Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21.433
Filter
1.
World J Gastroenterol ; 30(28): 3367-3372, 2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39091718

ABSTRACT

In this editorial, the roles of tata-box-binding protein-associated factor 15 (TAF15) in oncogenesis, tumor behavior, and as a therapeutic target in cancers in the context of gastrointestinal (GI) tumors are discussed concerning the publication by Guo et al. TAF15 is a member of the FET protein family with a comprehensive range of cellular processes. Besides, evidence has shown that TAF15 is involved in many diseases, including cancers. TAF15 contributes to carcinogenesis and tumor behavior in many tumors. Besides, its relationship with the mitogen-activated protein kinases (MAPK) signaling pathway makes TAF15 a new target for therapy. Although, the fact that there is few studies investigating the expression of TAF15 constitutes a potential limitation in GI system, the association of TAF15 expression with aggressive tumor behavior and, similar to other organ tumors, the influence of TAF15 on the MAPK signaling pathway emphasize that this protein could serve as a new molecular biomarker to predict tumor behavior and target therapeutic intervention in GI cancers. In conclusion, more studies should be performed to better understand the prognostic and therapeutic role of TAF15 in GI tumors, especially in tumors resistant to therapy.


Subject(s)
Biomarkers, Tumor , Gastrointestinal Neoplasms , TATA-Binding Protein Associated Factors , Humans , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/metabolism , TATA-Binding Protein Associated Factors/metabolism , TATA-Binding Protein Associated Factors/genetics , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/analysis , Prognosis , MAP Kinase Signaling System , Molecular Targeted Therapy/methods , Gene Expression Regulation, Neoplastic , Carcinogenesis/genetics
2.
Immun Inflamm Dis ; 12(8): e1348, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39105572

ABSTRACT

BACKGROUND: Mecapegfilgrastim, a long-acting granulocyte-colony stimulating factor has been approved for reducing the incidence of infection, particularly febrile neutropenia (FN), in China. OBJECTIVE: We conducted a multicenter prospective observational study to examine the safety and effectiveness of mecapegfilgrastim in preventing neutropenia in gastrointestinal patients receiving the chemotherapy, including S-1/capecitabine-based regimens or the fluorouracil, leucovorin, oxaliplatin, and irinotecan (FOLFOXIRI)/fluorouracil, leucovorin, and oxaliplatin (FOLFOX)/fluorouracil, leucovorin, oxaliplatin, and irinotecan (FOLFIRINOX) regimens. METHOD: Five hundred and sixty-one gastrointestinal patients from 40 sites across China, between May 2019 and November 2021, were included. The administration of mecapegfilgrastim was prescribed at the discretion of local physicians. RESULTS: The most common adverse drug reactions (ADRs) of any grade for all patients was increased white blood cells (2.9%). Grade 3/4 ADRs were observed for anemia (0.2%), decreased white blood cells (0.2%), and decreased neutrophil count (0.2%). Among the 116 patients who received S-1/capecitabine-based chemotherapy throughout all cycles, ADRs of any grade included anemia (1.7%), myalgia (0.9%), and increased alanine aminotransferase (0.9%). No grade 3/4 ADRs were observed. In 414 cycles of patients who underwent S-1/capecitabine-based regimens, only one (0.2%) cycle experienced grade 4 neutropenia. In the FOLFIRINOX, FOLFOXIRI, and FOLFOX chemotherapy regimens, grade 4 neutropenia occurred in one (2.7%) of 37 cycles, four (4.7%) of 85 cycles, and two (1.2%) of 167 cycles, respectively. CONCLUSION: In a real-world setting, mecapegfilgrastim has proven effective in preventing severe neutropenia in gastrointestinal patients following chemotherapy. This includes commonly used moderate or high-risk FN regimens or regimens containing S1/capecitabine, all of which have demonstrated favorable efficacy and safety profiles.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Fluorouracil , Gastrointestinal Neoplasms , Neutropenia , Humans , Male , Female , Middle Aged , Prospective Studies , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aged , Gastrointestinal Neoplasms/drug therapy , Neutropenia/prevention & control , Neutropenia/chemically induced , Neutropenia/epidemiology , Adult , Fluorouracil/adverse effects , Fluorouracil/therapeutic use , Leucovorin/therapeutic use , Leucovorin/adverse effects , Irinotecan/therapeutic use , Irinotecan/adverse effects , Oxaliplatin/adverse effects , Oxaliplatin/therapeutic use , China/epidemiology
3.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 128-133, 2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39097885

ABSTRACT

To assess the diagnostic efficacy of SEPT9 along with PAX5 gene methylation detection in gastrointestinal cancer and precancerous lesions, the peripheral blood of 62 patients with gastric cancer (GC) and 60 patients with no evidence of disease (as the control group) were retrospectively collected. The methylation rates of PAX5 and SEPT9 gene promoters in blood samples of GC group were detected by PCR. At the same time, the differences in methylation rates of genes in the two groups were compared, and the predictive value of plasma methylation PAX5 and SEPT9 in GC was evaluated by receiver operating characteristic (ROC) curve. We found that there were 41 cases of methylated PAX5 gene promoter region and 39 cases of methylated SEPT9 gene promoter region in GC group. The control group contained 14 cases of PAX5 gene promoter methylation and 12 cases of RNF¹80 gene promoter methylation. The occurrence of PAX5 promoter methylation was correlated with age of GC patients. There were statistically significant differences in mSEPT9 gene in patients with different TNM stages. Kaplan-Meier survival curve analysis revealed that the three-year overall survival rate of GC patients with PAX5 methylation was lower than that of GC patients without PAX5 methylation. No significant difference was discovered in 3-year overall survival rate between GC patients with SEPT9 methylation and those without SEPT9 methylation. Combined detection could not improve the diagnostic value of GC, but could promote diagnosis sensitivity. In summary, the risk of PAX5 and SEPT9 gene methylation in GC patients presents higher when compared with healthy people. PAX5 gene methylation is closely related to age, while SEPT9 is closely related to tumor TNM stage, and PAX5 gene methylation can decrease the survival rate of GC patients. Detection of PAX5 gene methylation level can assist in evaluating the prognosis of GC patients.


Subject(s)
DNA Methylation , PAX5 Transcription Factor , Precancerous Conditions , Promoter Regions, Genetic , Septins , Humans , Septins/genetics , DNA Methylation/genetics , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , Female , Male , Middle Aged , Promoter Regions, Genetic/genetics , Aged , Precancerous Conditions/genetics , Precancerous Conditions/diagnosis , Precancerous Conditions/pathology , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/pathology , ROC Curve , Kaplan-Meier Estimate , Stomach Neoplasms/genetics , Stomach Neoplasms/diagnosis , Stomach Neoplasms/pathology , Adult , Biomarkers, Tumor/genetics , Retrospective Studies , Neoplasm Staging
5.
Curr Oncol ; 31(8): 4455-4475, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39195316

ABSTRACT

Lymph node metastasis (LNM) is one of the major prognostic factors in human gastrointestinal carcinomas (GICs). The lymph node-positive patients have poorer survival than node-negative patients. LNM is directly associated with the recurrence and poor survival of patients with GICs. The early detection of LNM in patients and designing effective therapies to suppress LNM may significantly impact the survival of these patients. The rapid progress made in proteomic technologies could be successfully applied to identify molecular targets for cancers at high-throughput levels. LC-MS/MS analysis enables the identification of proteins involved in LN metastasis, which can be utilized for diagnostic and therapeutic applications. This review summarizes the studies on LN metastasis in GICs using proteomic approaches to date.


Subject(s)
Gastrointestinal Neoplasms , Lymphatic Metastasis , Proteomics , Humans , Proteomics/methods , Gastrointestinal Neoplasms/pathology
6.
Cells ; 13(16)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39195202

ABSTRACT

Cancer is a leading cause of death worldwide. Around one-third of the total global cancer incidence and mortality are related to gastrointestinal (GI) cancers. Over the past few years, rapid developments have been made in patient-derived organoid (PDO) models for gastrointestinal cancers. By closely mimicking the molecular properties of their parent tumors in vitro, PDOs have emerged as powerful tools in personalized medicine and drug discovery. Here, we review the current literature on the application of PDOs of common gastrointestinal cancers in the optimization of drug treatment strategies in the clinic and their rising importance in pre-clinical drug development. We discuss the advantages and limitations of gastrointestinal cancer PDOs and outline the microfluidics-based strategies that improve the throughput of PDO models in order to extract the maximal benefits in the personalized medicine and drug discovery process.


Subject(s)
Gastrointestinal Neoplasms , Organoids , Precision Medicine , Humans , Precision Medicine/methods , Organoids/drug effects , Organoids/pathology , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/pathology , Drug Evaluation, Preclinical/methods , Drug Discovery/methods , Drug Screening Assays, Antitumor/methods
7.
Anticancer Res ; 44(9): 3937-3943, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39197902

ABSTRACT

BACKGROUND/AIM: Intraoperative identification of the cancer location is often difficult during robot-assisted surgery, especially in early stage cancers. This study aimed to investigate the feasibility and accuracy of a novel endoscopic clip emitting near-infrared (NIR) fluorescence during robot-assisted surgery for gastrointestinal cancer. PATIENTS AND METHODS: Preoperative placement of endoscopic marking clips equipped with NIR fluorescent resin was performed to determine the resection margins in six patients with gastrointestinal cancer. During robot-assisted surgery, a NIR fluorescence imaging system was used to detect the fluorescence. The evaluation examined whether fluorescence from the clips was visualized during robot-assisted surgery. RESULTS: The NIR fluorescent signals emitted from the clips were successfully detected in all six patients from the serosal surfaces, resulting in the quick and accurate identification of the resection line. There were no significant differences in age, sex, or body mass index between the patients in whom we could detect NIR fluorescence. CONCLUSION: This novel NIR fluorescent clip is a promising diagnostic tool for accurately detecting tumor locations during robot-assisted surgery for gastrointestinal cancer.


Subject(s)
Gastrointestinal Neoplasms , Indocyanine Green , Optical Imaging , Robotic Surgical Procedures , Humans , Male , Female , Gastrointestinal Neoplasms/surgery , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/diagnostic imaging , Aged , Optical Imaging/methods , Robotic Surgical Procedures/methods , Middle Aged , Aged, 80 and over
8.
Turk J Gastroenterol ; 35(5): 354-359, 2024 May.
Article in English | MEDLINE | ID: mdl-39128095

ABSTRACT

BACKGROUND/AIMS:  The endoscopic features of small-bowel gastrointestinal stromal tumors (GISTs) are not well defined. The objective of this study was to describe the endoscopic features of GISTs of the small intestine detected via single-balloon enteroscopy (SBE). MATERIALS AND METHODS:  Patients with surgically confirmed small intestinal GISTs from January 2014 to September 2022 were retrospectively analyzed. The hospital's electronic medical record system was used to retrieve the patients' data, including their demographics, clinical symptoms, hemoglobin on admission, endoscopic and computerized tomography findings, clinicopathological findings, and surgical management data. RESULTS:  In total, 46 GIST patients (23 men and 23 women) with overt bleeding were included, with a mean age of 52 years (23-80 years). The typical duration of the symptoms was 48 hours. Four patients (8.70%) had lesions in the duodenum, 32 (69.56%) had lesions in the jejunum, 8 (17.39%) had lesions in the ileum, and 2 (4.35%) had lesions around the junction of the jejunum and ileum. Out of the 46 patients, 27 underwent SBE, and GISTs were visualized in 25, while the lesions could not be visualized in the remaining 2. Submucosal round (n = 13), submucosal sessile (n = 8), and invasive/penetrating (n = 4) were among the endoscopic tumor features. Twenty patients exhibited submucosal protuberant lesions, with ulceration, vascular nodules/congestion, or erosion on the surface, and 5 patients presented ulcerative infiltrative lesions. The multiple logistic regression analysis indicated that the invasive/penetrating characteristics of GISTs under SBE evaluation are significantly correlated with the risk level of GIST malignancy (P < .05). CONCLUSION:  A variety of endoscopic characteristics could be observed during the preoperative SBE evaluation of small-intestine GISTs.


Subject(s)
Gastrointestinal Stromal Tumors , Intestine, Small , Single-Balloon Enteroscopy , Humans , Gastrointestinal Stromal Tumors/pathology , Gastrointestinal Stromal Tumors/diagnostic imaging , Gastrointestinal Stromal Tumors/surgery , Gastrointestinal Stromal Tumors/diagnosis , Female , Middle Aged , Male , Retrospective Studies , Aged , Adult , Aged, 80 and over , Single-Balloon Enteroscopy/methods , Intestine, Small/pathology , Intestine, Small/diagnostic imaging , Young Adult , Gastrointestinal Hemorrhage/etiology , Intestinal Neoplasms/pathology , Intestinal Neoplasms/surgery , Intestinal Neoplasms/diagnostic imaging , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/diagnostic imaging , Gastrointestinal Neoplasms/surgery
9.
Pol J Pathol ; 75(2): 126-137, 2024.
Article in English | MEDLINE | ID: mdl-39166521

ABSTRACT

The incidence and prevalence of neuroendocrine neoplasms (NENs) in many organs are increasing. Although such NENs have similar grades, they may exhibit quite different behaviors. In this multicenter study, we aimed to investigate the prevalence and distribution of different morphological NEN variants in the non-pancreatic gastrointestinal (GI) tract and determine whether they can guide prognosis prediction. Two hundred and fifty-six patients diagnosed with NENs originating from the GI tract from 7 different centers were included in the study. In 89 (36.6%) cases, different morphological variants were detected. When the variants were grouped according to their aggressiveness as described in the literature, a statistically significant relationship between aggressiveness and the variables organ and age was found ( p < 0.05). The oncocytic variant was found to metastasize more than the other aggressive types (42.9%). The paraganglioma-like variant was found to have a smaller size, lower proliferation index, and a more benign clinical course. This study demonstrated that well-differentiated GI neuroendocrine tumors (GI-NETs) have considerable morphological diversity. Generally, case reports of rare morphological variants of GI-NETs are available in the literature. We believe that our study contributes to a better understanding of the prevalence, localization, and significance of morphological variations in GI-NETs.


Subject(s)
Gastrointestinal Neoplasms , Neuroendocrine Tumors , Humans , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/epidemiology , Female , Middle Aged , Male , Prevalence , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/epidemiology , Aged , Adult , Prognosis , Aged, 80 and over , Young Adult , Adolescent
10.
Cancer Lett ; 599: 217152, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39094825

ABSTRACT

Monoclonal antibodies targeting immune checkpoints have been widely applied in gastrointestinal cancer immunotherapy. However, systemic administration of various monoclonal antibodies does not often result in sustained effects in reversing the immunosuppressive tumor microenvironment (TME), which may be due to the spatiotemporal dynamic changes of immune checkpoints. Herein, we reported a novel immune checkpoint reprogramming strategy for gastrointestinal cancer immunotherapy. It was achieved by the sequential delivery of siPD-L1 (siRNA for programmed cell death ligand 1) and pOX40L (plasmid for OX40 ligand), which were complexed with two cationic polymer brush-grafted carbon nanotubes (dense short (DS) and dense long (DL)) designed based on the structural characteristics of nucleic acids and brush architectures. Upon administrating DL/pOX40L for the first three dosages, then followed by DS/siPD-L1 for the next three dosages to the TME, it upregulated the stimulatory checkpoint OX40L on dendritic cells (DCs) and downregulated inhibitory checkpoint PD-L1 on tumor cells and DCs in a sequential reprogramming manner. Compared with other combination treatments, this sequential strategy drastically boosted the DCs maturation, and CD8+ cytotoxic T lymphocytes infiltration in tumor site. Furthermore, it could augment the local antitumor response and improve the T cell infiltration in tumor-draining lymph nodes to reverse the peripheral immunosuppression. Our study demonstrated that sequential nucleic acid delivery strategy via personalized nanoplatforms effectively reversed the immunosuppression status in both tumor microenvironment and peripheral immune landscape, which significantly enhanced the systemic antitumor immune responses and established an optimal immunotherapy strategy against gastrointestinal cancer.


Subject(s)
B7-H1 Antigen , Dendritic Cells , Gastrointestinal Neoplasms , Immunotherapy , OX40 Ligand , Tumor Microenvironment , Animals , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , Mice , Immunotherapy/methods , Gastrointestinal Neoplasms/immunology , Gastrointestinal Neoplasms/therapy , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/genetics , B7-H1 Antigen/immunology , Humans , Dendritic Cells/immunology , Cell Line, Tumor , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Mice, Inbred C57BL , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/pharmacology , Female
11.
Biochem Pharmacol ; 227: 116463, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39102994

ABSTRACT

Gastrointestinal cancers (GICs) are highly prevalent cancers that threaten human health worldwide. The Wnt/ß-catenin signaling pathway has been reported to play a pivotal role in the carcinogenesis of GICs. Numerous interventions targeting the Wnt/ß-catenin signaling in GICs are currently being tested in clinical trials with promising results. Unfortunately, there are no clinically approved drugs that effectively target this pathway. This comprehensive review aims to evaluate the impact of clinical therapies targeting the Wnt/ß-catenin signaling pathway in GICs. By integrating data from bioinformatics databases and recent literature from the past five years, we examine the heterogeneous expression and regulatory mechanisms of Wnt/ß-catenin pathway genes and proteins in GICs. Specifically, we focus on expression patterns, mutation frequencies, and clinical prognoses to understand their implications for treatment strategies. Additionally, we discuss recent clinical trial efforts targeting this pathway. Understanding the inhibitors currently under clinical investigation may help optimize foundational research and clinical strategies. We hope that elucidating the current status of precision therapeutic stratification for patients targeting the Wnt/ß-catenin pathway will guide future innovations in precision medicine for GICs.


Subject(s)
Antineoplastic Agents , Gastrointestinal Neoplasms , Wnt Signaling Pathway , Humans , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/metabolism , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/physiology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Animals , beta Catenin/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/genetics , Molecular Targeted Therapy/methods
12.
Support Care Cancer ; 32(9): 592, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39150573

ABSTRACT

BACKGROUND: The effectiveness of generalist palliative care interventions in hospitals is unknown. AIM: This study aimed to explore the impact of a palliative care case management intervention for patients with gastrointestinal cancer (PalMaGiC) on hospital admissions, healthcare use, and place of death. DESIGN: This was a register-based cohort study analyzing data from the Danish Register on Causes of Death, the Danish National Patient Register, and the Danish Palliative Database. SETTING/PARTICIPANTS: Deceased patients with gastrointestinal cancer from 2010 to 2020 exposed to PalMaGiC were compared over three periods of time to patients receiving standard care. RESULTS: A total of 43,969 patients with gastrointestinal cancers were included in the study, of whom 1518 were exposed to PalMaGiC. In the last 30 days of life, exposed patients were significantly more likely to be hospitalized (OR of 1.62 (95% CI 1.26-2.01)), spend more days at the hospital, estimate of 1.21 (95% CI 1.02-1.44), and have a higher number of hospital admissions (RR of 1.13 (95% CI 1.01-1.27)), and were more likely to die at the hospital (OR of 1.94 (95% CI 1.55-2.44)) with an increasing trend over time. No differences were found for hospital healthcare use. CONCLUSION: Patients exposed to the PalMaGiC intervention had a greater likelihood of hospitalizations and death at the hospital compared to unexposed patients, despite the opposite intention. Sensitivity analyses show that regional differences may hold some of the explanation for this. Future development of generalist palliative care in hospitals should focus on integrating a home-based approach, community care, and PC physician involvement.


Subject(s)
Gastrointestinal Neoplasms , Palliative Care , Registries , Humans , Gastrointestinal Neoplasms/therapy , Palliative Care/methods , Palliative Care/statistics & numerical data , Male , Female , Denmark , Aged , Registries/statistics & numerical data , Cohort Studies , Aged, 80 and over , Middle Aged , Hospitalization/statistics & numerical data
13.
BMC Health Serv Res ; 24(1): 932, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143501

ABSTRACT

BACKGROUND: Upper gastrointestinal cancers (UGICs) are increasingly prevalent. With a poor prognosis and significant longer-term effects, UGICs present significant adjustment challenges for individuals with cancer and their informal caregivers. However, the supportive care needs of these informal caregivers are largely unknown. This systematic review of qualitative studies synthesises and critically evaluates the current evidence base on the experience of informal caregivers of individuals with UGIC. METHODS: A Joanna Briggs Institute systematic review was conducted. Searches were performed in four databases (MEDLINE, PsycINFO, Embase, CINAHL) from database inception to February 2021. Included studies explored experiences of informal caregivers of individuals diagnosed with primary cancer of the oesophagus, stomach, pancreas, bile duct, gallbladder, or liver. Studies were independently screened for eligibility and included studies were appraised for quality by two reviewers. Data were extracted and synthesised using meta-aggregation. RESULTS: 19 papers were included in this review, and 328 findings were extracted. These were aggregated into 16 categories across three findings: (1) UGIC caregiver burden; UGIC caregivers undertake extensive responsibilities, especially around patient diet as digestion is severely impacted by UGICs. (2) Mediators of caregiver burden; The nature of UGICs, characterised by disruptive life changes for caregivers, was identified as a mediator for caregiver burden. (3) Consequences of caregiver burden: UGIC caregivers' experiences were shaped by unmet needs, a lack of information and a general decline in social interaction. CONCLUSIONS: The findings of this review suggest the need for a cultural shift within health services. Caregiving for UGIC patients is suggested to adversely affect caregivers' quality of life, similarly to other cancer caregiving populations and therefore they should be better incorporated as co-clients in care-planning and execution by including them in discussions about the patient's diagnosis, treatment options, and potential side effects.


Subject(s)
Caregivers , Gastrointestinal Neoplasms , Humans , Caregivers/psychology , Gastrointestinal Neoplasms/psychology , Caregiver Burden/psychology , Qualitative Research , Quality of Life
14.
JAMA Netw Open ; 7(8): e2428828, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39158910

ABSTRACT

Importance: The incidence of gastrointestinal stromal tumors (GISTs) increased after the implementation of GIST-specific histology coding in 2001, but updated data on trends and survival are lacking. Objective: To examine the evolving epidemiology of GISTs in major organ sites. Design, Setting, and Participants: This descriptive, population-based cohort study used nationally representative data from the National Cancer Institute Surveillance, Epidemiology, and End Results (SEER) Program, including the SEER-22 and SEER-17 registries. Data were from evaluated patients aged 20 years or older with GISTs diagnosed between January 1, 2000, and December 31, 2019. Analyses were last updated on October 29, 2023. Main Outcomes and Measures: Organ site-specific trends in age-standardized incidence rates and annual percent changes (APCs) in rates were estimated by race and ethnicity and, when possible, by sex, age, and primary indicator. Multivariable Cox proportional hazards regression models were used to examine racial and ethnic differences in overall and GIST-specific survival by site. Results: The SEER-22 and SEER-17 datasets contained 23 001 and 12 109 case patients with GISTs, respectively. Patients in the SEER-22 registry had a mean (SD) age of 64 (13) years and 51.3% were men. With regard to race and ethnicity, 9.7% of patients were Asian or Pacific Islander, 12.3% were Hispanic, 19.6% were non-Hispanic Black, and 57.7% were non-Hispanic White. Overall incidence rates of GISTs in the SEER-22 cohort increased substantially over time for all organ sites but the colon (APCs: esophagus, 7.3% [95% CI, 4.4% to 10.2%]; gastric, 5.1% [95% CI, 4.2% to 6.1%]; small intestine, 2.7% [95% CI, 1.8% to 3.7%]; colon, -0.2% [95% CI, -1.3% to 0.9%]; and rectum, 1.9% [95% CI, 0.1% to 3.8%]). There were similar increasing trends by age groups (<50 vs ≥50 years), sex, race and ethnicity, and primary indicator for gastric and small intestine GISTs. Increases were mainly restricted to localized stage disease. Patients in the SEER-17 cohort had a mean (SD) age of 64 (14) years and 51.9% were men. With regard to race and ethnicity, 13.3% of patients were Asian or Pacific Islander, 11.6% were Hispanic, 17.8% were non-Hispanic Black, and 56.6% were non-Hispanic White. Non-Hispanic Black individuals had higher overall mortality for esophageal (adjusted hazard ratio [HR], 6.4 [95% CI, 2.0 to 20.3]) and gastric (adjusted HR, 1.4 [95% CI, 1.2 to 1.5]) GISTs compared with non-Hispanic White individuals. Asian or Pacific Islander individuals also had higher overall mortality for esophageal GISTs (adjusted HR, 5.6 [95% CI, 1.5 to 20.2]). Results were similar for GIST-specific survival. Conclusions and Relevance: In this cohort study using SEER data, the incidence of GISTs in major organ sites increased in the last 2 decades among several population groups. These findings suggest that additional studies are warranted to identify risk factors, because histologic reclassification and higher availability of endoscopy and imaging do not fully explain these unfavorable incidence trends. Prevention efforts are needed to reduce the substantial survival disparities among racial and ethnic minoritized populations.


Subject(s)
Gastrointestinal Stromal Tumors , SEER Program , Humans , Gastrointestinal Stromal Tumors/mortality , Gastrointestinal Stromal Tumors/epidemiology , Gastrointestinal Stromal Tumors/ethnology , Male , Female , Middle Aged , Incidence , Aged , United States/epidemiology , Adult , Cohort Studies , Aged, 80 and over , Gastrointestinal Neoplasms/mortality , Gastrointestinal Neoplasms/epidemiology
15.
Biomolecules ; 14(8)2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39199305

ABSTRACT

Gastrointestinal (GI) cancers impose a substantial global health burden, highlighting the necessity for deeper understanding of their intricate pathogenesis and treatment strategies. This review explores the interplay between intratumoral microbiota, tumor metabolism, and major types of GI cancers (including esophageal, gastric, liver, pancreatic, and colorectal cancers), summarizing recent studies and elucidating their clinical implications and future directions. Recent research revealed altered microbial signatures within GI tumors, impacting tumor progression, immune responses, and treatment outcomes. Dysbiosis-induced alterations in tumor metabolism, including glycolysis, fatty acid metabolism, and amino acid metabolism, play critical roles in cancer progression and therapeutic resistance. The integration of molecular mechanisms and potential biomarkers into this understanding further enhances the prognostic significance of intratumoral microbiota composition and therapeutic opportunities targeting microbiota-mediated tumor metabolism. Despite advancements, challenges remain in understanding the dynamic interactions within the tumor microenvironment (TME). Future research directions, including advanced omics technologies and prospective clinical studies, offer promising avenues for precision oncology and personalized treatment interventions in GI cancer. Overall, integrating microbiota-based approaches and molecular biomarkers into GI cancer management holds promise for improving patient outcomes and survival.


Subject(s)
Biomarkers, Tumor , Gastrointestinal Neoplasms , Tumor Microenvironment , Humans , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/microbiology , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Dysbiosis/microbiology , Dysbiosis/metabolism , Microbiota , Gastrointestinal Microbiome , Animals
16.
Int Immunopharmacol ; 140: 112728, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39098227

ABSTRACT

Imatinib-induced skin rash poses a significant challenge for patients with gastrointestinal stromal tumor, often resulting in treatment interruption or discontinuation and subsequent treatment failure. However, the underlying mechanism of imatinib-induced skin rashes in gastrointestinal stromal tumor patients remains unclear. A total of 51 patients (27 with rash and 24 without rash) were enrolled in our study. Blood samples were collected concomitantly with the onset of clinical manifestations of rashes, and simultaneously collecting clinical relevant information. The imatinib concentration and untargeted metabolomics were performed by ultra-high-performance liquid chromatography-tandem mass spectrometry. There were no significant differences in age, gender, imatinib concentration and white blood cells count between the rash group and the control group. However, the rash group exhibited a higher eosinophil count (P<0.05) and lower lymphocyte count (P<0.05) compared to the control group. Untargeted metabolomics analysis found that 105 metabolites were significantly differentially abundant. The univariate analysis highlighted erucamide, linoleoylcarnitine, and valine betaine as potential predictive markers (AUC≥0.80). Further enriched pathway analysis revealed primary metabolic pathways, including sphingolipid signaling pathway, sphingolipid metabolism, cysteine and methionine metabolism, biosynthesis of unsaturated fatty acids, arginine and proline metabolism, and biosynthesis of amino acids. These findings suggest that the selected differential metabolites could serve as a foundation for the prediction and management of imatinib-induced skin rash in gastrointestinal stromal tumor patients.


Subject(s)
Antineoplastic Agents , Exanthema , Gastrointestinal Neoplasms , Gastrointestinal Stromal Tumors , Imatinib Mesylate , Metabolomics , Humans , Gastrointestinal Stromal Tumors/drug therapy , Imatinib Mesylate/adverse effects , Imatinib Mesylate/therapeutic use , Female , Male , Middle Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Exanthema/chemically induced , Gastrointestinal Neoplasms/drug therapy , Aged , Adult
18.
Lancet Oncol ; 25(9): 1163-1175, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39127063

ABSTRACT

BACKGROUND: The long-term impact of tyrosine kinase inhibitor (TKI) discontinuation on resistance and survival in patients with advanced gastrointestinal stromal tumours (GIST) is unclear. We report the exploratory long-term outcomes of patients with advanced GIST stopping imatinib in the BFR14 trial. METHODS: BFR14, an open-label, randomised, phase 3 trial, was done in 17 comprehensive cancer centres or hospitals across France. Patients with advanced GIST aged 18 years or older with an Eastern Cooperative Oncology Group performance status of 0-3, no previous treatment with imatinib, and no previous malignancy were eligible. Patients were treated with oral imatinib 400 mg daily. Patients with a complete or partial response, or stable disease, according to Response Evaluation Criteria in Solid Tumours (1.0) at 1 year, 3 years, and 5 years from the start of treatment were randomly assigned (1:1) to treatment discontinuation until progression (interruption group) or treatment continuation until progression (continuation group). Randomisation was done centrally with computer-generated permuted blocks of two and six patients stratified by participating centre and presence or absence of residual disease on CT scan. The primary endpoint was progression-free survival. Secondary endpoints included time to imatinib resistance and overall survival. Analyses were conducted on an intention-to-treat basis in all randomly assigned patients who were not lost to follow-up. This trial is registered with ClinicalTrial.gov, NCT00367861. FINDINGS: Between May 12, 2003, and March 16, 2004, after 1 year of imatinib, 32 patients were randomly assigned to the interruption group and 26 to the continuation group. Between June 13, 2005, and May 30, 2007, after 3 years of imatinib, 25 patients were randomly assigned to the interruption group and 25 to the continuation group. Between Nov 9, 2007, and July 12, 2010, after 5 years of imatinib, 14 patients were randomly assigned to the interruption group and 13 to the continuation group. Median follow-up was 235·2 months (IQR 128·8-236·6) after the 1-year randomisation, 200·9 months (190·2-208·4) after the 3-year randomisation, and 164·5 months (134·4-176·4) after the 5-year randomisation. Median progression-free survival in the interruption group versus the continuation group after 1 year of imatinib was 6·1 months (95% CI 2·5-10·1) versus 27·8 months (19·5-37·9; hazard ratio [HR] 0·36 [95% CI 0·20-0·64], log-rank p=0·0003), after 3 years of imatinib was 7·0 months (3·5-11·7) versus 67·0 months (48·8-85·6; 0·15 [0·07-0·32], log-rank p<0·0001), and after 5 years of imatinib was 12·0 months (9·0-16·6) versus not reached (NR; NR-NR; 0·13 [0·03-0·58], log-rank p=0·0016). The median time to imatinib resistance after 1 year of imatinib was 28·7 months (95% CI 18·1-39·1) versus 90·6 months (25·3-156·1; HR 0·93 [95% CI 0·51-1·71], log-rank p=0·82), after 3 years was 66·2 months (43·0-89·6) versus 127·3 months (15·0-239·7; 0·35 [0·17-0·72, log-rank p=0·0028), and after 5 years was 58·6 months (0·0-167·4) versus NR (NR-NR; 0·24 [0·05-1·12], log-rank p=0·049). Median overall survival after 1 year of imatinib was 56·0 months (95% CI 30·3-82·9) versus 105·0 months (20·6-189·6; HR 0·84 [95% CI 0·46-1·54], log-rank p=0·57), after 3 years was 104·0 months (90·7-118·7) versus 134·0 months (89·7-178·3; 0·40 [0·20-0·82], log-rank p=0·0096), and after 5 years was NR (NR-NR) versus 110·4 months (82·7-154·1; 1·28 [0·41-3·99]; log-rank p=0·67), INTERPRETATION: Imatinib interruption in patients with GIST without progressive disease is not recommended. Imatinib interruption in non-progressing patients with GIST was associated with rapid progression, faster resistance to imatinib, and shorter overall survival in the long-term follow-up when compared with imatinib continuation in patients after 3 years and 5 years of imatinib. FUNDING: Centre Léon Bérard, INCa, CONTICANET, Ligue Contre le Cancer, and Novartis.


Subject(s)
Gastrointestinal Stromal Tumors , Imatinib Mesylate , Protein Kinase Inhibitors , Humans , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/pathology , Gastrointestinal Stromal Tumors/mortality , Imatinib Mesylate/administration & dosage , Imatinib Mesylate/therapeutic use , Male , Female , Middle Aged , Aged , Follow-Up Studies , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/adverse effects , France , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/mortality , Progression-Free Survival , Adult , Time Factors , Drug Resistance, Neoplasm , Withholding Treatment/statistics & numerical data , Drug Administration Schedule
19.
World J Gastroenterol ; 30(31): 3628-3634, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39192999

ABSTRACT

This editorial comments on the manuscript by Chang et al, focusing on the still elusive interplay between epigenetic regulation and autophagy in gastrointestinal diseases, particularly cancer. Autophagy, essential for cellular homeostasis, exhibits diverse functions ranging from cell survival to death, and is particularly implicated in physiological gastrointestinal cell functions. However, its role in pathological backgrounds remains intricate and context-dependent. Studies underscore the dual nature of autophagy in cancer, where its early suppressive effects in early stages are juxtaposed with its later promotion, contributing to chemoresistance. This discrepancy is attributed to the dysregulation of autophagy-related genes and their intricate involvement in cellular processes. Epigenetic modifications and regulations of gene expression, including non-coding RNAs (ncRNAs), emerge as critical players in exerting regulatory control over autophagy flux, influencing treatment responses and tumor progression. Targeting epigenetic mechanisms and improving strategies involving the inhibition or induction of autophagy through pharmacological or genetic means present potential avenues to sensitize tumor cells to chemotherapy. Additionally, nanocarrier-based delivery of ncRNAs offers innovative therapeutic approaches. Understanding the intricate interaction between autophagy and ncRNA regulation opens avenues for the development of targeted therapies, thereby improving the prognosis of gastrointestinal malignancies with poor outcomes.


Subject(s)
Autophagy , Epigenesis, Genetic , Gastrointestinal Neoplasms , Gene Expression Regulation, Neoplastic , RNA, Untranslated , Humans , Autophagy/drug effects , Autophagy/genetics , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Drug Resistance, Neoplasm/genetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology
20.
BMC Palliat Care ; 23(1): 214, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39182074

ABSTRACT

BACKGROUND: Cancer cachexia is characterized by the loss of body weight (BW) and anorexia. Anamorelin (ANAM) is a selective ghrelin receptor agonist with appetite-enhancing anabolic action. The ONO-7643-05 trial demonstrated that ANAM increased lean body mass and improved anorexia in a Japanese population. However, the clinical outcomes of patients on ANAM have not yet been reported. PATIENTS AND METHODS: We investigated the clinical outcomes of patients with unresectable, advanced, or recurrent gastrointestinal cancer (colorectal, gastric, or pancreatic cancer) who were treated with ANAM between April 2017 and August 2022. Cachexia was defined as the presence of anorexia and a loss of ≥ 5% of BW within 6 months. To evaluate the response to ANAM, the patients who had discontinued ANAM within 3 weeks were excluded. Response to ANAM was defined as maintenance of or increase in BW and improved appetite from baseline at every 3-week evaluation. We also collected data on the reasons for the discontinuation of ANAM and the correlation between clinical factors and ANAM response. Safety analysis of ANAM was performed for all patients who received ANAM. RESULTS: Seventy-four patients were included in this study (49 males and 25 females), with a median age of 67.1 years (range, 36-83). The primary tumors were colorectal cancer in 27 (36.5%), gastric cancer in 20 (27.0%), and pancreatic cancer in 27 (36.5%). The Eastern Cooperative Oncology Group performance status was 0 in 10 (13.5%), 1 in 44 (59.5%), and ≥ 2 in 20 (27.0%). The number of previous chemotherapy regimens was 0 in 20 (27.0%), 1 in 22 (29.7%), and ≥ 2 in 32 (43.2%). ANAM was discontinued within 3 weeks in 28 patients for the following reasons: low-grade (grade 1 or 2) adverse events in 15 patients, ileus in three, grade 3 fatigue in one, progressive disease in one, censored follow-up in six, and unknown reasons in three. The proportion of ANAM responders was 63.6% (95% confidence interval, 47.8-77.6%). Among baseline characteristics, age ≥ 75 attenuated the ANAM response (p = 0.03). ANAM responders showed better disease control with chemotherapy than non-responders (75.0% vs. 37.5%, p = 0.02). CONCLUSIONS: ANAM may improve the outcomes of patients with gastrointestinal cancer cachexia in clinical practice.


Subject(s)
Cachexia , Gastrointestinal Neoplasms , Humans , Male , Cachexia/drug therapy , Cachexia/etiology , Female , Aged , Middle Aged , Gastrointestinal Neoplasms/complications , Gastrointestinal Neoplasms/drug therapy , Aged, 80 and over , Adult , Oligopeptides/therapeutic use , Oligopeptides/pharmacology , Japan , Hydrazines
SELECTION OF CITATIONS
SEARCH DETAIL