Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 16.463
2.
PLoS One ; 19(5): e0301969, 2024.
Article En | MEDLINE | ID: mdl-38771787

PURPOSE: This study aims to introduce an innovative multi-step pipeline for automatic tumor-stroma ratio (TSR) quantification as a potential prognostic marker for pancreatic cancer, addressing the limitations of existing staging systems and the lack of commonly used prognostic biomarkers. METHODS: The proposed approach involves a deep-learning-based method for the automatic segmentation of tumor epithelial cells, tumor bulk, and stroma from whole-slide images (WSIs). Models were trained using five-fold cross-validation and evaluated on an independent external test set. TSR was computed based on the segmented components. Additionally, TSR's predictive value for six-month survival on the independent external dataset was assessed. RESULTS: Median Dice (inter-quartile range (IQR)) of 0.751(0.15) and 0.726(0.25) for tumor epithelium segmentation on internal and external test sets, respectively. Median Dice of 0.76(0.11) and 0.863(0.17) for tumor bulk segmentation on internal and external test sets, respectively. TSR was evaluated as an independent prognostic marker, demonstrating a cross-validation AUC of 0.61±0.12 for predicting six-month survival on the external dataset. CONCLUSION: Our pipeline for automatic TSR quantification offers promising potential as a prognostic marker for pancreatic cancer. The results underscore the feasibility of computational biomarker discovery in enhancing patient outcome prediction, thus contributing to personalized patient management.


Biomarkers, Tumor , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/mortality , Prognosis , Female , Stromal Cells/pathology , Male , Deep Learning , Aged , Middle Aged , Image Processing, Computer-Assisted/methods
3.
Cancer J ; 30(3): 185-193, 2024.
Article En | MEDLINE | ID: mdl-38753753

ABSTRACT: Neuroendocrine tumors (NETs) are rare tumors that develop from cells of the neuroendocrine system and can originate in multiple organs and tissues such as the bowels, pancreas, adrenal glands, ganglia, thyroid, and lungs. This review will focus on gastroenteropancreatic NETs (more commonly called NETs) characterized by frequent somatostatin receptor (SSTR) overexpression and pheochromocytomas/paragangliomas (PPGLs), which typically overexpress norepinephrine transporter. Advancements in SSTR-targeted imaging and treatment have revolutionized the management of patients with NETs. This comprehensive review delves into the current practice, discussing the use of the various Food and Drug Administration-approved SSTR-agonist positron emission tomography tracers and the predictive imaging biomarkers, and elaborating on 177Lu-DOTATATE peptide receptor radionuclide therapy including the evolving areas of posttherapy imaging practices and peptide receptor radionuclide therapy retreatment. SSTR-targeted imaging and therapy can also be used in patients with PPGL; however, this patient population has demonstrated the best outcomes from norepinephrine transporter-targeted therapy with 131I-metaiodobenzylguanidine. Metaiodobenzylguanidine theranostics for PPGL will be discussed, noting that in 2024 it became commercially unavailable in the United States. Therefore, the use and reported success of SSTR theranostics for PPGL will also be explored.


Neuroendocrine Tumors , Humans , Neuroendocrine Tumors/therapy , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/pathology , Receptors, Somatostatin/metabolism , Radiopharmaceuticals/therapeutic use , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/pathology , Theranostic Nanomedicine/methods , Precision Medicine/methods , Positron-Emission Tomography/methods , Intestinal Neoplasms/therapy , Intestinal Neoplasms/diagnosis , Intestinal Neoplasms/pathology
4.
J Exp Clin Cancer Res ; 43(1): 145, 2024 May 15.
Article En | MEDLINE | ID: mdl-38750539

BACKGROUND: Plasma cell-free DNA (cfDNA) fragmentomics has demonstrated significant differentiation power between cancer patients and healthy individuals, but little is known in pancreatic and biliary tract cancers. The aim of this study is to characterize the cfDNA fragmentomics in biliopancreatic cancers and develop an accurate method for cancer detection. METHODS: One hundred forty-seven patients with biliopancreatic cancers and 71 non-cancer volunteers were enrolled, including 55 patients with cholangiocarcinoma, 30 with gallbladder cancer, and 62 with pancreatic cancer. Low-coverage whole-genome sequencing (median coverage: 2.9 ×) was performed on plasma cfDNA. Three cfDNA fragmentomic features, including fragment size, end motif and nucleosome footprint, were subjected to construct a stacked machine learning model for cancer detection. Integration of carbohydrate antigen 19-9 (CA19-9) was explored to improve model performance. RESULTS: The stacked model presented robust performance for cancer detection (area under curve (AUC) of 0.978 in the training cohort, and AUC of 0.941 in the validation cohort), and remained consistent even when using extremely low-coverage sequencing depth of 0.5 × (AUC: 0.905). Besides, our method could also help differentiate biliopancreatic cancer subtypes. By integrating the stacked model and CA19-9 to generate the final detection model, a high accuracy in distinguishing biliopancreatic cancers from non-cancer samples with an AUC of 0.995 was achieved. CONCLUSIONS: Our model demonstrated ultrasensitivity of plasma cfDNA fragementomics in detecting biliopancreatic cancers, fulfilling the unmet accuracy of widely-used serum biomarker CA19-9, and provided an affordable way for accurate noninvasive biliopancreatic cancer screening in clinical practice.


Biliary Tract Neoplasms , Cell-Free Nucleic Acids , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/blood , Biliary Tract Neoplasms/genetics , Biliary Tract Neoplasms/diagnosis , Biliary Tract Neoplasms/blood , Male , Female , Middle Aged , Aged , Biomarkers, Tumor/blood , Adult
5.
Expert Rev Mol Diagn ; 24(5): 355-362, 2024 May.
Article En | MEDLINE | ID: mdl-38708441

INTRODUCTION: A marked histomolecular heterogeneity characterizes pancreatic cancer. Thus, different tumor histologies with divergent genomic profiles exist within the same category. AREAS COVERED: Using data from PubMed, SCOPUS, and Embase (last search date: 04/04/2024), this expert-based, narrative review presents and discusses the essential molecular determinants of biological aggressiveness and poor prognosis in pancreatic cancer. First, KRAS mutation still represents one of the most critical difficulties in treating pancreatic cancers. In this district, it is mutated in > 90% of malignant tumors. Notably, actionable alterations for molecular-based therapies are typically lacking in KRAS-mutated pancreatic cancer. Furthermore, transcriptome-based studies clarified that the squamous phenotype is characterized by poorer prognosis and response to standard chemotherapy. We also discuss molecular biomarkers related to dismal prognosis in specific subsets of pancreatic cancer, such as SMAD4 in signet-ring cell carcinoma and TP53 in invasive cancers derived from intraductal tubulopapillary neoplasms. EXPERT OPINION: The identification of the subgroups of pancreatic cancer with particularly unfavorable prognoses is a critical step for addressing specific research efforts. In addition to implementing and strengthening current precision oncology strategies, the decisive step for improving the survival of patients affected by pancreatic cancer must pass through targeting the KRAS gene.


Biomarkers, Tumor , Mutation , Pancreatic Neoplasms , Proto-Oncogene Proteins p21(ras) , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/mortality , Prognosis , Proto-Oncogene Proteins p21(ras)/genetics , Biomarkers, Tumor/genetics , Genomics/methods
6.
Zhonghua Nei Ke Za Zhi ; 63(5): 474-479, 2024 May 01.
Article Zh | MEDLINE | ID: mdl-38715484

Objective: Objective To analyze the relationship between the survival outcomes of pancreatic cancer patients with obstructive jaundice and various clinical and pathological factors. Methods: A case series study was conducted, where clinical data from pancreatic cancer patients with obstructive jaundice, who were admitted to the Cancer Hospital of Tianjin Medical University between March 2022 and May 2023, were retrospectively gathered. Factors potentially affecting patient prognosis were initially analyzed using univariate analysis, followed by multivariate analysis using the Cox regression model for selected factors. A P-value of less than 0.05 was deemed statistically significant. Results: The study included 104 patients, comprising 69 males and 35 females, with a median age of 62 years (ranging from 38 to 85 years). Of these, 76 patients (73.1%) were followed until death, with a median survival time of 8.9 (6.2,11.5) months. The number of deaths versus surviving cases at 6 and 12 months were 20/75 and 64/14, respectively, resulting in estimated survival rates of 79.6% and 22.8%. Univariate analysis identified factors such as weight loss, primary site, TNM stage, liver metastasis, number of organs with tumor, stage at which jaundice appeared, CA19-9 levels, albumin levels, and D-dimer levels as significant in influencing prognosis (all P<0.05). Multivariate analysis revealed TNM stage, number of organs with tumor, method of jaundice treatment, albumin levels, and D-dimer levels as independent prognostic factors (all P<0.05). Conclusion: In pancreatic cancer patients presenting with obstructive jaundice, close monitoring of weight loss, primary site, TNM stage, liver metastasis, number of organs with tumor, the timing of jaundice occurrence, method of jaundice treatment, CA19-9, albumin, and D-dimer levels is crucial, as these factors may significantly impact the patient's survival and prognosis.


Jaundice, Obstructive , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/diagnosis , Male , Female , Middle Aged , Prognosis , Aged , Retrospective Studies , Adult , Aged, 80 and over , Survival Rate
7.
BMC Cancer ; 24(1): 555, 2024 May 03.
Article En | MEDLINE | ID: mdl-38702616

Periampullary cancers, including pancreatic ductal adenocarcinoma, ampullary-, cholangio-, and duodenal carcinoma, are frequently diagnosed in an advanced stage and are associated with poor overall survival. They are difficult to differentiate from each other and challenging to distinguish from benign periampullary disease preoperatively. To improve the preoperative diagnostics of periampullary neoplasms, clinical or biological markers are warranted.In this study, 28 blood plasma amino acids and derivatives from preoperative patients with benign (N = 45) and malignant (N = 72) periampullary disease were analyzed by LC-MS/MS.Principal component analysis and consensus clustering both separated the patients with cancer and the patients with benign disease. Glutamic acid had significantly higher plasma expression and 15 other metabolites significantly lower plasma expression in patients with malignant disease compared with patients having benign disease. Phenylalanine was the only metabolite associated with improved overall survival (HR = 0.50, CI 0.30-0.83, P < 0.01).Taken together, plasma metabolite profiles from patients with malignant and benign periampullary disease were significantly different and have the potential to distinguish malignant from benign disease preoperatively.


Amino Acids , Biomarkers, Tumor , Humans , Male , Female , Amino Acids/blood , Middle Aged , Aged , Biomarkers, Tumor/blood , Ampulla of Vater/pathology , Tandem Mass Spectrometry , Diagnosis, Differential , Common Bile Duct Neoplasms/blood , Common Bile Duct Neoplasms/diagnosis , Common Bile Duct Neoplasms/surgery , Common Bile Duct Neoplasms/pathology , Duodenal Neoplasms/blood , Duodenal Neoplasms/diagnosis , Duodenal Neoplasms/pathology , Duodenal Neoplasms/surgery , Adult , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/surgery , Pancreatic Neoplasms/mortality , Chromatography, Liquid , Principal Component Analysis , Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/pathology
8.
Sci Rep ; 14(1): 10199, 2024 05 03.
Article En | MEDLINE | ID: mdl-38702437

In pancreatic ductal adenocarcinoma (PDAC) patients, the importance of peritoneal lavage cytology, which indicates unresectability, remains controversial. This study sought to determine whether positive peritoneal lavage cytology (CY+) precludes pancreatectomy. Furthermore, we propose a novel liquid biopsy using peritoneal lavage fluid to detect viable peritoneal tumor cells (v-PTCs) with TelomeScan F35, a telomerase-specific replication-selective adenovirus engineered to express green fluorescent protein. Resectable cytologically or histologically proven PDAC patients (n = 53) were enrolled. CY was conducted immediately following laparotomy. The resulting fluid was examined by conventional cytology (conv-CY; Papanicolaou staining and MOC-31 immunostaining) and by the novel technique (Telo-CY; using TelomeScan F35). Of them, 5 and 12 were conv-CY+ and Telo-CY+, respectively. All underwent pancreatectomy. The two double-CY+ (conv-CY+ and Telo-CY+) patients showed early peritoneal recurrence (P-rec) postoperatively, despite adjuvant chemotherapy. None of the three conv-CY+ Telo-CY- patients exhibited P-rec. Six of the 10 Telo-CY+ conv-CY- patients (60%) relapsed with P-rec. Of the remaining 38 double-CY- [conv-CY-, Telo-CY-, conv-CY± (Class III)] patients, 3 (8.3%) exhibited P-rec. Although conv-CY+ status predicted poor prognosis and a higher risk of P-rec, Telo-CY was more sensitive for detecting v-PTC. Staging laparoscopy and performing conv-CY and Telo-CY are needed to confirm the indication for pancreatectomy.


Carcinoma, Pancreatic Ductal , Pancreatectomy , Pancreatic Neoplasms , Peritoneal Lavage , Humans , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/surgery , Male , Female , Aged , Middle Aged , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/diagnosis , Cytodiagnosis/methods , Aged, 80 and over , Neoplasm Recurrence, Local/pathology , Liquid Biopsy/methods , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/diagnosis , Adult , Cytology
9.
Mymensingh Med J ; 33(2): 533-539, 2024 Apr.
Article En | MEDLINE | ID: mdl-38557537

In biliary-pancreatic malignancy, the serum CA 19-9 is considered as a tumor marker. Its high level may indicate the presence of a malignant disorder, but it can also be raised in benign conditions and also in malignancies from other organs. The value may be normal even in malignant condition. This comparative study was conducted in the Department of Surgery of Bangabandhu Sheikh Mujib Medical University (BSMMU), Bangladesh from 1st June 2016 to 31st May 2017 to determine the sensitivity and specificity of CA 19-9 as a tumor marker in pancreatic malignancy in our perspective and to establish a cut-off value of CA 19-9 which might prove as a definitive indication of pancreatic malignancy. We found that when the cut off value of CA 19-9 is 38 U/ml (according to ROC curve), the sensitivity, specificity, PPV and NPV were 77.8%, 77.8%, 77.8%, 77.8% respectively. And if the serum CA 19-9 threshold was raised to 100 and 120 to diagnose pancreatic cancer, sensitivity became 72.2% and 66.7% and NPV 76.2% and 73.9% respectively. However, the specificity increased to 88.9% and 94.4% and the PPV increased to 86.7% and 92.3% respectively.


Biomarkers, Tumor , Pancreatic Neoplasms , Humans , Sensitivity and Specificity , ROC Curve , Pancreatic Neoplasms/diagnosis , Bangladesh
10.
JCO Precis Oncol ; 8: e2300355, 2024 Mar.
Article En | MEDLINE | ID: mdl-38564682

PURPOSE: Pancreatic cancer (PC) is a deadly disease most often diagnosed in late stages. Identification of high-risk subjects could both contribute to preventative measures and help diagnose the disease at earlier timepoints. However, known risk factors, assessed independently, are currently insufficient for accurately stratifying patients. We use large-scale data from the UK Biobank (UKB) to identify genetic variant-smoking interaction effects and show their importance in risk assessment. METHODS: We draw data from 15,086,830 genetic variants and 315,512 individuals in the UKB. There are 765 cases of PC. Crucially, robust resampling corrections are used to overcome well-known challenges in hypothesis testing for interactions. Replication analysis is conducted in two independent cohorts totaling 793 cases and 570 controls. Integration of functional annotation data and construction of polygenic risk scores (PRS) demonstrate the additional insight provided by interaction effects. RESULTS: We identify the genome-wide significant variant rs77196339 on chromosome 2 (per minor allele odds ratio in never-smokers, 2.31 [95% CI, 1.69 to 3.15]; per minor allele odds ratio in ever-smokers, 0.53 [95% CI, 0.30 to 0.91]; P = 3.54 × 10-8) as well as eight other loci with suggestive evidence of interaction effects (P < 5 × 10-6). The rs77196339 region association is validated (P < .05) in the replication sample. PRS incorporating interaction effects show improved discriminatory ability over PRS of main effects alone. CONCLUSION: This study of genome-wide germline variants identified smoking to modify the effect of rs77196339 on PC risk. Interactions between known risk factors can provide critical information for identifying high-risk subjects, given the relative inadequacy of models considering only main effects, as demonstrated in PRS. Further studies are necessary to advance toward comprehensive risk prediction approaches for PC.


Genetic Predisposition to Disease , Pancreatic Neoplasms , Humans , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Smoking/genetics , Smoking/adverse effects , Risk Factors , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Germ Cells
12.
Am J Surg Pathol ; 48(5): 511-520, 2024 May 01.
Article En | MEDLINE | ID: mdl-38567813

The diagnosis of solid pseudopapillary neoplasm of the pancreas (SPN) can be challenging due to potential confusion with other pancreatic neoplasms, particularly pancreatic neuroendocrine tumors (NETs), using current pathological diagnostic markers. We conducted a comprehensive analysis of bulk RNA sequencing data from SPNs, NETs, and normal pancreas, followed by experimental validation. This analysis revealed an increased accumulation of peroxisomes in SPNs. Moreover, we observed significant upregulation of the peroxisome marker ABCD1 in both primary and metastatic SPN samples compared with normal pancreas and NETs. To further investigate the potential utility of ABCD1 as a diagnostic marker for SPN via immunohistochemistry staining, we conducted verification in a large-scale patient cohort with pancreatic tumors, including 127 SPN (111 primary, 16 metastatic samples), 108 NET (98 nonfunctional pancreatic neuroendocrine tumor, NF-NET, and 10 functional pancreatic neuroendocrine tumor, F-NET), 9 acinar cell carcinoma (ACC), 3 pancreatoblastoma (PB), 54 pancreatic ductal adenocarcinoma (PDAC), 20 pancreatic serous cystadenoma (SCA), 19 pancreatic mucinous cystadenoma (MCA), 12 pancreatic ductal intraepithelial neoplasia (PanIN) and 5 intraductal papillary mucinous neoplasm (IPMN) samples. Our results indicate that ABCD1 holds promise as an easily applicable diagnostic marker with exceptional efficacy (AUC=0.999, sensitivity=99.10%, specificity=100%) for differentiating SPN from NET and other pancreatic neoplasms through immunohistochemical staining.


Carcinoma, Pancreatic Ductal , Neuroendocrine Tumors , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreas/pathology , Carcinoma, Pancreatic Ductal/pathology , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Pancreatic Ducts/chemistry , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis , ATP Binding Cassette Transporter, Subfamily D, Member 1
13.
Cells ; 13(7)2024 Mar 29.
Article En | MEDLINE | ID: mdl-38607041

Pancreatic cancer is a highly lethal disease with a 5-year survival rate of around 11-12%. Surgery, being the treatment of choice, is only possible in 20% of symptomatic patients. The main reason is that when it becomes symptomatic, IT IS the tumor is usually locally advanced and/or has metastasized to distant organs; thus, early diagnosis is infrequent. The lack of specific early symptoms is an important cause of late diagnosis. Unfortunately, diagnostic tumor markers become positive at a late stage, and there is a lack of early-stage markers. Surgical and non-surgical cases are treated with neoadjuvant and/or adjuvant chemotherapy, and the results are usually poor. However, personalized targeted therapy directed against tumor drivers may improve this situation. Until recently, many pancreatic tumor driver genes/proteins were considered untargetable. Chemical and physical characteristics of mutated KRAS are a formidable challenge to overcome. This situation is slowly changing. For the first time, there are candidate drugs that can target the main driver gene of pancreatic cancer: KRAS. Indeed, KRAS inhibition has been clinically achieved in lung cancer and, at the pre-clinical level, in pancreatic cancer as well. This will probably change the very poor outlook for this disease. This paper reviews the genetic characteristics of sporadic and hereditary predisposition to pancreatic cancer and the possibilities of a personalized treatment according to the genetic signature.


Lung Neoplasms , Pancreatic Neoplasms , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/diagnosis , Lung Neoplasms/genetics , Neoadjuvant Therapy , Biomarkers, Tumor/genetics
14.
Gigascience ; 132024 Jan 02.
Article En | MEDLINE | ID: mdl-38608280

Pancreatic ductal adenocarcinoma (PDAC) remains a lethal malignancy, largely due to the paucity of reliable biomarkers for early detection and therapeutic targeting. Existing blood protein biomarkers for PDAC often suffer from replicability issues, arising from inherent limitations such as unmeasured confounding factors in conventional epidemiologic study designs. To circumvent these limitations, we use genetic instruments to identify proteins with genetically predicted levels to be associated with PDAC risk. Leveraging genome and plasma proteome data from the INTERVAL study, we established and validated models to predict protein levels using genetic variants. By examining 8,275 PDAC cases and 6,723 controls, we identified 40 associated proteins, of which 16 are novel. Functionally validating these candidates by focusing on 2 selected novel protein-encoding genes, GOLM1 and B4GALT1, we demonstrated their pivotal roles in driving PDAC cell proliferation, migration, and invasion. Furthermore, we also identified potential drug repurposing opportunities for treating PDAC. SIGNIFICANCE: PDAC is a notoriously difficult-to-treat malignancy, and our limited understanding of causal protein markers hampers progress in developing effective early detection strategies and treatments. Our study identifies novel causal proteins using genetic instruments and subsequently functionally validates selected novel proteins. This dual approach enhances our understanding of PDAC etiology and potentially opens new avenues for therapeutic interventions.


Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Proteome , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/genetics , Glycosyltransferases , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Biomarkers , Membrane Proteins
15.
BMC Cancer ; 24(1): 478, 2024 Apr 15.
Article En | MEDLINE | ID: mdl-38622651

BACKGROUND: Pancreatic cancer is the foremost contributor to cancer-related deaths globally, and its prevalence continues to rise annually. Nevertheless, the underlying mechanisms behind its development remain unclear and necessitate comprehensive investigation. METHODS: In this study, a total of 29 fresh stool samples were collected from patients diagnosed with pancreatic cancer. The gut microbial data of healthy controls were obtained from the SRA database (SRA data number: SRP150089). Additionally, 28 serum samples and diseased tissues were collected from 14 patients with confirmed pancreatic cancer and 14 patients with chronic pancreatitis. Informed consent was obtained from both groups of patients. Microbial sequencing was performed using 16s rRNA. RESULTS: The results showed that compared with healthy controls, the species abundance index of intestinal flora in patients with pancreatic cancer was increased (P < 0.05), and the number of beneficial bacteria at the genus level was reduced (P < 0.05). Compared with patients with chronic pancreatitis, the expression levels of CA242 and CA199 in the serum of patients with pancreatic cancer were increased (P < 0.05). The bacterial richness index of tumor microorganisms in patients with pancreatic cancer increased, while the diversity index decreased(P < 0.05). Furthermore, there was a change in the species composition at the genus level. Additionally, the expression level of CA242 was found to be significantly positively correlated with the relative abundance of Acinetobacter(P < 0.05). CONCLUSION: Over all, the expression levels of serum tumor markers CA242 and CA19-9 in patients with pancreatic cancer are increased, while the beneficial bacteria in the intestine and tumor microenvironment are reduced and pathogenic bacteria are increased. Acinetobacter is a specific bacterial genus highly expressed in pancreatic cancer tissue.


Microbiota , Pancreatic Neoplasms , Pancreatitis, Chronic , Humans , Antigens, Tumor-Associated, Carbohydrate , RNA, Ribosomal, 16S/genetics , Pancreatic Neoplasms/diagnosis , Bacteria/genetics , Pancreatitis, Chronic/genetics , Tumor Microenvironment
17.
J Natl Compr Canc Netw ; 22(4): 237-243, 2024 04 17.
Article En | MEDLINE | ID: mdl-38631387

BACKGROUND: Germline genetic testing is a vital component of guideline-recommended cancer care for males with pancreatic, breast, or metastatic prostate cancers. We sought to determine whether there were racial disparities in germline genetic testing completion in this population. PATIENTS AND METHODS: This retrospective cohort study included non-Hispanic White and Black males with incident pancreatic, breast, or metastatic prostate cancers between January 1, 2019, and September 30, 2021. Two nationwide cohorts were examined: (1) commercially insured individuals in an administrative claims database, and (2) Veterans receiving care in the Veterans Health Administration. One-year germline genetic testing rates were estimated by using Kaplan-Meier methods. Cox proportional hazards regression was used to test the association between race and genetic testing completion. Causal mediation analyses were performed to investigate whether socioeconomic variables contributed to associations between race and germline testing. RESULTS: Our cohort consisted of 7,894 males (5,142 commercially insured; 2,752 Veterans). One-year testing rates were 18.0% (95% CI, 16.8%-19.2%) in commercially insured individuals and 14.2% (95% CI, 11.5%-15.0%) in Veterans. Black race was associated with a lower hazard of testing among commercially insured individuals (adjusted hazard ratio [aHR], 0.73; 95% CI, 0.58-0.91; P=.005) but not among Veterans (aHR, 0.99; 95% CI, 0.75-1.32; P=.960). In commercially insured individuals, income (aHR, 0.90; 95% CI, 0.86-0.96) and net worth (aHR, 0.92; 95% CI, 0.86-0.98) mediated racial disparities, whereas education (aHR, 0.98; 95% CI, 0.94-1.01) did not. CONCLUSIONS: Overall rates of guideline-recommended genetic testing are low in males with pancreatic, breast, or metastatic prostate cancers. Racial disparities in genetic testing among males exist in a commercially insured population, mediated by net worth and household income; these disparities are not seen in the equal-access Veterans Health Administration. Alleviating financial and access barriers may mitigate racial disparities in genetic testing.


Genetic Testing , Pancreatic Neoplasms , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/diagnosis , Genetic Testing/statistics & numerical data , Genetic Testing/methods , Middle Aged , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/diagnosis , Retrospective Studies , Aged , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/diagnosis , Healthcare Disparities/statistics & numerical data , Germ-Line Mutation , Breast Neoplasms, Male/genetics , Breast Neoplasms, Male/diagnosis , Breast Neoplasms, Male/pathology , United States , Adult , Genetic Predisposition to Disease , Black or African American/statistics & numerical data , Black or African American/genetics
18.
Biomarkers ; 29(4): 194-204, 2024 Jun.
Article En | MEDLINE | ID: mdl-38644767

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.


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
19.
J Am Soc Cytopathol ; 13(3): 213-218, 2024.
Article En | MEDLINE | ID: mdl-38575468

INTRODUCTION: Insulinoma-associated protein 1 (INSM1) is an immunohistochemical marker commonly used to confirm cytomorphological concordant neuroendocrine tumors/carcinomas (NETs/NECs), demonstrating high utility in small samples. Previous reports have suggested comparable INSM1 staining in CytoLyt-fixed cell blocks and formalin-fixed surgical pathology specimens. This study aimed to assess INSM1 immunoreactivity using both fixation methods and investigate potential factors contributing to its variable expression. MATERIALS AND METHODS: A retrospective query was performed (03/31/21-05/31/22) for NET/NEC cases that had both formalin- and CytoLyt-fixed cell blocks. We collected clinical data and reporting of immunostains for each case. INSM1 staining was evaluated in both fixation methods, and reported as positive, negative, or equivocal. Equivocal INSM1 staining was further scored as a percentage of 1%-100% and intensity of weak (faint staining), moderate (darker staining), and strong (dense staining). RESULTS: Our search identified 20 cases from diverse body sites, including mediastinal lymph nodes (40%), pancreas (35%), lung (20%), and porta hepatis lymph nodes (5%). All cases exhibited a widespread positivity (over 90%) in formalin-fixed cell blocks. In contrast, CytoLyt fixed cells showed a negative stain in 65% of cases and 30% exhibited an equivocal positivity. CONCLUSIONS: While INSM1 is previously reported as a sensitive (75%-100%) and specific (82.7%-100%) marker for NET/NECs, our study found a reduced immunohistochemical staining in CytoLyt-fixed cell blocks. Consequently, false negative INSM1 immunohistochemical results in CytoLyt-fixed cell block material may pose a pitfall in the diagnosis of NET/NEC.


Biomarkers, Tumor , Formaldehyde , Immunohistochemistry , Repressor Proteins , Tissue Fixation , Humans , Retrospective Studies , Repressor Proteins/metabolism , Biomarkers, Tumor/metabolism , Immunohistochemistry/methods , Tissue Fixation/methods , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/diagnosis , Female , Middle Aged , Male , Aged , Adult , Fixatives , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/diagnosis , Carcinoma, Neuroendocrine/pathology , Carcinoma, Neuroendocrine/diagnosis , Carcinoma, Neuroendocrine/metabolism
20.
OMICS ; 28(4): 182-192, 2024 Apr.
Article En | MEDLINE | ID: mdl-38634790

Over a decade ago, longitudinal multiomics analysis was pioneered for early disease detection and individually tailored precision health interventions. However, high sample processing costs, expansive multiomics measurements along with complex data analysis have made this approach to precision/personalized medicine impractical. Here we describe in a case report, a more practical approach that uses fewer measurements, annual sampling, and faster decision making. We also show how this approach offers promise to detect an exceedingly rare and potentially fatal condition before it fully manifests. Specifically, we describe in the present case report how longitudinal multiomics monitoring (LMOM) helped detect a precancerous pancreatic tumor and led to a successful surgical intervention. The patient, enrolled in an annual blood-based LMOM since 2018, had dramatic changes in the June 2021 and 2022 annual metabolomics and proteomics results that prompted further clinical diagnostic testing for pancreatic cancer. Using abdominal magnetic resonance imaging, a 2.6 cm lesion in the tail of the patient's pancreas was detected. The tumor fluid from an aspiration biopsy had 10,000 times that of normal carcinoembryonic antigen levels. After the tumor was surgically resected, histopathological findings confirmed it was a precancerous pancreatic tumor. Postoperative omics testing indicated that most metabolite and protein levels returned to patient's 2018 levels. This case report illustrates the potentials of blood LMOM for precision/personalized medicine, and new ways of thinking medical innovation for a potentially life-saving early diagnosis of pancreatic cancer. Blood LMOM warrants future programmatic translational research with the goals of precision medicine, and individually tailored cancer diagnoses and treatments.


Pancreatic Neoplasms , Precancerous Conditions , Humans , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/surgery , Pancreatic Neoplasms/genetics , Precancerous Conditions/diagnosis , Precancerous Conditions/blood , Precancerous Conditions/pathology , Proteomics/methods , Biomarkers, Tumor/blood , Metabolomics/methods , Male , Precision Medicine/methods , Magnetic Resonance Imaging , Middle Aged , Early Detection of Cancer/methods , Multiomics
...