Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 668
Filter
1.
Anticancer Res ; 44(8): 3481-3491, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39060051

ABSTRACT

BACKGROUND/AIM: Precision medicine aims to revolutionize healthcare by tailoring treatment regimens. This study aimed to integrate comprehensive tumor genomic profiling (CTGP) by targeted-gene panel sequencing and drug screening by circulating tumor cell-derived organoids (CTOs) into clinical practice for the treatment of gastrointestinal (GI) cancers. PATIENTS AND METHODS: Nine patients with various GI cancers underwent CTGP and CTO drug sensitivity testing. CTGP results guided targeted therapy and immunotherapy, while CTO drug sensitivity predicted response to chemotherapy and targeted agents. The drug recommendations from two platforms were correlated with the treatment response to the suggested medications retrospectively. RESULTS: Five patients received therapies aligned with CTGP, including HER2-targeted treatment, immunotherapy, and BRAF/MEK dual inhibition, showing positive responses. CTO drug sensitivity predicted progression under regorafenib (low potential benefit) and good response to chemotherapy with high potential benefit. The combination of CTGP and CTO drug sensitivity may exhibit significant correlation with clinical outcomes during treatment with candidate drugs, demonstrating a sensitivity of 79% and an accuracy of 75%. This encompasses various treatment modalities, such as chemotherapy, targeted therapy, and immunotherapy. CONCLUSION: The present investigation elucidated the integration of CTGP and CTO drug sensitivity screening into clinical practice in a complementary manner, showcasing a significant correlation between treatment response and testing outcomes. Additional prospective evaluation of these two testing modalities in a large cohort is warranted to confirm whether the inclusion of CTO drug sensitivity screening confers enhanced survival benefits compared to utilizing CTGP alone.


Subject(s)
Gastrointestinal Neoplasms , Neoplastic Cells, Circulating , Organoids , Humans , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/blood , Female , Male , Organoids/pathology , Organoids/drug effects , Middle Aged , Aged , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/drug effects , Precision Medicine/methods , Genomics/methods , Retrospective Studies , Gene Expression Profiling/methods , Biomarkers, Tumor/genetics , Molecular Targeted Therapy/methods , Adult , Drug Screening Assays, Antitumor/methods
2.
J Cancer Res Clin Oncol ; 150(7): 331, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38951269

ABSTRACT

OBJECTIVE: To conduct a systematic review and meta-analysis of case-control and cohort human studies evaluating metabolite markers identified using high-throughput metabolomics techniques on esophageal cancer (EC), cancer of the gastroesophageal junction (GEJ), and gastric cancer (GC) in blood and tissue. BACKGROUND: Upper gastrointestinal cancers (UGC), predominantly EC, GEJ, and GC, are malignant tumour types with high morbidity and mortality rates. Numerous studies have focused on metabolomic profiling of UGC in recent years. In this systematic review and meta-analysis, we have provided a collective summary of previous findings on metabolites and metabolomic profiling associated with EC, GEJ and GC. METHODS: Following the PRISMA procedure, a systematic search of four databases (Embase, PubMed, MEDLINE, and Web of Science) for molecular epidemiologic studies on the metabolomic profiles of EC, GEJ and GC was conducted and registered at PROSPERO (CRD42023486631). The Newcastle-Ottawa Scale (NOS) was used to benchmark the risk of bias for case-controlled and cohort studies. QUADOMICS, an adaptation of the QUADAS-2 (Quality Assessment of Diagnostic Accuracy) tool, was used to rate diagnostic accuracy studies. Original articles comparing metabolite patterns between patients with and without UGC were included. Two investigators independently completed title and abstract screening, data extraction, and quality evaluation. Meta-analysis was conducted whenever possible. We used a random effects model to investigate the association between metabolite levels and UGC. RESULTS: A total of 66 original studies involving 7267 patients that met the required criteria were included for review. 169 metabolites were differentially distributed in patients with UGC compared to healthy patients among 44 GC, 9 GEJ, and 25 EC studies including metabolites involved in glycolysis, anaerobic respiration, tricarboxylic acid cycle, and lipid metabolism. Phosphatidylcholines, eicosanoids, and adenosine triphosphate were among the most frequently reported lipids and metabolites of cellular respiration, while BCAA, lysine, and asparagine were among the most commonly reported amino acids. Previously identified lipid metabolites included saturated and unsaturated free fatty acids and ketones. However, the key findings across studies have been inconsistent, possibly due to limited sample sizes and the majority being hospital-based case-control analyses lacking an independent replication group. CONCLUSION: Thus far, metabolomic studies have provided new opportunities for screening, etiological factors, and biomarkers for UGC, supporting the potential of applying metabolomic profiling in early cancer diagnosis. According to the results of our meta-analysis especially BCAA and TMAO as well as certain phosphatidylcholines should be implicated into the diagnostic procedure of patients with UGC. We envision that metabolomics will significantly enhance our understanding of the carcinogenesis and progression process of UGC and may eventually facilitate precise oncological and patient-tailored management of UGC.


Subject(s)
Metabolomics , Humans , Metabolomics/methods , Esophageal Neoplasms/blood , Esophageal Neoplasms/metabolism , Stomach Neoplasms/blood , Stomach Neoplasms/metabolism , Stomach Neoplasms/diagnosis , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/diagnosis , Metabolome/physiology , Case-Control Studies , Esophagogastric Junction/pathology , Esophagogastric Junction/metabolism
3.
Curr Opin Oncol ; 36(4): 282-290, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38726808

ABSTRACT

PURPOSE OF REVIEW: This review explores the role of circulating tumor (ct)DNA as a biomarker for clinical decision-making and monitoring purposes in metastatic gastrointestinal stromal tumor (GIST) patients. We discuss key insights from recent clinical trials and anticipate the future perspectives of ctDNA profiling within the clinical landscape of GIST. RECENT FINDINGS: The identification and molecular characterization of KIT/platelet-derived growth factor receptor alpha (PDGFRA) mutations from ctDNA in metastatic GIST is feasible and reliable. Such identification through ctDNA serves as a predictor of clinical outcomes to tyrosine-kinase inhibitors (TKIs) in metastatic patients. Additionally, conjoined ctDNA analysis from clinical trials reveal the evolving mutational landscapes and increase in intratumoral heterogeneity across treatment lines. Together, this data positions ctDNA determination as a valuable tool for monitoring disease progression and guiding therapy in metastatic patients. These collective efforts culminated in the initiation of a ctDNA-based randomized clinical trial in GIST, marking a significant milestone in integrating ctDNA testing into the clinical care of GIST patients. SUMMARY: The dynamic field of ctDNA technologies is rapidly evolving and holds significant promise for research. Several trials have successfully validated the clinical utility of ctDNA in metastatic GIST, laying the foundations for its prospective integration into the routine clinical management of GIST patients.


Subject(s)
Circulating Tumor DNA , Gastrointestinal Neoplasms , Gastrointestinal Stromal Tumors , Humans , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/blood , Circulating Tumor DNA/blood , Circulating Tumor DNA/genetics , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/drug therapy , Biomarkers, Tumor/genetics , Biomarkers, Tumor/blood , Protein Kinase Inhibitors/therapeutic use , Mutation , Randomized Controlled Trials as Topic
4.
PLoS One ; 19(5): e0303290, 2024.
Article in English | MEDLINE | ID: mdl-38743680

ABSTRACT

Genike, the imatinib (IM)-alpha form is widely used in the treatment of gastrointestinal stromal tumor (GIST) patients in China. We wanted to investigate whether there are differences in IM plasma concentrations, adverse events, health-related quality of life (QOL) and outcomes between patients treated with Genike and Glivec. Thirty included GIST patients receiving IM treatment were matched to either Genike or Glivec according to gastrectomy, body weight, body surface area and sex. There was no statistically significant difference in IM trough plasma levels between the two groups. There were no significant differences in very common adverse events of IM between the Genike and Glivec groups. IM was well tolerated, although it was associated with a significant change in cognitive function (P < 0.001), fatigue (P = 0.015), pain (P = 0.015), nausea/vomiting (P = 0.029), insomnia (P = 0.019), diarrhea (P = 0.003) and financial difficulties (P < 0.001). Physical functioning, financial burden and insomnia were significantly different between the two groups (P = 0.026). Until Aug. 2022, there was no significant difference in time to imatinib treatment failure (TTF) between the two groups. In conclusion, there was no difference in IM plasma concentration and adverse events between Genike and Glivec. Both Genike and Glivec could partially decrease the QOL of GIST patients. Physical functioning was worse in Genike group than in Glivec group, while the economic burden and symptoms of insomnia in Glivec patients were worse. There was no significant difference in TTF between the two groups.


Subject(s)
Antineoplastic Agents , Gastrointestinal Stromal Tumors , Imatinib Mesylate , Quality of Life , Humans , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/blood , Imatinib Mesylate/therapeutic use , Imatinib Mesylate/adverse effects , Female , Male , Middle Aged , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Adult , Treatment Outcome , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/blood , Case-Control Studies
5.
PLoS One ; 19(5): e0304574, 2024.
Article in English | MEDLINE | ID: mdl-38814898

ABSTRACT

BACKGROUND: The prevalence of gastrointestinal tumors continues to be significant. To uncover promising therapeutic targets for these tumors, we rigorously executed a Mendelian randomization (MR) study to comprehensively screen the blood metabolomes for potential causal mediators of five frequently encountered gastrointestinal tumors (Liver Cancer, Colorectal Cancer, Esophageal Cancer, Gastric Cancer and Pancreatic Cancer). METHODS: We selected a comprehensive set of 137 distinct blood metabolites derived from three large-scale genome-wide association studies (GWASs) involving a total of 147827 participants of European ancestry. The gastrointestinal tumors-related data were obtained from a GWAS conducted within the Finnish study. Through meticulous MR analyses, we thoroughly assessed the associations between blood metabolites and gastrointestinal tumors. Additionally, a phenome-wide MR (Phe-MR) analysis was employed to investigate the potential on-target side effects of metabolite interventions. RESULTS: We have identified 1 blood metabolites, namely isovalerylcarnitine (ORlog10: 1.01; 95%CI, 1.01-1.02; P = 1.81×10-7), as the potential causal mediators for liver cancer. However, no potential pathogenic mediators were detected for the other four tumors. CONCLUSIONS: The current systematic MR analysis elucidated the potential role of isovalerylcarnitine as a causal mediator in the development of liver cancer. Leveraging the power of Phe-MR study facilitated the identification of potential adverse effects associated with drug targets for liver cancer prevention. Considering the weighing of pros and cons, isovalerylcarnitine emerges as a promising candidate for targeted drug interventions in the realm of liver cancer prevention.


Subject(s)
Gastrointestinal Neoplasms , Genome-Wide Association Study , Mendelian Randomization Analysis , Metabolome , Humans , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/genetics , Male , Female , Finland/epidemiology , Carnitine/blood , Carnitine/analogs & derivatives , Middle Aged , Risk Factors , Liver Neoplasms/blood , Liver Neoplasms/genetics
6.
Biomarkers ; 29(5): 233-243, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38696280

ABSTRACT

BACKGROUND: Despite numerous reports on the alterations of microRNA-1246 (miR-1246) expression level in digestive system cancers, its role in gastrointestinal cancers (GICs) remains unclear. This meta-analysis aimed to assess the diagnostic potential of circulating miR-1246 in GICs. METHODS: Meta-disc version 1.4 and Comprehensive Meta-Analysis (CMA) version 3.7 software were used to calculate pooled sensitivity, specificity, likelihood ratios, diagnostic odds ratio (DOR), area under the curve (AUC), Q*index and summary receiver-operating characteristic (SROC). Subgroup analyses were conducted for cancer type, sample type and geographical region. Publication bias was assessed using Begg's and Egger's tests. RESULTS: A total of 14 articles involving 18 studies and 1526 participants (972 cases and 554 controls) were included. The diagnostic accuracy of miRNA-1246 in GICs was as follows: pooled sensitivity: 0.81 (95% CI: 0.79 - 0.83), specificity: 0.74 (95% CI: 0.71 - 0.77), PLR: 3.315 (95% CI: 2.33 - 4.72), NLR: 0.221 (95% CI: 0.153 - 0.319), DOR: 16.87 (95% CI: 9.45 - 30.09), AUC: 0.891, and Q*-index: 0.807. No publication bias was found based on Begg's (p = 0.172) and Egger's (p = 0.113) tests. CONCLUSION: Circulating miR-1246 shows promise as a non-invasive biomarker for early detection of GICs.


Subject(s)
Biomarkers, Tumor , Gastrointestinal Neoplasms , MicroRNAs , Humans , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/genetics , MicroRNAs/blood , MicroRNAs/genetics , ROC Curve , Sensitivity and Specificity , Circulating MicroRNA/blood , Circulating MicroRNA/genetics
7.
Biomarkers ; 29(4): 194-204, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38644767

ABSTRACT

INTRODUCTION: Methylated circulating tumour DNA (ctDNA) blood tests for BCAT1/IKZF1 (COLVERA) and SEPT9 (Epi proColon) are used to detect colorectal cancer (CRC). However, there are no ctDNA assays approved for other gastrointestinal adenocarcinomas. We aimed to characterize BCAT1, IKZF1 and SEPT9 methylation in different gastrointestinal adenocarcinoma and non-gastrointestinal tumours to determine if these validated CRC biomarkers might be useful for pan-gastrointestinal adenocarcinoma detection. METHODS: Tissue DNA methylation data from colorectal (COAD, READ), gastroesophageal (ESCA, STAD), pancreatic (PAAD) and cholangiocarcinoma (CHOL) adenocarcinoma cohorts within The Cancer Genome Atlas were used for differential methylation analyses. Clinicodemographic predictors of BCAT1, IKZF1 and SEPT9 methylation, and the selectivity of hypermethylated BCAT1, IKZF1 and SEPT9 for colorectal adenocarcinomas in comparison to other cancers were each explored with beta regression. RESULTS: Hypermethylated BCAT1, IKZF1 and SEPT9 were each differentially methylated in colorectal and gastroesophageal adenocarcinomas. IKZF1 was differentially methylated in pancreatic adenocarcinoma. Hypermethylated DNA biomarkers BCAT1, IKZF1 and SEPT9 were largely stable across different stages of disease and were highly selective for gastrointestinal adenocarcinomas relative to other cancer types. DISCUSSION: Existing CRC methylated ctDNA blood tests for BCAT1/IKZF1 and SEPT9 might be usefully repurposed for use in other gastrointestinal adenocarcinomas and warrant further prospective ctDNA studies.


Subject(s)
Adenocarcinoma , Biomarkers, Tumor , DNA Methylation , Gastrointestinal Neoplasms , Ikaros Transcription Factor , Septins , Humans , Septins/genetics , Septins/blood , Ikaros Transcription Factor/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/blood , Adenocarcinoma/genetics , Adenocarcinoma/diagnosis , Adenocarcinoma/blood , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/blood , Male , Circulating Tumor DNA/genetics , Circulating Tumor DNA/blood , Female , Colorectal Neoplasms/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/blood , Cholangiocarcinoma/genetics , Cholangiocarcinoma/diagnosis , Cholangiocarcinoma/blood , Middle Aged , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/blood
8.
J Proteome Res ; 23(5): 1679-1688, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38546438

ABSTRACT

Previous metabolomics studies have highlighted the predictive value of metabolites on upper gastrointestinal (UGI) cancer, while most of them ignored the potential effects of lifestyle and genetic risk on plasma metabolites. This study aimed to evaluate the role of lifestyle and genetic risk in the metabolic mechanism of UGI cancer. Differential metabolites of UGI cancer were identified using partial least-squares discriminant analysis and the Wilcoxon test. Then, we calculated the healthy lifestyle index (HLI) score and polygenic risk score (PRS) and divided them into three groups, respectively. A total of 15 metabolites were identified as UGI-cancer-related differential metabolites. The metabolite model (AUC = 0.699) exhibited superior discrimination ability compared to those of the HLI model (AUC = 0.615) and the PRS model (AUC = 0.593). Moreover, subgroup analysis revealed that the metabolite model showed higher discrimination ability for individuals with unhealthy lifestyles compared to that with healthy individuals (AUC = 0.783 vs 0.684). Furthermore, in the genetic risk subgroup analysis, individuals with a genetic predisposition to UGI cancer exhibited the best discriminative performance in the metabolite model (AUC = 0.770). These findings demonstrated the clinical significance of metabolic biomarkers in UGI cancer discrimination, especially in individuals with unhealthy lifestyles and a high genetic risk.


Subject(s)
Gastrointestinal Neoplasms , Healthy Lifestyle , Aged , Female , Humans , Male , Middle Aged , Biological Specimen Banks , Biomarkers, Tumor/genetics , Biomarkers, Tumor/blood , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/blood , Genetic Risk Score , Metabolomics/methods , UK Biobank , United Kingdom/epidemiology
10.
BMC Cancer ; 21(1): 989, 2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34479528

ABSTRACT

BACKGROUND: Gastrointestinal tumors are a leading cause of mortality worldwide. As shown in our previous study, miR-1290 is overexpressed in colorectal cancer (CRC) and promotes tumor progression. We therefore aimed to explore the potential of circulating miR-1290 as a biomarker for gastrointestinal cancer. METHODS: A serum miRNA sequencing analysis was performed. Then, circulating miRNA detection technologies were established. The expression of miR-1290 was analyzed in gastrointestinal tumor cell lines and culture supernatants. Expression levels of circulating miR-1290 in clinical samples were examined. Associations between miR-1290 expression and clinicopathologic characteristics were analyzed. Xenograft models were generated to assess the fluctuation in serum miR-1290 levels during disease progression. RESULTS: Through miRNA sequencing, we identified that miR-1290 was overexpressed in serum samples from patients with CRC. We confirmed that human gastrointestinal tumor cells express and secrete miR-1290. The circulating miR-1290 levels was up-regulated in patients with pancreatic cancer (PC) (p < 0.01), CRC (p < 0.05), and gastric cancer (GC) (p < 0.01). High miR-1290 expression levels were associated with tumor size, lymphatic invasion, vascular invasion, distant metastasis, tumor differentiation and AJCC stage in patients with PC and CRC. The area under the curve (AUC) was 0.8857 in patients with PC, with 60.9% sensitivity and 90.0% specificity. The AUC was 0.7852 in patients with CRC, with 42.0% sensitivity and 90.0% specificity. In patients with GC, the AUC was 0.6576, with 26.0% sensitivity and 90.0% specificity. The in vivo model verified that the circulating miR-1290 level was significantly increased after tumor formation and decreased after drug treatment. CONCLUSIONS: Our findings indicate that circulating miR-1290 is a potential biomarker for gastrointestinal cancer diagnosis and monitoring.


Subject(s)
Biomarkers, Tumor/genetics , Gastrointestinal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Animals , Apoptosis , Biomarkers, Tumor/blood , Case-Control Studies , Cell Proliferation , Female , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/blood , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Clin. transl. oncol. (Print) ; 23(8): 1646-1656, ago. 2021.
Article in English | IBECS | ID: ibc-222163

ABSTRACT

Background Identifying valid biomarkers for patient selection impressively promotes the success of anti-PD-1 therapy. However, the unmet need for biomarkers in gastrointestinal (GI) cancers remains significant. We aimed to explore the predictive value of the circulating T-cell receptor (TCR) repertoire for clinical outcomes in GI cancers who received anti-PD-1 therapy. Methods 137 pre- and 79 post-treated peripheral blood samples were included. The TCR repertoire was evaluated by sequencing of complementarity-determining region 3 (CDR3) in the TRB gene. The Shannon index was used to measure the diversity of the TCR repertoire, and Morisita’s overlap index was used to determine TCR repertoire similarities between pre- and post-treated samples. Results Among all enrolled patients, 76 received anti-PD-1 monotherapy and 61 received anti-PD-1 combination therapy. In the anti-PD-1 monotherapy cohort, patients with higher baseline TCR diversity exhibited a significantly higher disease control rate (77.8% vs. 47.2%; hazard ratio [HR] 3.92; 95% confidence interval [CI] 1.14–13.48; P = 0.030) and a longer progression-free survival (PFS) (median: 6.47 months vs. 2.77 months; HR 2.10; 95% CI 1.16–3.79; P = 0.014) and overall survival (OS) (median: NA vs. 8.97 months; HR 3.53; 95% CI 1.49–8.38; P = 0.004) than those with lower diversity. Moreover, patients with a higher TCR repertoire similarity still showed a superior PFS (4.43 months vs. 1.84 months; HR 13.98; 95% CI 4.37–44.68; P < 0.001) and OS (13.40 months vs. 6.12 months; HR 2.93; 95% CI 1.22–7.03; P = 0.016) even in the cohort with lower baseline diversity. However, neither biomarker showed predictive value in the anti-PD-1 combination therapy cohort. Interestingly, the combination of TCR diversity and PD-L1 expression can facilitate patient stratification in a pooled cohort. Conclusion The circulating TCR repertoire can serve as a predictor of clinical outcomes in anti-PD-1 therapy in GI cancers (AU)


Subject(s)
Humans , Male , Female , Young Adult , Adult , Middle Aged , Aged , Antineoplastic Agents, Immunological/therapeutic use , Gastrointestinal Neoplasms/therapy , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/blood , B7-H1 Antigen/metabolism , Biomarkers, Tumor/blood , Complementarity Determining Regions/genetics , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/mortality , Progression-Free Survival , Treatment Outcome
12.
Cancer Med ; 10(20): 7021-7039, 2021 10.
Article in English | MEDLINE | ID: mdl-34423578

ABSTRACT

BACKGROUND: The prognostic significance of programmed cell death-ligand 1 (PD-L1) expression on circulating tumor cells (CTCs) has been explored but is still in controversy. We performed, for the first time, a meta-analysis to systematically evaluate its prognostic value in human cancers. METHODS: Literature databases were searched for eligible studies prior to June 30, 2021. The pooled hazard ratios (HRs) and 95% confidence intervals (95% CIs) were calculated for the associations of pre-treatment and post-treatment PD-L1+ CTCs with progression-free survival (PFS) and overall survival (OS). Subgroup analyses with regards to cancer type, treatment, CTC enrichment method, PD-L1 detection method, cut-off, and specifically the comparison model were performed. RESULTS: We included 30 eligible studies (32 cohorts, 1419 cancer patients) in our analysis. Pre-treatment PD-L1+ CTCs detected by immunofluorescence (IF) tended to predict better PFS (HR = 0.55, 95% CI 0.28-1.08, p = 0.084) and OS (HR = 0.61, 95% CI 0.36-1.04, p = 0.067) for immune checkpoint inhibitor (ICI) treatment, but were significantly associated with unfavorable survival for non-ICI therapies (PFS: HR = 1.85, 95% CI 1.21-2.85, p = 0.005; OS: HR = 2.44, 95% CI 1.69-3.51, p < 0.001). Post-treatment PD-L1+ CTCs predicted markedly worse PFS and OS. The prognostic value was obviously modulated by comparison models. Among patients with detectable CTCs, PD-L1+ individuals had comparable survival to PD-L1- individuals, except ICI treatment for which PD-L1+ may predict better PFS (HR = 0.42, 95% CI 0.17-1.06, p = 0.067). Patients with PD-L1+ CTCs had worse survival prognosis compared to those without PD-L1+ CTCs in overall analysis (PFS: HR = 2.10, 95% CI 1.59-2.77, p < 0.001; OS: HR = 2.55, 95% CI 1.70-3.81, p < 0.001) and in most subgroups. CONCLUSIONS: Our analysis demonstrated that PD-L1 positive expression on CTCs predicted better survival prognosis for ICI treatment but worse survival for other therapies, which thus can be potentially used as a prognostic marker of malignant tumor treatment. However, the prognostic value of PD-L1+ CTCs for ICI treatment needs validation by more large-scale studies in the future.


Subject(s)
B7-H1 Antigen/metabolism , Neoplasms/blood , Neoplasms/mortality , Neoplastic Cells, Circulating/metabolism , Breast Neoplasms/blood , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Confidence Intervals , Female , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/mortality , Gastrointestinal Neoplasms/pathology , Humans , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/blood , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Neoplasms/pathology , Prognosis , Progression-Free Survival , Proportional Hazards Models , Publication Bias , Urogenital Neoplasms/blood , Urogenital Neoplasms/mortality , Urogenital Neoplasms/pathology
13.
Epigenomics ; 13(16): 1327-1339, 2021 08.
Article in English | MEDLINE | ID: mdl-34369810

ABSTRACT

DNA methylation is of paramount importance for the evolution of human cancers. Its high sensitivity and specificity make it a potential biomarker for early cancer screening in the context of an increasing global burden of gastrointestinal (GI) carcinoma. More DNA methylation biomarkers are emerging with the development of liquid biopsy and sensitive DNA methylation detection technology. This review provides an overview of DNA methylation, focusing on the presentation and comparison of 5-methylcytosine detection technologies, and introduces the promising plasma-based cell-free DNA (cfDNA) methylation biomarkers published in recent years for early screening of GI carcinoma. Finally, we summarize and discuss the future of plasma cfDNA methylation markers detection as a clinical tool for early screening of GI carcinoma.


Lay abstract In the context of an increasing global burden of gastrointestinal (GI) carcinoma, early cancer screening is of paramount importance, so there is an urgent need for efficient and reliable potential biomarkers. More biomarkers are emerging with the development of noninvasive test and related detection technologies. This review provides an overview of certain potential biomarkers named DNA methylation, focusing on the presentation and comparison of related detection technologies for this type of biomarkers and introduces the promising plasma-based biomarkers published in recent years for early screening of GI carcinoma. Finally, we summarize and discuss the future of plasma-based biomarkers detection as a clinical tool for early screening of GI carcinoma.


Subject(s)
Circulating Tumor DNA , DNA Methylation , Epigenesis, Genetic , Epigenomics/methods , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/genetics , Liquid Biopsy/methods , Biomarkers, Tumor , Early Detection of Cancer/methods , Gastrointestinal Neoplasms/blood , Humans , Prognosis
14.
Biomed Pharmacother ; 141: 111929, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34328118

ABSTRACT

BACKGROUND: Clinical evidence of thiamine-related neuropsychiatric symptoms, including the initial stage, is limited because serum thiamine levels tend to be evaluated only for patients who develop severe neuropsychiatric symptoms suspected to be related to severe thiamine deficiency. This study aimed to evaluate the relationship between thiamine decline and neuropsychiatric symptoms, including initial symptoms, and the effect of chemotherapy on serum thiamine levels in gastrointestinal and hematological cancer patients receiving chemotherapy. METHOD: We retrospectively identified 87 patients who were diagnosed with gastrointestinal and hematological cancers at our hospital. We evaluated the risk factors associated with neuropsychiatric symptoms, including initial symptoms (neuropsychiatric symptoms), the relationship between the presence of neuropsychiatric symptoms and serum thiamine levels, and changes in serum thiamine levels after chemotherapy. RESULTS: Logistic regression analysis identified thiamine decline as a significant factor associated with neuropsychiatric symptoms (p < 0.001, odds ratio = 0.040, 95% confidence interval [CI]: 0.010-0.163). The Mann-Whitney U test showed that patients with neuropsychiatric symptoms had significantly lower serum thiamine levels (19.5 ± 5.4 ng/mL, n = 39) than patients without neuropsychiatric symptoms (31.9 ± 14.2 ng/mL, n = 48) (p = 0.001). In hematological cancer patients, serum thiamine levels gradually declined after chemotherapy, with the lowest levels at 5-8 weeks (23.5 ± 7.6 ng/mL, P = 0.035 vs. 0 weeks, Wilcoxon rank sum test). CONCLUSION: Our study showed that a decrease in serum thiamine levels can be a risk factor for neuropsychiatric symptoms, and chemotherapy can lead to a decrease in serum thiamine levels.


Subject(s)
Antineoplastic Agents/adverse effects , Gastrointestinal Neoplasms/blood , Hematologic Neoplasms/blood , Mental Disorders/blood , Thiamine Deficiency/blood , Thiamine/blood , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Female , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/epidemiology , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/epidemiology , Humans , Male , Mental Disorders/epidemiology , Middle Aged , Retrospective Studies , Thiamine Deficiency/epidemiology , Young Adult
15.
Radiat Oncol ; 16(1): 83, 2021 May 03.
Article in English | MEDLINE | ID: mdl-33941218

ABSTRACT

BACKGROUND: This communication reports the identification of a new panel of transcriptional changes in inflammation-associated genes observed in response to ionising radiation received by radiotherapy patients. METHODS: Peripheral blood samples were taken with ethical approval and informed consent from a total of 20 patients undergoing external beam radiotherapy for breast, lung, gastrointestinal or genitourinary tumours. Nanostring nCounter analysis of transcriptional changes was carried out in samples prior and 24 h post-delivery of the 1st radiotherapy fraction, just prior to the 5th or 6th fraction, and just before the last fraction. RESULTS: Statistical analysis with BRB-ArrayTools, GLM MANOVA and nSolver, revealed a radiation responsive panel of genes which varied by patient group (type of cancer) and with time since exposure (as an analogue for dose received), which may be useful as a biomarker of radiation response. CONCLUSION: Further validation in a wider group of patients is ongoing, together with work towards a full understanding of patient specific responses in support of personalised approaches to radiation medicine.


Subject(s)
Biomarkers, Tumor/blood , Gene Expression Regulation, Neoplastic/radiation effects , Inflammation/genetics , Neoplasms/blood , Radiation, Ionizing , Transcriptome/radiation effects , Biomarkers, Tumor/genetics , Breast Neoplasms/blood , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/radiotherapy , Female , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/immunology , Gastrointestinal Neoplasms/radiotherapy , Humans , Lung Neoplasms/blood , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/radiotherapy , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/radiotherapy , Pilot Projects , Prognosis , Urogenital Neoplasms/blood , Urogenital Neoplasms/genetics , Urogenital Neoplasms/immunology , Urogenital Neoplasms/radiotherapy
16.
Int J Mol Sci ; 22(5)2021 Feb 26.
Article in English | MEDLINE | ID: mdl-33652558

ABSTRACT

Cancer immunotherapy, specifically immune checkpoint blockade, has been found to be effective in the treatment of metastatic cancers. However, only a subset of patients achieve clinical responses. Elucidating pretreatment biomarkers predictive of sustained clinical response is a major research priority. Another research priority is evaluating changes in the immune system before and after treatment in responders vs. nonresponders. Our group has been studying immune networks as an accurate reflection of the global immune state. Flow cytometry (FACS, fluorescence-activated cell sorting) data characterizing immune cell panels in peripheral blood mononuclear cells (PBMC) from gastroesophageal adenocarcinoma (GEA) patients were used to analyze changes in immune networks in this setting. Here, we describe a novel computational pipeline to perform secondary analyses of FACS data using systems biology/machine learning techniques and concepts. The pipeline is centered around comparative Bayesian network analyses of immune networks and is capable of detecting strong signals that conventional methods (such as FlowJo manual gating) might miss. Future studies are planned to validate and follow up the immune biomarkers (and combinations/interactions thereof) associated with clinical responses identified with this computational pipeline.


Subject(s)
Adenocarcinoma , Flow Cytometry , Gastrointestinal Neoplasms , Immunotherapy , Leukocytes, Mononuclear , Adenocarcinoma/blood , Adenocarcinoma/immunology , Adenocarcinoma/therapy , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/immunology , Gastrointestinal Neoplasms/therapy , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology
17.
PLoS One ; 16(2): e0245052, 2021.
Article in English | MEDLINE | ID: mdl-33544706

ABSTRACT

Emerging evidence that an elevated serum gamma-glutamyltransferase (GGT) level is associated with an increased risk of gastrointestinal cancer, but still controversial. The aim of this study to assess the relationship between GGT level and risk of gastrointestinal cancer, and the contribution of the interaction of hyperglycemia with elevated GGT level to the incidence of gastrointestinal cancer by the stratified analysis. A total of 8,120,665 Koreans who received medical checkups in 2009 were included. Subjects were classified according to the quartile of GGT level for women and men. The incidence rates of gastrointestinal cancer for each group were analyzed using Cox proportional hazards models. During follow-up, 129,853 cases of gastrointestinal cancer newly occurred (esophagus, 3,792; stomach, 57,932; and colorectal, 68,789 cases). The highest GGT quartile group showed an increased risk of gastrointestinal cancer (esophagus, hazard ratio = 2.408 [95% confidence interval, 2.184-2.654]; stomach, 1.121 [1.093-1.149]; and colorectal, 1.185 [1.158-1.211]). The risk increased significantly with the rise in GGT quartile level, regardless of the site of cancer. The stratified analysis according to glycemic status showed that the effect of elevated GGT was predominant in the risk of esophageal cancer. The effect of elevated GGT further increased the risk of stomach and colorectal cancers in diabetic patients. An elevated level of GGT was associated with an increased risk of gastrointestinal cancer, regardless of the site of cancer. The effect of the increase in GGT level on the risk of gastrointestinal cancer depended on the type of cancer and glycemic status.


Subject(s)
Gastrointestinal Neoplasms/epidemiology , gamma-Glutamyltransferase/blood , Adult , Blood Glucose , Cohort Studies , Female , Gastrointestinal Neoplasms/blood , Humans , Incidence , Male , Middle Aged , Risk
18.
Clin Transl Oncol ; 23(8): 1646-1656, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33583004

ABSTRACT

BACKGROUND: Identifying valid biomarkers for patient selection impressively promotes the success of anti-PD-1 therapy. However, the unmet need for biomarkers in gastrointestinal (GI) cancers remains significant. We aimed to explore the predictive value of the circulating T-cell receptor (TCR) repertoire for clinical outcomes in GI cancers who received anti-PD-1 therapy. METHODS: 137 pre- and 79 post-treated peripheral blood samples were included. The TCR repertoire was evaluated by sequencing of complementarity-determining region 3 (CDR3) in the TRB gene. The Shannon index was used to measure the diversity of the TCR repertoire, and Morisita's overlap index was used to determine TCR repertoire similarities between pre- and post-treated samples. RESULTS: Among all enrolled patients, 76 received anti-PD-1 monotherapy and 61 received anti-PD-1 combination therapy. In the anti-PD-1 monotherapy cohort, patients with higher baseline TCR diversity exhibited a significantly higher disease control rate (77.8% vs. 47.2%; hazard ratio [HR] 3.92; 95% confidence interval [CI] 1.14-13.48; P = 0.030) and a longer progression-free survival (PFS) (median: 6.47 months vs. 2.77 months; HR 2.10; 95% CI 1.16-3.79; P = 0.014) and overall survival (OS) (median: NA vs. 8.97 months; HR 3.53; 95% CI 1.49-8.38; P = 0.004) than those with lower diversity. Moreover, patients with a higher TCR repertoire similarity still showed a superior PFS (4.43 months vs. 1.84 months; HR 13.98; 95% CI 4.37-44.68; P < 0.001) and OS (13.40 months vs. 6.12 months; HR 2.93; 95% CI 1.22-7.03; P = 0.016) even in the cohort with lower baseline diversity. However, neither biomarker showed predictive value in the anti-PD-1 combination therapy cohort. Interestingly, the combination of TCR diversity and PD-L1 expression can facilitate patient stratification in a pooled cohort. CONCLUSION: The circulating TCR repertoire can serve as a predictor of clinical outcomes in anti-PD-1 therapy in GI cancers.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Gastrointestinal Neoplasms/therapy , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/blood , Adult , Aged , B7-H1 Antigen/metabolism , Biomarkers, Tumor/blood , Complementarity Determining Regions/genetics , Female , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/mortality , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Progression-Free Survival , Receptors, Antigen, T-Cell, alpha-beta/blood , Time Factors , Treatment Outcome , Young Adult
19.
Pancreas ; 50(2): 130-137, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33560090

ABSTRACT

OBJECTIVE: The aim of the study was to assess the impact of systemic markers of inflammation on the outcomes in patients with neuroendocrine tumors (NETs) treated with everolimus or placebo (as measured by baseline neutrophil-to-lymphocyte ratio [NLR] and lymphocyte-to-monocyte ratio [LMR]). METHODS: Patient data (gastrointestinal, pancreatic, and lung NETs) from 2 large phase 3 studies, RADIANT-3 (n = 410) and RADIANT-4 (n = 302), were pooled and analyzed. The primary end point was centrally assessed progression-free survival (PFS) as estimated by the Kaplan-Meier method. RESULTS: In the pooled population, elevated LMR (median PFS, 11.1 months; 95% confidence interval, 9.3-13.7; hazard ratio, 0.69; P < 0.001) and reduced NLR (median PFS, 10.8 months; 95% confidence interval, 9.2-11.7; hazard ratio, 0.75; P = 0.0060) correlated with longer PFS among all patients. These markers were also found to be prognostic in the everolimus- and placebo-treated subgroups. CONCLUSIONS: Data from this study suggest that LMR and NLR are robust prognostic markers for NETs and could potentially be used to identify patients who may receive or are receiving the most benefit from targeted therapies. As both are derived from a complete blood count, they can be routinely used in clinical practice, providing valuable information to clinicians and patients alike.


Subject(s)
Gastrointestinal Neoplasms/blood , Inflammation/blood , Lung Neoplasms/blood , Lymphocytes , Monocytes , Neuroendocrine Tumors/blood , Neutrophils , Antineoplastic Agents/therapeutic use , Clinical Decision-Making , Clinical Trials, Phase III as Topic , Everolimus/therapeutic use , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/mortality , Humans , Inflammation/diagnosis , Inflammation/drug therapy , Inflammation/mortality , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lymphocyte Count , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/mortality , Predictive Value of Tests , Progression-Free Survival , Randomized Controlled Trials as Topic , Time Factors
20.
Future Oncol ; 17(12): 1507-1518, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33626926

ABSTRACT

Aim: To evaluate the role of clinical features and blood markers in patients with malignant digestive tract tumors bone metastasis. Materials & methods: A total of 267 patients were included in this trial. Age, gender, primary tumor site, metastatic sites, T/N stage, high-density lipoprotein, low-density lipoprotein, total cholesterol, triglycerides, alkaline phosphatase, LDH, Ca levels, platelet, neutrophils to absolute value of lymphocytes (NLR), ratio of platelets to absolute values of lymphocytes (PLR) were analyzed. Results: T stage, lymph node metastasis, N stage and liver and lung metastasis were independent risk factors. LDH + alkaline phosphatase + NLR + PLR and LDH + NLR, respectively have higher predictive value for bone metastasis compared with patients with early-stage malignant digestive tract tumor and patients with advanced malignant digestive tract tumor without bone metastasis. Conclusion: Some clinical features or blood markers have the potential to detect bone metastasis early to avoid skeletal complications.


Subject(s)
Biomarkers, Tumor/blood , Bone Neoplasms/diagnosis , Early Detection of Cancer/methods , Gastrointestinal Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Alkaline Phosphatase/blood , Bone Neoplasms/blood , Bone Neoplasms/secondary , Female , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/diagnosis , Humans , Lactate Dehydrogenases/blood , Male , Middle Aged , Neoplasm Staging , Platelet Count , Predictive Value of Tests , Prognosis , Retrospective Studies , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL