCorrelation of molecular subtypes with histological response after neoadjuvant therapy in patients with EC and GC.

[i] WGS, whole genome analysis; WTS, whole transcriptome analysis; CRC, colorectal cancer; GC, gastric cancer; PC, pancreatic cancer; EC, esophageal cancer; N/A, not applicable.

Conclusion

Multi-gene testing should be widely applied in clinical settings, not only for greater insights into tumor biology but also to drive cancer treatment. New clinical studies should apply multigene testing toward the goal of finding novel targeted therapies. The rapid analysis of genetic alterations with real-time monitoring of therapy responses by ctDNA can optimize new therapeutic strategies. The comprehensive characterization of GI cancers by genetic testing will contribute to a better molecular-level understanding of cancer, and it will contribute to more appropriate and effective genomic-driven therapies for patients who might not benefit from standardized therapy or experimental interferences in the context of clinical studies. Due to several challenges to be resolved such as the costs, restricted sensitivity, and time consumption to carry out, genetic testing should be used when standard therapeutic approaches have been completed at present.

Acknowledgements

Not applicable.

Funding

This study was funded by KAKENHI (Grant-in-Aid for Scientific Research) (grant no. 18H02883).

Availability of data and materials

Not applicable.

Authors' contributions

TM and MY performed the literature research. TM wrote the manuscript and performed the revision and approval of the final version. MY designed research, coordinated and corrected the writing of the paper. Both authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Authors' information

Dr Tasuku Matsuoka (ORCID: 0000-0001-5019-8519); Dr Masakazu Yashiro (ORCID: 0000-0001-5743-7228).

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

GI

gastrointestinal

NGS

next-generation sequencing

FDA

U.S. Food and Drug Administration

MSI

microsatellite instability

TMB

tumor mutational burden

OS

overall survival

PARP

poly (ADP-ribose) polymerase

ICB

immune checkpoint blockade

PC

pancreatic cancer

DDR

DNA damage response

EGFR

epidermal growth factor receptor

MAPK

mitogen-activated protein kinases

CRC

colorectal cancer

mCRC

metastatic CRC

HR

hazard ratio

T-DXd

Trastuzumab-Deruxtecan

HER2

human epidermal growth factor receptor 2

WGS

whole genome sequencing

PDAC

pancreatic ductal adenocarcinoma

HRD

homologous recombination deficiency

ASCO

American Society of Clinical Oncology

PFS

progression-free survival

GC

gastric cancer

FGFR

fibroblast growth factor receptor

HCC

hepatocellular carcinoma

TERT

telomerase reverse transcriptase

ctDNR

circulating tumor DNA

cfDNA

cell-free DNA

MRD

minimal residual disease

LS

Lynch syndrome

FDR

first-degree-relative

HDGC

hereditary diffuse gastric cancer

DGC

diffuse type gastric cancer

WES

whole exome sequencing

EC

esophageal cancer

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Ceasovschih A, Voloc G, Sorodoc V, Vâță D, Lupașcu CD, Preda C, Lionte C, Stoica A, Sîrbu O, Grigorescu ED, et al: From chronic pruritus to neuroendocrine tumor: A case report. Exp Ther Med. 23:1892022. View Article : Google Scholar : PubMed/NCBI

3 

Watanabe M, Baba H, Ishioka C, Nishimura Y and Muto M: Recent advances in diagnosis and treatment for malignancies of the gastrointestinal tract. Digestion. 85:95–98. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Matsuoka T and Yashiro M: Precision medicine for gastrointestinal cancer: Recent progress and future perspective. World J Gastrointest Oncol. 12:1–20. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Nagahashi M, Shimada Y, Ichikawa H, Kameyama H, Takabe K, Okuda S and Wakai T: Next generation sequencing-based gene panel tests for the management of solid tumors. Cancer Sci. 110:6–15. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Bahassi el M and Stambrook PJ: Next-generation sequencing technologies: Breaking the sound barrier of human genetics. Mutagenesis. 29:303–310. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Sicklick JK, Kato S, Okamura R, Schwaederle M, Hahn ME, Williams CB, De P, Krie A, Piccioni DE, Miller VA, et al: Molecular profiling of cancer patients enables personalized combination therapy: The I-PREDICT study. Nat Med. 25:744–750. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Massard C, Michiels S, Ferté C, Le Deley MC, Lacroix L, Hollebecque A, Verlingue L, Ileana E, Rosellini S, Ammari S, et al: High-throughput genomics and clinical outcome in hard-to-treat advanced cancers: Results of the MOSCATO 01 trial. Cancer Discov. 7:586–595. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Cobain EF, Wu YM, Vats P, Chugh R, Worden F, Smith DC, Schuetze SM, Zalupski MM, Sahai V, Alva A, et al: Assessment of clinical benefit of integrative genomic profiling in advanced solid tumors. JAMA Oncol. 7:525–533. 2021.PubMed/NCBI

10 

Pishvaian MJ, Blais EM, Brody JR, Lyons E, DeArbeloa P, Hendifar A, Mikhail S, Chung V, Sahai V, Sohal DPS, et al: Overall survival in patients with pancreatic cancer receiving matched therapies following molecular profiling: A retrospective analysis of the know your tumor registry trial. Lancet Oncol. 21:508–518. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, Aranda Aguilar E, Bardelli A, Benson A, Bodoky G, et al: ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. 27:1386–1422. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Venderbosch S, Nagtegaal ID, Maughan TS, Smith CG, Cheadle JP, Fisher D, Kaplan R, Quirke P, Seymour MT, Richman SD, et al: Mismatch repair status and BRAF mutation status in metastatic colorectal cancer patients: A pooled analysis of the CAIRO, CAIRO2, COIN, and FOCUS studies. Clin Cancer Res. 20:5322–5330. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Jones JC, Renfro LA, Al-Shamsi HO, Schrock AB, Rankin A, Zhang BY, Kasi PM, Voss JS, Leal AD, Sun J, et al: Non-V600 BRAF mutations define a clinically distinct molecular subtype of metastatic colorectal cancer. J Clin Oncol. 35:2624–2630. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Yaeger R, Kotani D, Mondaca S, Parikh AR, Bando H, Van Seventer EE, Taniguchi H, Zhao H, Thant CN, de Stanchina E, et al: Response to anti-EGFR therapy in patients with BRAF non-V600-mutant metastatic colorectal cancer. Clin Cancer Res. 25:7089–7097. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Johnson B, Loree JM, Jacome AA, Mendis S, Syed M, Morris Ii VK, Parseghian CM, Dasari A, Pant S, Raymond VM, et al: Atypical, non-V600 BRAF mutations as a potential mechanism of resistance to EGFR inhibition in metastatic colorectal cancer. JCO Precis Oncol. 3:PO.19.00102. 2019.

16 

Corcoran RB, Ebi H, Turke AB, Coffee EM, Nishino M, Cogdill AP, Brown RD, Della Pelle P, Dias-Santagata D, Hung KE, et al: EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discov. 2:227–235. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Yaeger R, Cercek A, O'Reilly EM, Reidy DL, Kemeny N, Wolinsky T, Capanu M, Gollub MJ, Rosen N, Berger MF, et al: Pilot trial of combined BRAF and EGFR inhibition in BRAF-mutant metastatic colorectal cancer patients. Clin Cancer Res. 21:1313–1320. 2015. View Article : Google Scholar : PubMed/NCBI

18 

van Geel RMJM, Tabernero J, Elez E, Bendell JC, Spreafico A, Schuler M, Yoshino T, Delord JP, Yamada Y, Lolkema MP, et al: A phase Ib dose-escalation study of encorafenib and cetuximab with or without alpelisib in metastatic BRAF-mutant colorectal cancer. Cancer Discov. 7:610–619. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Kopetz S, Guthrie KA, Morris VK, Lenz HJ, Magliocco AM, Maru D, Yan Y, Lanman R, Manyam G, Hong DS, et al: Randomized trial of irinotecan and cetuximab with or without vemurafenib in BRAF-mutant metastatic colorectal cancer (SWOG S1406). J Clin Oncol. 39:285–294. 2021. View Article : Google Scholar : PubMed/NCBI

20 

Corcoran RB, André T, Atreya CE, Schellens JHM, Yoshino T, Bendell JC, Hollebecque A, McRee AJ, Siena S, Middleton G, et al: Combined BRAF, EGFR, and MEK inhibition in patients with BRAFV600E-mutant colorectal cancer. Cancer Discov. 8:428–443. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Roviello G, D'Angelo A, Petrioli R, Roviello F, Cianchi F, Nobili S, Mini E and Lavacchi D: Encorafenib, binimetinib, and cetuximab in BRAF V600E-mutated colorectal cancer. Transl Oncol. 13:1007952020. View Article : Google Scholar : PubMed/NCBI

22 

Grothey A, Fakih M and Tabernero J: Management of BRAF-mutant metastatic colorectal cancer: A review of treatment options and evidence-based guidelines. Ann Oncol. 32:959–967. 2021. View Article : Google Scholar : PubMed/NCBI

23 

Nagasaka M, Li Y, Sukari A, Ou SHI, Al-Hallak MN and Azmi AS: KRAS G12C Game of Thrones, which direct KRAS inhibitor will claim the iron throne? Cancer Treat Rev. 84:1019742020. View Article : Google Scholar : PubMed/NCBI

24 

Wang C and Fakih M: Targeting KRAS in colorectal cancer. Curr Oncol Rep. 23:282021. View Article : Google Scholar : PubMed/NCBI

25 

Stadler ZK, Battaglin F, Middha S, Hechtman JF, Tran C, Cercek A, Yaeger R, Segal NH, Varghese AM, Reidy-Lagunes DL, et al: Reliable detection of mismatch repair deficiency in colorectal cancers using mutational load in next-generation sequencing panels. J Clin Oncol. 34:2141–2147. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Maby P, Tougeron D, Hamieh M, Mlecnik B, Kora H, Bindea G, Angell HK, Fredriksen T, Elie N, Fauquembergue E, et al: Correlation between density of CD8+ T-cell infiltrate in microsatellite unstable colorectal cancers and frameshift mutations: A rationale for personalized immunotherapy. Cancer Res. 75:3446–3455. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Andre T, Amonkar M, Norquist JM, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt CJA, Smith D, Garcia-Carbonero R, et al: Health-related quality of life in patients with microsatellite instability-high or mismatch repair deficient metastatic colorectal cancer treated with first-line pembrolizumab versus chemotherapy (KEYNOTE-177): An open-label, randomised, phase 3 trial. Lancet Oncol. 22:665–677. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Richman SD, Southward K, Chambers P, Cross D, Barrett J, Hemmings G, Taylor M, Wood H, Hutchins G, Foster JM, et al: HER2 overexpression and amplification as a potential therapeutic target in colorectal cancer: Analysis of 3256 patients enrolled in the QUASAR, FOCUS and PICCOLO colorectal cancer trials. J Pathol. 238:562–570. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Wang G, He Y, Sun Y, Wang W, Qian X, Yu X and Pan Y: Prevalence, prognosis and predictive status of HER2 amplification in anti-EGFR-resistant metastatic colorectal cancer. Clin Transl Oncol. 22:813–822. 2020. View Article : Google Scholar : PubMed/NCBI

30 

Sartore-Bianchi A, Trusolino L, Martino C, Bencardino K, Lonardi S, Bergamo F, Zagonel V, Leone F, Depetris I, Martinelli E, et al: Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): A proof-of-concept, multicentre, open-label, phase 2 trial. Lancet Oncol. 17:738–746. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Meric-Bernstam F, Hurwitz H, Raghav KPS, McWilliams RR, Fakih M, VanderWalde A, Swanton C, Kurzrock R, Burris H, Sweeney C, et al: Pertuzumab plus trastuzumab for HER2-amplified metastatic colorectal cancer (MyPathway): An updated report from a multicentre, open-label, phase 2a, multiple basket study. Lancet Oncol. 20:518–530. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Siena S, Di Bartolomeo M, Raghav K, Masuishi T, Loupakis F, Kawakami H, Yamaguchi K, Nishina T, Fakih M, Elez E, et al: Trastuzumab deruxtecan (DS-8201) in patients with HER2-expressing metastatic colorectal cancer (DESTINY-CRC01): A multicentre, open-label, phase 2 trial. Lancet Oncol. 22:779–789. 2021. View Article : Google Scholar : PubMed/NCBI

33 

Hallberg B and Palmer RH: The role of the ALK receptor in cancer biology. Ann Oncol. 27 (Suppl 3):iii4–iii15. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Pietrantonio F, Di Nicolantonio F, Schrock AB, Lee J, Tejpar S, Sartore-Bianchi A, Hechtman JF, Christiansen J, Novara L, Tebbutt N, et al: ALK, ROS1, and NTRK rearrangements in metastatic colorectal cancer. J Natl Cancer Inst. 109:2017. View Article : Google Scholar : PubMed/NCBI

35 

Ruela AL, de Figueiredo EC, de Araújo MB, Carvalho FC and Pereira GR: Molecularly imprinted microparticles in lipid-based formulations for sustained release of donepezil. Eur J Pharm Sci. 93:114–122. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Hosoda W, Chianchiano P, Griffin JF, Pittman ME, Brosens LA, Noë M, Yu J, Shindo K, Suenaga M, Rezaee N, et al: Genetic analyses of isolated high-grade pancreatic intraepithelial neoplasia (HG-PanIN) reveal paucity of alterations in TP53 and SMAD4. J Pathol. 242:16–23. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, Johns AL, Miller D, Nones K, Quek K, et al: Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 518:495–501. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, Mollaee M, Wagner KU, Koduru P, Yopp A, et al: Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun. 6:67442015. View Article : Google Scholar : PubMed/NCBI

39 

Sohal DPS, Kennedy EB, Cinar P, Conroy T, Copur MS, Crane CH, Garrido-Laguna I, Lau MW, Johnson T, Krishnamurthi S, et al: Metastatic pancreatic cancer: ASCO guideline update. J Clin Oncol. 38:3217–3230. 2020. View Article : Google Scholar : PubMed/NCBI

40 

Robson M, Im SA, Senkus E, Xu B, Domchek SM, Masuda N, Delaloge S, Li W, Tung N, Armstrong A, et al: Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med. 377:523–533. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Del Campo JM, Matulonis UA, Malander S, Provencher D, Mahner S, Follana P, Waters J, Berek JS, Woie K, Oza AM, et al: Niraparib maintenance therapy in patients with recurrent ovarian cancer after a partial response to the last platinum-based chemotherapy in the ENGOT-OV16/NOVA trial. J Clin Oncol. 37:2968–2973. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Curtin NJ and Szabo C: Poly(ADP-ribose) polymerase inhibition: Past, present and future. Nat Rev Drug Discov. 19:711–736. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Park W, Chen J, Chou JF, Varghese AM, Yu KH, Wong W, Capanu M, Balachandran V, McIntyre CA, El Dika I, et al: Genomic methods identify homologous recombination deficiency in pancreas adenocarcinoma and optimize treatment selection. Clin Cancer Res. 26:3239–3247. 2020. View Article : Google Scholar : PubMed/NCBI

44 

Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, Park JO, Hochhauser D, Arnold D, Oh DY, et al: Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N Engl J Med. 381:317–327. 2019. View Article : Google Scholar : PubMed/NCBI

45 

Hu ZI, Shia J, Stadler ZK, Varghese AM, Capanu M, Salo-Mullen E, Lowery MA, Diaz LA Jr, Mandelker D, Yu KH, et al: Evaluating mismatch repair deficiency in pancreatic adenocarcinoma: Challenges and recommendations. Clin Cancer Res. 24:1326–1336. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Latham A, Srinivasan P, Kemel Y, Shia J, Bandlamudi C, Mandelker D, Middha S, Hechtman J, Zehir A, Dubard-Gault M, et al: Microsatellite instability is associated with the presence of lynch syndrome pan-cancer. J Clin Oncol. 37:286–295. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, Lordick F, Ohtsu A, Omuro Y, Satoh T, et al: Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet. 376:687–697. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Takegawa N, Tsurutani J, Kawakami H, Yonesaka K, Kato R, Haratani K, Hayashi H, Takeda M, Nonagase Y, Maenishi O and Nakagawa K: [fam-] trastuzumab deruxtecan, antitumor activity is dependent on HER2 expression level rather than on HER2 amplification. Int J Cancer. 145:3414–3424. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Shitara K, Bang YJ, Iwasa S, Sugimoto N, Ryu MH, Sakai D, Chung HC, Kawakami H, Yabusaki H, Lee J, et al: Trastuzumab deruxtecan in previously treated HER2-positive gastric cancer. N Engl J Med. 382:2419–2430. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Helsten T, Elkin S, Arthur E, Tomson BN, Carter J and Kurzrock R: The FGFR landscape in cancer: Analysis of 4,853 tumors by next-generation sequencing. Clin Cancer Res. 22:259–267. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Hur JY, Chao J, Kim K, Kim ST, Kim KM, Klempner SJ and Lee J: High-level FGFR2 amplification is associated with poor prognosis and lower response to chemotherapy in gastric cancers. Pathol Res Pract. 216:1528782020. View Article : Google Scholar : PubMed/NCBI

52 

Catenacci DVT, Rasco D, Lee J, Rha SY, Lee KW, Bang YJ, Bendell J, Enzinger P, Marina N, Xiang H, et al: Phase I escalation and expansion study of bemarituzumab (FPA144) in patients with advanced solid tumors and FGFR2b-selected gastroesophageal adenocarcinoma. J Clin Oncol. 38:2418–2426. 2020. View Article : Google Scholar : PubMed/NCBI

53 

Catenacci DV, Tesfaye A, Tejani M, Cheung E, Eisenberg P, Scott AJ, Eng C, Hnatyszyn J, Marina N, Powers J and Wainberg Z: Bemarituzumab with modified FOLFOX6 for advanced FGFR2-positive gastroesophageal cancer: FIGHT Phase III study design. Future Oncol. 15:2073–2082. 2019. View Article : Google Scholar : PubMed/NCBI

54 

Nault JC, Mallet M, Pilati C, Calderaro J, Bioulac-Sage P, Laurent C, Laurent A, Cherqui D, Balabaud C and Zucman-Rossi J: High frequency of telomerase reverse-transcriptase promoter somatic mutations in hepatocellular carcinoma and preneoplastic lesions. Nat Commun. 4:22182013. View Article : Google Scholar : PubMed/NCBI

55 

Schulze K, Imbeaud S, Letouzé E, Alexandrov LB, Calderaro J, Rebouissou S, Couchy G, Meiller C, Shinde J, Soysouvanh F, et al: Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat Genet. 47:505–511. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Jhunjhunwala S, Jiang Z, Stawiski EW, Gnad F, Liu J, Mayba O, Du P, Diao J, Johnson S, Wong KF, et al: Diverse modes of genomic alteration in hepatocellular carcinoma. Genome Biol. 15:4362014. View Article : Google Scholar : PubMed/NCBI

57 

Cancer Genome Atlas Research Network. Electronic address, . simplewheeler@bcm.edu; Cancer Genome Atlas Research Network: Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell. 169:1327–1341.e23. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Xu Z, Hu J, Cao H, Pilo MG, Cigliano A, Shao Z, Xu M, Ribback S, Dombrowski F, Calvisi DF and Chen X: Loss of Pten synergizes with c-Met to promote hepatocellular carcinoma development via mTORC2 pathway. Exp Mol Med. 50:e4172018. View Article : Google Scholar : PubMed/NCBI

59 

Jin M, Piao Z, Kim NG, Park C, Shin EC, Park JH, Jung HJ, Kim CG and Kim H: p16 is a major inactivation target in hepatocellular carcinoma. Cancer. 89:60–68. 2000. View Article : Google Scholar : PubMed/NCBI

60 

Kudo M, Finn RS, Qin S, Han KH, Ikeda K, Piscaglia F, Baron A, Park JW, Han G, Jassem J, et al: Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: A randomised phase 3 non-inferiority trial. Lancet. 391:1163–1173. 2018. View Article : Google Scholar : PubMed/NCBI

61 

Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, Kudo M, Breder V, Merle P, Kaseb AO, et al: Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med. 382:1894–1905. 2020. View Article : Google Scholar : PubMed/NCBI

62 

Bruix J, Qin S, Merle P, Granito A, Huang YH, Bodoky G, Pracht M, Yokosuka O, Rosmorduc O, Breder V, et al: Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 389:56–66. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Abou-Alfa GK, Meyer T, Cheng AL, El-Khoueiry AB, Rimassa L, Ryoo BY, Cicin I, Merle P, Chen Y, Park JW, et al: Cabozantinib in patients with advanced and progressing hepatocellular carcinoma. N Engl J Med. 379:54–63. 2018. View Article : Google Scholar : PubMed/NCBI

64 

Valle J, Wasan H, Palmer DH, Cunningham D, Anthoney A, Maraveyas A, Madhusudan S, Iveson T, Hughes S, Pereira SP, et al: Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N Engl J Med. 362:1273–1281. 2010. View Article : Google Scholar : PubMed/NCBI

65 

Lamarca A, Palmer DH, Wasan HS, Ross PJ, Ma YT, Arora A, Falk S, Gillmore R, Wadsley J, Patel K, et al: Second-line FOLFOX chemotherapy versus active symptom control for advanced biliary tract cancer (ABC-06): A phase 3, open-label, randomised, controlled trial. Lancet Oncol. 22:690–701. 2021. View Article : Google Scholar : PubMed/NCBI

66 

Javle M, Bekaii-Saab T, Jain A, Wang Y, Kelley RK, Wang K, Kang HC, Catenacci D, Ali S, Krishnan S, et al: Biliary cancer: Utility of next-generation sequencing for clinical management. Cancer. 122:3838–3847. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Nakamura Y and Yoshino T: Clinical utility of analyzing circulating tumor DNA in patients with metastatic colorectal cancer. Oncologist. 23:1310–1318. 2018. View Article : Google Scholar : PubMed/NCBI

68 

Nakamura Y and Shitara K: Development of circulating tumour DNA analysis for gastrointestinal cancers. ESMO Open. 5 (Suppl 1):e0006002020. View Article : Google Scholar : PubMed/NCBI

69 

Parikh AR, Van Seventer EE, Siravegna G, Hartwig AV, Jaimovich A, He Y, Kanter K, Fish MG, Fosbenner KD, Miao B, et al: Minimal residual disease detection using a plasma-only circulating tumor DNA assay in patients with colorectal cancer. Clin Cancer Res. 27:5586–5594. 2021. View Article : Google Scholar : PubMed/NCBI

70 

Kwapisz D: The first liquid biopsy test approved. Is it a new era of mutation testing for non-small cell lung cancer? Ann Transl Med. 5:462017.PubMed/NCBI

71 

Merker JD, Oxnard GR, Compton C, Diehn M, Hurley P, Lazar AJ, Lindeman N, Lockwood CM, Rai AJ, Schilsky RL, et al: Circulating tumor DNA analysis in patients with cancer: American society of clinical oncology and college of american pathologists joint review. J Clin Oncol. 36:1631–1641. 2018. View Article : Google Scholar : PubMed/NCBI

72 

Luo H, Zhao Q, Wei W, Zheng L, Yi S, Li G, Wang W, Sheng H, Pu H, Mo H, et al: Circulating tumor DNA methylation profiles enable early diagnosis, prognosis prediction, and screening for colorectal cancer. Sci Transl Med. 12:eaax75332020. View Article : Google Scholar : PubMed/NCBI

73 

Balgkouranidou I, Matthaios D, Karayiannakis A, Bolanaki H, Michailidis P, Xenidis N, Amarantidis K, Chelis L, Trypsianis G, Chatzaki E, et al: Prognostic role of APC and RASSF1A promoter methylation status in cell free circulating DNA of operable gastric cancer patients. Mutat Res. 778:46–51. 2015. View Article : Google Scholar : PubMed/NCBI

74 

Ng CKY, Di Costanzo GG, Tosti N, Paradiso V, Coto-Llerena M, Roscigno G, Perrina V, Quintavalle C, Boldanova T, Wieland S, et al: Genetic profiling using plasma-derived cell-free DNA in therapy-naïve hepatocellular carcinoma patients: A pilot study. Ann Oncol. 29:1286–1291. 2018. View Article : Google Scholar : PubMed/NCBI

75 

Kisiel JB, Dukek BA, V S R Kanipakam R, Ghoz HM, Yab TC, Berger CK, Taylor WR, Foote PH, Giama NH, Onyirioha K, et al: Hepatocellular carcinoma detection by plasma methylated DNA: Discovery, phase I pilot, and phase II clinical validation. Hepatology. 69:1180–1192. 2019. View Article : Google Scholar : PubMed/NCBI

76 

Maleddu A, Pantaleo MA, Nannini M and Biasco G: The role of mutational analysis of KIT and PDGFRA in gastrointestinal stromal tumors in a clinical setting. J Transl Med. 9:752011. View Article : Google Scholar : PubMed/NCBI

77 

Larribère L and Martens UM: Advantages and challenges of using ctDNA NGS to assess the presence of minimal residual disease (MRD) in solid tumors. Cancers (Basel). 13:56982021. View Article : Google Scholar : PubMed/NCBI

78 

Tabernero J, Lenz HJ, Siena S, Sobrero A, Falcone A, Ychou M, Humblet Y, Bouché O, Mineur L, Barone C, et al: Analysis of circulating DNA and protein biomarkers to predict the clinical activity of regorafenib and assess prognosis in patients with metastatic colorectal cancer: A retrospective, exploratory analysis of the CORRECT trial. Lancet Oncol. 16:937–948. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Taniguchi H, Nakamura Y, Kotani D, Yukami H, Mishima S, Sawada K, Shirasu H, Ebi H, Yamanaka T, Aleshin A, et al: CIRCULATE-Japan: Circulating tumor DNA-guided adaptive platform trials to refine adjuvant therapy for colorectal cancer. Cancer Sci. 112:2915–2920. 2021. View Article : Google Scholar : PubMed/NCBI

80 

Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, Valtorta E, Schiavo R, Buscarino M, Siravegna G, et al: Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 486:532–536. 2012. View Article : Google Scholar : PubMed/NCBI

81 

Siravegna G, Mussolin B, Buscarino M, Corti G, Cassingena A, Crisafulli G, Ponzetti A, Cremolini C, Amatu A, Lauricella C, et al: Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat Med. 21:795–801. 2015. View Article : Google Scholar : PubMed/NCBI

82 

Wang DS, Liu ZX, Lu YX, Bao H, Wu X, Zeng ZL, Liu Z, Zhao Q, He CY, Lu JH, et al: Liquid biopsies to track trastuzumab resistance in metastatic HER2-positive gastric cancer. Gut. 68:1152–1161. 2019. View Article : Google Scholar : PubMed/NCBI

83 

Martinelli E, Ciardiello D, Martini G, Troiani T, Cardone C, Vitiello PP, Normanno N, Rachiglio AM, Maiello E, Latiano T, et al: Implementing anti-epidermal growth factor receptor (EGFR) therapy in metastatic colorectal cancer: Challenges and future perspectives. Ann Oncol. 31:30–40. 2020. View Article : Google Scholar : PubMed/NCBI

84 

Cremolini C, Rossini D, Dell'Aquila E, Lonardi S, Conca E, Del Re M, Busico A, Pietrantonio F, Danesi R, Aprile G, et al: Rechallenge for patients with RAS and BRAF wild-type metastatic colorectal cancer with acquired resistance to first-line cetuximab and irinotecan: A phase 2 single-arm clinical trial. JAMA Oncol. 5:343–350. 2019. View Article : Google Scholar : PubMed/NCBI

85 

Nakamura Y, Taniguchi H, Ikeda M, Bando H, Kato K, Morizane C, Esaki T, Komatsu Y, Kawamoto Y, Takahashi N, et al: Clinical utility of circulating tumor DNA sequencing in advanced gastrointestinal cancer: SCRUM-Japan GI-SCREEN and GOZILA studies. Nat Med. 26:1859–1864. 2020. View Article : Google Scholar : PubMed/NCBI

86 

Catana A, Apostu AP and Antemie RG: Multi gene panel testing for hereditary breast cancer-is it ready to be used? Med Pharm Rep. 92:220–225. 2019.PubMed/NCBI

87 

Stjepanovic N, Moreira L, Carneiro F, Balaguer F, Cervantes A, Balmaña J and Martinelli E; ESMO Guidelines Committee. Electronic address, : simpleclinicalguidelines@esmo.org: Hereditary gastrointestinal cancers: ESMO clinical practice guidelines for diagnosis, treatment and follow-up†. Ann Oncol. 30:1558–1571. 2019. View Article : Google Scholar : PubMed/NCBI

88 

Daly MB, Pal T, Berry MP, Buys SS, Dickson P, Domchek SM, Elkhanany A, Friedman S, Goggins M, Hutton ML, et al: Genetic/familial high-risk assessment: Breast, ovarian, and pancreatic, version 2.2021, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 19:77–102. 2021. View Article : Google Scholar : PubMed/NCBI

89 

Taylor A, Brady AF, Frayling IM, Hanson H, Tischkowitz M, Turnbull C and Side L; UK Cancer Genetics Group (UK-CGG), : Consensus for genes to be included on cancer panel tests offered by UK genetics services: Guidelines of the UK Cancer Genetics Group. J Med Genet. 55:372–377. 2018. View Article : Google Scholar : PubMed/NCBI

90 

Cerretelli G, Ager A, Arends MJ and Frayling IM: Molecular pathology of Lynch syndrome. J Pathol. 250:518–531. 2020. View Article : Google Scholar : PubMed/NCBI

91 

Win AK, Lindor NM, Young JP, Macrae FA, Young GP, Williamson E, Parry S, Goldblatt J, Lipton L, Winship I, et al: Risks of primary extracolonic cancers following colorectal cancer in lynch syndrome. J Natl Cancer Inst. 104:1363–1372. 2012. View Article : Google Scholar : PubMed/NCBI

92 

Syngal S, Brand RE, Church JM, Giardiello FM, Hampel HL and Burt RW; American College of Gastroenterology, : ACG clinical guideline: Genetic testing and management of hereditary gastrointestinal cancer syndromes. Am J Gastroenterol. 110:223–263. 2015. View Article : Google Scholar : PubMed/NCBI

93 

Le DT, Kim TW, Van Cutsem E, Geva R, Jäger D, Hara H, Burge M, O'Neil B, Kavan P, Yoshino T, et al: Phase II open-label study of pembrolizumab in treatment-refractory, microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: KEYNOTE-164. J Clin Oncol. 38:11–19. 2020. View Article : Google Scholar : PubMed/NCBI

94 

Overbeek KA, Cahen DL, Canto MI and Bruno MJ: Surveillance for neoplasia in the pancreas. Best Pract Res Clin Gastroenterol. 30:971–986. 2016. View Article : Google Scholar : PubMed/NCBI

95 

Shindo K, Yu J, Suenaga M, Fesharakizadeh S, Cho C, Macgregor-Das A, Siddiqui A, Witmer PD, Tamura K, Song TJ, et al: Deleterious germline mutations in patients with apparently sporadic pancreatic adenocarcinoma. J Clin Oncol. 35:3382–3390. 2017. View Article : Google Scholar : PubMed/NCBI

96 

Chaffee KG, Oberg AL, McWilliams RR, Majithia N, Allen BA, Kidd J, Singh N, Hartman AR, Wenstrup RJ and Petersen GM: Prevalence of germ-line mutations in cancer genes among pancreatic cancer patients with a positive family history. Genet Med. 20:119–127. 2018. View Article : Google Scholar : PubMed/NCBI

97 

Hu C, Hart SN, Polley EC, Gnanaolivu R, Shimelis H, Lee KY, Lilyquist J, Na J, Moore R, Antwi SO, et al: Association between inherited germline mutations in cancer predisposition genes and risk of pancreatic cancer. JAMA. 319:2401–2409. 2018. View Article : Google Scholar : PubMed/NCBI

98 

Bartsch DK, Gress TM and Langer P: Familial pancreatic cancer-current knowledge. Nat Rev Gastroenterol Hepatol. 9:445–453. 2012. View Article : Google Scholar : PubMed/NCBI

99 

Hampel H, Bennett RL, Buchanan A, Pearlman R and Wiesner GL: Guideline Development Group, American College of Medical Genetics and Genomics Professional Practice and Guidelines Committee and National Society of Genetic Counselors Practice Guidelines Committee: A practice guideline from the American college of medical genetics and genomics and the national society of genetic counselors: Referral indications for cancer predisposition assessment. Genet Med. 17:70–87. 2015. View Article : Google Scholar : PubMed/NCBI

100 

Pilonis ND, Tischkowitz M, Fitzgerald RC and di Pietro M: Hereditary diffuse gastric cancer: Approaches to screening, surveillance, and treatment. Annu Rev Med. 72:263–280. 2021. View Article : Google Scholar : PubMed/NCBI

101 

Donner I, Kiviluoto T, Ristimäki A, Aaltonen LA and Vahteristo P: Exome sequencing reveals three novel candidate predisposition genes for diffuse gastric cancer. Fam Cancer. 14:241–246. 2015. View Article : Google Scholar : PubMed/NCBI

102 

Berger MF and Mardis ER: The emerging clinical relevance of genomics in cancer medicine. Nat Rev Clin Oncol. 15:353–365. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Nakagawa H, Wardell CP, Furuta M, Taniguchi H and Fujimoto A: Cancer whole-genome sequencing: Present and future. Oncogene. 34:5943–5950. 2015. View Article : Google Scholar : PubMed/NCBI

104 

Colomer R, Mondejar R, Romero-Laorden N, Alfranca A, Sanchez-Madrid F and Quintela-Fandino M: When should we order a next generation sequencing test in a patient with cancer? EClinicalMedicine. 25:1004872020. View Article : Google Scholar : PubMed/NCBI

105 

ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium, . Pan-cancer analysis of whole genomes. Nature. 578:82–93. 2020. View Article : Google Scholar : PubMed/NCBI

106 

Frank MO, Koyama T, Rhrissorrakrai K, Robine N, Utro F, Emde AK, Chen BJ, Arora K, Shah M, Geiger H, et al: Sequencing and curation strategies for identifying candidate glioblastoma treatments. BMC Med Genomics. 12:562019. View Article : Google Scholar : PubMed/NCBI

107 

Takai E, Nakamura H, Chiku S, Kubo E, Ohmoto A, Totoki Y, Shibata T, Higuchi R, Yamamoto M, Furuse J, et al: Whole-exome Sequencing reveals new potential susceptibility genes for Japanese familial pancreatic cancer. Ann Surg. 275:e652–e658. 2022. View Article : Google Scholar : PubMed/NCBI

108 

Tan M, Brusgaard K, Gerdes AM, Mortensen MB, Detlefsen S, Schaffalitzky de Muckadell OB and Joergensen MT: Whole genome sequencing identifies rare germline variants enriched in cancer related genes in first degree relatives of familial pancreatic cancer patients. Clin Genet. 100:551–562. 2021. View Article : Google Scholar : PubMed/NCBI

109 

Bala P, Singh AK, Kavadipula P, Kotapalli V, Sabarinathan R and Bashyam MD: Exome sequencing identifies ARID2 as a novel tumor suppressor in early-onset sporadic rectal cancer. Oncogene. 40:863–874. 2021. View Article : Google Scholar : PubMed/NCBI

110 

Mendelaar PAJ, Smid M, van Riet J, Angus L, Labots M, Steeghs N, Hendriks MP, Cirkel GA, van Rooijen JM, Ten Tije AJ, et al: Whole genome sequencing of metastatic colorectal cancer reveals prior treatment effects and specific metastasis features. Nat Commun. 12:5742021. View Article : Google Scholar : PubMed/NCBI

111 

Ishaque N, Abba ML, Hauser C, Patil N, Paramasivam N, Huebschmann D, Leupold JH, Balasubramanian GP, Kleinheinz K, Toprak UH, et al: Whole genome sequencing puts forward hypotheses on metastasis evolution and therapy in colorectal cancer. Nat Commun. 9:47822018. View Article : Google Scholar : PubMed/NCBI

112 

Alves JM, Prado-López S, Tomás L, Valecha M, Estévez-Gómez N, Alvariño P, Geisel D, Modest DP, Sauer IM, Pratschke J, et al: Clonality and timing of relapsing colorectal cancer metastasis revealed through whole-genome single-cell sequencing. Cancer Lett. 543:2157672022. View Article : Google Scholar : PubMed/NCBI

113 

Erkizan HV, Sukhadia S, Natarajan TG, Marino G, Notario V, Lichy JH and Wadleigh RG: Exome sequencing identifies novel somatic variants in African American esophageal squamous cell carcinoma. Sci Rep. 11:148142021. View Article : Google Scholar : PubMed/NCBI

114 

Rose Brannon A, Jayakumaran G, Diosdado M, Patel J, Razumova A, Hu Y, Meng F, Haque M, Sadowska J, Murphy BJ, et al: Enhanced specificity of clinical high-sensitivity tumor mutation profiling in cell-free DNA via paired normal sequencing using MSK-ACCESS. Nat Commun. 12:37702021. View Article : Google Scholar : PubMed/NCBI

115 

Razavi P, Li BT, Brown DN, Jung B, Hubbell E, Shen R, Abida W, Juluru K, De Bruijn I, Hou C, et al: High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. Nat Med. 25:1928–1937. 2019. View Article : Google Scholar : PubMed/NCBI

116 

Modi A, Vai S, Caramelli D and Lari M: The illumina sequencing protocol and the novaseq 6000 system. Methods Mol Biol. 2242:15–42. 2021. View Article : Google Scholar : PubMed/NCBI

117 

Subramanian I, Verma S, Kumar S, Jere A and Anamika K: Multi-omics data integration, interpretation, and its application. Bioinform Biol Insights. 14:11779322198990512020. View Article : Google Scholar : PubMed/NCBI

118 

Basu A, Bodycombe NE, Cheah JH, Price EV, Liu K, Schaefer GI, Ebright RY, Stewart ML, Ito D, Wang S, et al: An interactive resource to identify cancer genetic and lineage dependencies targeted by small molecules. Cell. 154:1151–1161. 2013. View Article : Google Scholar : PubMed/NCBI

119 

Zhang B, Wang J, Wang X, Zhu J, Liu Q, Shi Z, Chambers MC, Zimmerman LJ, Shaddox KF, Kim S, et al: Proteogenomic characterization of human colon and rectal cancer. Nature. 513:382–387. 2014. View Article : Google Scholar : PubMed/NCBI

120 

Li Z, Gao X, Peng X, May Chen MJ, Li Z, Wei B, Wen X, Wei B, Dong Y, Bu Z, et al: Multi-omics characterization of molecular features of gastric cancer correlated with response to neoadjuvant chemotherapy. Sci Adv. 6:eaay42112020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2024
Volume 27 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Matsuoka T and Matsuoka T: Current status and perspectives of genetic testing in gastrointestinal cancer (Review). Oncol Lett 27: 21, 2024
APA
Matsuoka, T., & Matsuoka, T. (2024). Current status and perspectives of genetic testing in gastrointestinal cancer (Review). Oncology Letters, 27, 21. https://doi.org/10.3892/ol.2023.14155
MLA
Matsuoka, T., Yashiro, M."Current status and perspectives of genetic testing in gastrointestinal cancer (Review)". Oncology Letters 27.1 (2024): 21.
Chicago
Matsuoka, T., Yashiro, M."Current status and perspectives of genetic testing in gastrointestinal cancer (Review)". Oncology Letters 27, no. 1 (2024): 21. https://doi.org/10.3892/ol.2023.14155