Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Gut ; 72(4): 624-637, 2023 04.
Article in English | MEDLINE | ID: mdl-36002248

ABSTRACT

OBJECTIVE: Many cancers engage embryonic genes for rapid growth and evading the immune system. SOX9 has been upregulated in many tumours, yet the role of SOX9 in mediating immunosuppressive tumour microenvironment is unclear. Here, we aim to dissect the role of SOX9-mediated cancer stemness attributes and immunosuppressive microenvironment in advanced gastric adenocarcinoma (GAC) for novel therapeutic discoveries. METHODS: Bulk RNAseq/scRNA-seq, patient-derived cells/models and extensive functional studies were used to identify the expression and functions of SOX9 and its target genes in vitro and in vivo. Immune responses were studied in PBMCs or CD45+ immune cells cocultured with tumour cells with SOX9high or knockout and the KP-Luc2 syngeneic models were used for efficacy of combinations. RESULTS: SOX9 is one of the most upregulated SOX genes in GAC and highly expressed in primary and metastatic tissues and associated with poor prognosis. Depletion of SOX9 in patient-derived GAC cells significantly decreased cancer stemness attributes, tumour formation and metastases and consistently increased CD8+ T cell responses when cocultured with PBMCs/CD45+ cells from GAC patients. RNA sequencing identified the leukaemia inhibitory factor (LIF) as the top secreted molecule regulated by SOX9 in tumour cells and was enriched in malignant ascites and mediated SOX9-induced M2 macrophage repolarisation and inhibited T cell function. CONCLUSION: Epithelial SOX9 is critical in suppressing CD8+ T cell responses and modified macrophage function in GAC through the paracrine LIF factor. Cotargeting LIF/LIFR and CSF1R has great potential in targeting SOX9-mediated cancer stemness, T cell immunosuppression and metastases suggesting the novel combination therapy against advanced GAC.


Subject(s)
Adenocarcinoma , Stomach Neoplasms , Humans , Tumor Microenvironment , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Immunosuppressive Agents , Immunosuppression Therapy , SOX9 Transcription Factor/genetics
2.
Mod Pathol ; 36(3): 100065, 2023 03.
Article in English | MEDLINE | ID: mdl-36788102

ABSTRACT

Distinguishing grade 3 pancreatic neuroendocrine tumor (G3 PanNET) from neuroendocrine carcinoma (PanNEC) is a known diagnostic challenge, and accurate classification is critical because clinical behavior and therapies differ. Although current recommendations suggest that immunohistochemistry for p53, Rb, ATRX, and DAXX can distinguish most cases, some cases remain difficult to classify using this approach. In this study, we reviewed 47 high-grade neoplasms originally diagnosed as pancreatic neuroendocrine neoplasms. In addition to the currently recommended stains, we performed capture-based sequencing of approximately 500 cancer genes and immunohistochemistry for p16 and trypsin or chymotrypsin. Using an integrated molecular and clinicopathologic approach, 42 (89%) of 47 cases had a clear final diagnosis of either G3 PanNET (n = 17), PanNEC (n = 17), or mixed acinar-NEC (n = 8). The 17 G3 PanNETs demonstrated frequent alterations in MEN1 (71%), DAXX (47%), ATRX (24%), TSC2 (35%), SETD2 (42%), and CDKN2A (41%). Contrary to prior reports, TP53 alterations were also common in G3 PanNETs (35%) but were always mutually exclusive with CDKN2A alterations in this group. The 17 PanNECs demonstrated frequent alterations in TP53 (88%), cell cycle genes RB1 (47%), CCNE1/CCND1 (12%), CDKN2A (29%), and in KRAS (53%) and SMAD4 (41%); TP53 was coaltered with a cell cycle gene in 76% of PanNECs. Diffuse strong p16 staining was observed in 69% of PanNECs in contrast to 0% of G3 PanNETs. The 8 acinar-NECs had recurrent alterations in ATM (25%), APC (25%), and STK11 (25%). Five cases remained difficult to classify, 3 of which exhibited overlapping molecular features with alterations in MEN1 with or without ATRX, and RB1 with or without TP53, making it unclear whether to classify as PanNET or PanNEC. Our data demonstrate that molecular profiling and immunohistochemistry for p16 greatly improve the diagnostic accuracy of high-grade pancreatic neuroendocrine neoplasms and identify a subset of rare cases with overlapping features of both PanNET and PanNEC.


Subject(s)
Carcinoma, Neuroendocrine , Neuroendocrine Tumors , Pancreatic Neoplasms , Humans , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Carcinoma, Neuroendocrine/diagnosis , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/pathology , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Genomics
3.
Br J Surg ; 110(1): 67-75, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36331867

ABSTRACT

BACKGROUND: Most tumour response scoring systems for resected pancreatic cancer after neoadjuvant therapy score tumour regression. However, whether treatment-induced changes, including tumour regression, can be identified reliably on haematoxylin and eosin-stained slides remains unclear. Moreover, no large study of the interobserver agreement of current tumour response scoring systems for pancreatic cancer exists. This study aimed to investigate whether gastrointestinal/pancreatic pathologists can reliably identify treatment effect on tumour by histology, and to determine the interobserver agreement for current tumour response scoring systems. METHODS: Overall, 23 gastrointestinal/pancreatic pathologists reviewed digital haematoxylin and eosin-stained slides of pancreatic cancer or treated tumour bed. The accuracy in identifying the treatment effect was investigated in 60 patients (30 treatment-naive, 30 after neoadjuvant therapy (NAT)). The interobserver agreement for the College of American Pathologists (CAP) and MD Anderson Cancer Center (MDACC) tumour response scoring systems was assessed in 50 patients using intraclass correlation coefficients (ICCs). An ICC value below 0.50 indicated poor reliability, 0.50 or more and less than 0.75 indicated moderate reliability, 0.75 or more and below 0.90 indicated good reliability, and above 0.90 indicated excellent reliability. RESULTS: The sensitivity and specificity for identifying NAT effect were 76.2 and 49.0 per cent respectively. After NAT in 50 patients, ICC values for both tumour response scoring systems were moderate: 0.66 for CAP and 0.71 for MDACC. CONCLUSION: Identification of the effect of NAT in resected pancreatic cancer proved unreliable, and interobserver agreement for the current tumour response scoring systems was suboptimal. These findings support the recently published International Study Group of Pancreatic Pathologists recommendations to score residual tumour burden rather than tumour regression after NAT.


Subject(s)
Neoadjuvant Therapy , Pancreatic Neoplasms , Humans , Eosine Yellowish-(YS) , Reproducibility of Results , Pancreatic Neoplasms/surgery , Pancreatic Neoplasms/pathology , Observer Variation , Pancreatic Neoplasms
4.
Radiographics ; 42(5): 1320-1337, 2022.
Article in English | MEDLINE | ID: mdl-35930475

ABSTRACT

Biliary malignancies include those arising from the intrahepatic and extrahepatic bile ducts as well as the gallbladder and hepatopancreatic ampulla of Vater. The majority of intrahepatic and extrahepatic malignancies are cholangiocarcinomas (CCAs). They arise owing to a complex interplay between the patient-specific genetic background and multiple risk factors and may occur in the liver (intrahepatic CCA), hilum (perihilar CCA), or extrahepatic bile ducts (distal CCA). Biliary-type adenocarcinoma constitutes the most common histologic type of ampullary and gallbladder malignancies. Its prognosis is poor and surgical resection is considered curative, so early detection is key, with multimodality imaging playing a central role in making the diagnosis. There are several risk factors for biliary malignancy as well as predisposing conditions that increase the risk; this review highlights the pertinent imaging features of these entities with histopathologic correlation. The predisposing factors are broken down into three major categories: (a) congenital malformations such as choledochal cyst and pancreaticobiliary maljunction; (b) infectious or inflammatory conditions such as parasitic infections, hepatolithiasis, primary sclerosing cholangitis, and porcelain gallbladder; and (c) preinvasive epithelial neoplasms such as biliary intraepithelial neoplasm, intraductal papillary neoplasm of the bile duct, intra-ampullary papillary tubular neoplasm, and intracholecystic papillary neoplasm of the gallbladder. Recognizing the baseline features of these premalignant biliary entities and changes in their appearance over time that indicate the advent of malignancy in high-risk patients can lead to early diagnosis and potentially curative management. An invited commentary by Volpacchio is available online. Online supplemental material is available for this article. ©RSNA, 2022.


Subject(s)
Bile Duct Neoplasms , Biliary Tract Neoplasms , Cholangiocarcinoma , Lithiasis , Liver Diseases , Pancreatic Neoplasms , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Biliary Tract Neoplasms/diagnostic imaging , Cholangiocarcinoma/pathology , Humans
5.
Gut ; 70(1): 148-156, 2021 01.
Article in English | MEDLINE | ID: mdl-32350089

ABSTRACT

OBJECTIVE: Recently, tumours with microsatellite instability (MSI)/defective DNA mismatch repair (dMMR) have gained considerable interest due to the success of immunotherapy in this molecular setting. Here, we aim to clarify clinical-pathological and/or molecular features of this tumour subgroup through a systematic review coupled with a comparative analysis with existing databases, also providing indications for a correct approach to the clinical identification of MSI/dMMR pancreatic ductal adenocarcinoma (PDAC). DESIGN: PubMed, SCOPUS and Embase were searched for studies reporting data on MSI/dMMR in PDAC up to 30 November 2019. Histological and molecular data of MSI/dMMR PDAC were compared with non-MSI/dMMR PDAC and with PDAC reference cohorts (including SEER database and The Cancer Genome Atlas Research Network - TCGA project). RESULTS: Overall, 34 studies with 8323 patients with PDAC were included in the systematic review. MSI/dMMR demonstrated a very low prevalence in PDAC (around 1%-2%). Compared with conventional PDAC, MSI/dMMR PDAC resulted strongly associated with medullary and mucinous/colloid histology (p<0.01) and with a KRAS/TP53 wild-type molecular background (p<0.01), with more common JAK genes mutations. Data on survival are still unclear. CONCLUSION: PDAC showing typical medullary or mucinous/colloid histology should be routinely examined for MSI/dMMR status using specific tests (immunohistochemistry, followed by MSI-PCR in cases with doubtful results). Next-generation sequencing (NGS) should be adopted either where there is limited tissue or as part of NGS tumour profiling in the context of precision oncology, acknowledging that conventional histology of PDAC may rarely harbour MSI/dMMR.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Microsatellite Instability , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/therapy , Databases, Factual , Humans , Pancreatic Neoplasms/therapy
6.
Oncologist ; 26(2): 90-96, 2021 02.
Article in English | MEDLINE | ID: mdl-32945065

ABSTRACT

Both metachronous and synchronous tumors pose a diagnostic and clinical challenge, more so when one of the specimens demonstrates the rare neuroendocrine histology. We describe a patient with sarcoidosis who was treated for endometrial and ovarian neoplasm, recurred with two separate histologies (adenocarcinoma and high grade neuroendocrine), both associated with microsatellite instability (MSI)-high status. Targeted next-generation sequencing of tumor with synonymous somatic alterations pointed to a common ancestry of all three tumors and patient was successfully treated with a tailored immunotherapy regimen. Her sarcoidosis worsened only slightly, and immunotherapy did not need to be discontinued. This case highlights the importance of molecular testing for the optimal therapy of complex synchronous tumors and the need for communication between surgical and medical oncologists in patients with MSI-high cancer. KEY POINTS: The case of a patient with a recurrent gynecological cancer presenting as microsatellite instability (MSI)-high endometrial adenocarcinoma and MSI-high neuroendocrine tumor is reported. This case demonstrated a common genetic lineage with good response to checkpoint inhibition without clinical worsening of autoimmune disease. This article adds to the literature, suggesting tumor evolution with neuroendocrine differentiation in some cancers, and argues that a molecular-based approach to treatment might achieve better understanding and possibly superior treatment outcomes.


Subject(s)
Carcinoma, Neuroendocrine , Endometrial Neoplasms , Ovarian Neoplasms , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/genetics , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Female , Humans , Microsatellite Instability , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics
7.
Radiology ; 299(2): 349-358, 2021 05.
Article in English | MEDLINE | ID: mdl-33754826

ABSTRACT

Background Conventional radiologic modalities perform poorly in the radiated rectum and are often unable to differentiate residual cancer from treatment scarring. Purpose To report the development and initial patient study of an imaging system comprising an endorectal coregistered photoacoustic (PA) microscopy (PAM) and US system paired with a convolution neural network (CNN) to assess the rectal cancer treatment response. Materials and Methods In this prospective study (ClinicalTrials.gov identifier NCT04339374), participants completed radiation and chemotherapy from September 2019 to September 2020 and images were obtained with the PAM/US system prior to surgery. Another group's colorectal specimens were studied ex vivo. The PAM/US system consisted of an endorectal imaging probe, a 1064-nm laser, and one US ring transducer. The PAM CNN and US CNN models were trained and validated to distinguish normal from malignant colorectal tissue using ex vivo and in vivo patient data. The PAM CNN and US CNN were then tested using additional in vivo patient data that had not been seen by the CNNs during training and validation. Results Twenty-two patients' ex vivo specimens and five patients' in vivo images (a total of 2693 US regions of interest [ROIs] and 2208 PA ROIs) were used for CNN training and validation. Data from five additional patients were used for testing. A total of 32 participants (mean age, 60 years; range, 35-89 years) were evaluated. Unique PAM imaging markers of the complete tumor response were found, specifically including recovery of normal submucosal vascular architecture within the treated tumor bed. The PAM CNN model captured this recovery process and correctly differentiated these changes from the residual tumor. The imaging system remained highly capable of differentiating tumor from normal tissue, achieving an area under the receiver operating characteristic curve of 0.98 (95% CI: 0.98, 0.99) for data from five participants. By comparison, the US CNN had an area under the receiver operating characteristic curve of 0.71 (95% CI: 0.70, 0.73). Conclusion An endorectal coregistered photoacoustic microscopy/US system paired with a convolutional neural network model showed high diagnostic performance in assessing the rectal cancer treatment response and demonstrated potential for optimizing posttreatment management. © RSNA, 2021 Supplemental material is available for this article. See also the editorial by Klibanov in this issue.


Subject(s)
Deep Learning , Neoplasm, Residual/diagnostic imaging , Photoacoustic Techniques , Rectal Neoplasms/diagnostic imaging , Ultrasonography/methods , Adult , Aged , Aged, 80 and over , Diagnosis, Differential , Female , Humans , Male , Middle Aged , Prospective Studies , Rectal Neoplasms/pathology , Rectal Neoplasms/therapy
8.
Mod Pathol ; 33(7): 1410-1419, 2020 07.
Article in English | MEDLINE | ID: mdl-32051556

ABSTRACT

Smooth muscle tumors represent the second most common mural mesenchymal neoplasm in the gastrointestinal tract, but established criteria for prognostic assessment of these tumors are lacking. A large cohort of surgically resected intramural gastrointestinal smooth muscle tumors from 31 institutions was analyzed to identify potential prognostic features. Pathologic features were assessed by expert gastrointestinal and/or soft tissue pathologists at each center. Immunohistochemical confirmation was required. A total of 407 cases from the esophagus (n = 97, 24%), stomach (n = 180, 44%), small bowel (n = 74, 18%), and colorectum (n = 56, 14%) were identified. Patients ranged in age from 19 to 92 years (mean 55 years), with a slight female predominance (57%). Mean tumor size was 5.4 cm, with the largest tumor measuring 29 cm. Disease progression following surgery, defined as local recurrence, metastasis, or disease-related death, occurred in 56 patients (14%). Colorectal tumors were most likely to progress, followed by small bowel and gastric tumors. None of the esophageal tumors in this series progressed. Receiver operator characteristic analysis identified optimal cutoffs of 9.8 cm and 3 mitoses/5 mm2 for discriminating between progressive and non-progressive tumors. Histologic features strongly associated with progression by univariate analysis included moderate-to-severe atypia, high cellularity, abnormal differentiation (defined as differentiation not closely resembling that of normal smooth muscle), tumor necrosis, mucosal ulceration, lamina propria involvement, and serosal involvement (P < 0.0001 for all features). Age, sex, and margin status were not significantly associated with progression (P = 0.23, 0.82, and 0.07, respectively). A risk assessment table was created based on tumor site, size, and mitotic count, and Kaplan-Meier plots of progression-free survival for each subgroup revealed progression-based tiers. Based on our findings, it appears that nonesophageal gastrointestinal smooth muscle tumors measuring >10 cm and/or showing ≥3 mitoses/5 mm2 may behave aggressively, and therefore close clinical follow-up is recommended in these cases.


Subject(s)
Gastrointestinal Neoplasms/pathology , Smooth Muscle Tumor/pathology , Adult , Aged , Aged, 80 and over , Disease Progression , Female , Humans , Male , Middle Aged , Prognosis , Progression-Free Survival
9.
Ann Surg Oncol ; 27(9): 3542-3550, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32206954

ABSTRACT

BACKGROUND: Accurate grading of neuroendocrine neoplasms (NENs) is crucial for proper assessment of prognosis. Estimation of the proliferative indices, if not performed properly, is largely erroneous due to significant intratumoral heterogeneity. We sought to establish the degree of error in the grading of a cohort of curatively resected pancreatic NENs (PanNENs) and the theoretical impact of that in a larger cohort of Surveillance, Epidemiology, and End Results (SEER) patients. METHODS: A retrospective query of an institutional surgical database was performed from 2000 to 2018 to identify optimally resected PanNENs, which were reviewed by two gastrointestinal pathologists and regraded according to the WHO 2017 classification. Overall survival and recurrence-free survival were estimated using the Kaplan-Meier method for original and new grading systems, respectively and Cox proportional hazards models were used to evaluate the effect of the interested variables, including new grading systems. RESULTS: A total of 176 cases were identified. After regrading, 17/64 (26.6%) G1 neoplasms were classified as G2 and 12/95 (12.6%) G2 neoplasms were classified as G1, while 1/11 (9.1%) G3 neoplasms were classified as G2. Our expert gastrointestinal pathologists agreed on 97% of reclassified cases by blind review. Application of the G1/G2 misclassification errors on various groups, including PanNENs, in a SEER database of 1385 patients rendered the reported survival differences nonsignificant (1000 repetitions; p = 0.063, 95% confidence interval 0.056-0.070). CONCLUSIONS: Mischaracterization of grade is common in optimally resected PanNENs but is eliminated with proper training and adherence to guidelines. The discrepancy rates can cast doubt on the generally accepted survival differences between G1 and G2 patients, as surmised by large database analyses.


Subject(s)
Diagnostic Errors , Neoplasm Grading , Neuroendocrine Tumors , Pancreatic Neoplasms , Humans , Neoplasm Grading/standards , Neuroendocrine Tumors/mortality , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/surgery , Pancreas/pathology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Prognosis , Retrospective Studies , SEER Program , Survival Analysis
10.
J Magn Reson Imaging ; 51(6): 1630-1640, 2020 06.
Article in English | MEDLINE | ID: mdl-31418986

ABSTRACT

Hepatocellular adenomas (HCAs) are benign hepatic tumors that can be complicated by bleeding and/or malignant transformation. The epidemiology of HCAs has changed over recent decades, primarily influenced by an increased incidence of obesity in both men and women. Currently, at least eight distinct pathomolecular subtypes of HCAs have been identified, several of which have distinguishing and pertinent imaging features on MRI. Emerging evidence suggests that hepatobiliary phase appearance may provide diagnostic and prognostic information. The purpose of this article is to review the current pathomolecular lexicon and imaging features with emphasis on hepatobiliary phase appearance. Level of Evidence: 5 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2020;51:1630-1640.


Subject(s)
Adenoma, Liver Cell , Carcinoma, Hepatocellular , Liver Neoplasms , Adenoma, Liver Cell/diagnostic imaging , Female , Hemorrhage , Humans , Liver Neoplasms/diagnostic imaging , Magnetic Resonance Imaging , Male
11.
Ann Diagn Pathol ; 45: 151452, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31945621

ABSTRACT

Neoadjuvant treatment (NAT) followed by total mesorectal excision is currently considered the standard of treatment for rectal adenocarcinoma. The degree of pathologic treatment response (pTR) correlates significantly with the recurrence free survival and overall survival (OS). However, it remains unclear which clinical and pathologic factors are associated with a more robust response to NAT, including showing pathologic complete response (pCR). Chemokine receptor 4 (CXCR4) overexpression has been associated with unfavorable OS in some studies. In this study, we sought to evaluate the clinicopathologic determinants of pTR in neoadjuvant treated rectal adenocarcinoma (NAT-RA). We retrospectively identified 91 patients who underwent pre-treatment diagnostic biopsy, NAT, and surgical resection at our institution. The archival slides were reviewed for pathologic features in the pre-treatment biopsies and for assessment of pTR in the resection specimens according to the current College of American Pathologist (CAP)'s guidelines. pCR was obtained in 16.5% of the cases, whereas 20.9% had near pCR, 30.8% had partial response, and 31.9% had a poor/no response. CXCR4 immunohistochemical analysis was also performed on the pre-treatment biopsies. Lower pre-treatment cT-stage (p = 0.019) and pre-treatment AJCC cTNM stage groups (p = 0.004), longer time interval between completion of NAT and resection (p = 0.022), and presence of tumor-infiltrating lymphocytes in the pre-treatment biopsies (p = 0.019) were significantly associated with a better pTR. CXCR4 nuclear expression was associated with a lower percentage of residual tumor (p = 0.036). Pre-treatment CEA levels, tumor differentiation, CAP treatment response groups and lower percentage of residual tumor were associated with a better OS.


Subject(s)
Adenocarcinoma/drug therapy , Neoadjuvant Therapy/methods , Rectal Neoplasms/pathology , Adenocarcinoma/surgery , Aged , Chemoradiotherapy/methods , Disease-Free Survival , Female , Humans , Immunohistochemistry/methods , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Neoplasm Staging/methods , Neoplasm, Residual/pathology , Receptors, CXCR4/metabolism , Retrospective Studies
12.
Am J Physiol Endocrinol Metab ; 316(4): E674-E686, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30860882

ABSTRACT

Studies show maternal obesity is a risk factor for metabolic syndrome and nonalcoholic fatty liver disease (NAFLD) in offspring. Here we evaluated potential mechanisms underlying these phenotypes. Female C57Bl6 mice were fed chow or an obesogenic high-fat/high-sucrose (HF/HS) diet with subsequent mating of F1 and F2 female offspring to lean males to develop F2 and F3 generations, respectively. Offspring were fed chow or fibrogenic (high transfat, cholesterol, fructose) diets, and histopathological, metabolic changes, and bile acid (BA) homeostasis was evaluated. Chow-fed F1 offspring from maternal HF/HS lineages (HF/HS) developed periportal fibrosis and inflammation with aging, without differences in hepatic steatosis but increased BA pool size and shifts in BA composition. F1, but not F2 or F3, offspring from HF/HS showed increased steatosis on a fibrogenic diet, yet inflammation and fibrosis were paradoxically decreased in F1 offspring, a trend continued in F2 and F3 offspring. HF/HS feeding leads to increased periportal fibrosis and inflammation in chow-fed offspring without increased hepatic steatosis. By contrast, fibrogenic diet-fed F1 offspring from HF/HS dams exhibited worse hepatic steatosis but decreased inflammation and fibrosis. These findings highlight complex adaptations in NAFLD phenotypes with maternal diet.


Subject(s)
Bile Acids and Salts/metabolism , Cholesterol/metabolism , Diet , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Prenatal Exposure Delayed Effects/metabolism , Triglycerides/metabolism , Animals , Diet, High-Fat , Dietary Fats , Dietary Sucrose , Female , Fibrosis , Fructose , Homeostasis , Inflammation , Liver/pathology , Male , Metabolic Syndrome , Mice , Mice, Inbred C57BL , Obesity , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Trans Fatty Acids
13.
Histopathology ; 75(4): 568-577, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31077609

ABSTRACT

AIMS: Insulinoma-associated protein 1 (INSM1) is a transcription factor that is expressed in developing and mature neuroendocrine tissue. Recent studies have shown that INSM1 is a sensitive marker for neuroendocrine tumours. The aims of this study were to evaluate INSM1 expression in primary gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) and in their known metastases, in order to assess its sensitivity as compared with chromogranin-A (CgA) and synaptophysin (SYN), and to evaluate any change in expression between primary and metastatic disease. METHODS AND RESULTS: We identified 30 patients with primary GEP-NEN. Liver metastatic tissue was available for 26 patients; two patients had two metachronous metastatic foci, yielding a total of 28 metastatic cases. An additional two and seven non-paired cases of primary and metastatic grade 3 GEP-NEN, respectively, were included. To assess specificity, we evaluated the expression of these markers in other primary tumours (colorectal adenocarcinoma, acinar cell carcinoma, solid pseudopapillary neoplasm, cholangiocarcinoma, and hepatocellular carcinoma) and metastatic tumours in the liver (adrenal cortical, breast and prostate carcinomas) that may present as differential diagnoses. In our cohort, all of the primary GEP-NENs and 94% of the metastatic GEP-NENs expressed INSM1. INSM1 showed similar sensitivity to SYN and higher sensitivity than CgA in both primary and metastatic neoplasms. INSM1 has comparable specificity to CgA, and higher specificity than SYN. CONCLUSIONS: The nuclear reactivity and the high sensitivity and specificity of INSM1 make it a preferred neuroendocrine marker. In conclusion, INSM1 can be used as a single first-line marker for primary and metastatic GEP-NEN.


Subject(s)
Biomarkers, Tumor/analysis , Intestinal Neoplasms/diagnosis , Neuroendocrine Tumors/diagnosis , Pancreatic Neoplasms/diagnosis , Repressor Proteins/biosynthesis , Stomach Neoplasms/diagnosis , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Neoplasm Metastasis/diagnosis , Repressor Proteins/analysis , Retrospective Studies , Sensitivity and Specificity
14.
Lab Invest ; 95(2): 207-22, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25485535

ABSTRACT

Many commercially available cell lines have been in culture for ages, acquiring phenotypes that differ from the original cancers from which these cell lines were derived. Therefore, research on new cell lines could improve the success rates of translational research in cancer. We have developed methods for the isolation and culture of human pancreatic ductal adenocarcinoma (PDAC) cells from murine xenografts of human PDAC. We hypothesize that phenotypes of PDAC cells are modified by in vitro culture conditions over time and by in vivo implantation. Patient-derived xenografts were created in immunodeficient mice using surgically resected tumor specimens. These murine xenografts were then used to establish human PDAC cell lines in culture. Earlier (<5) passage and later (>20) passage cell lines were evaluated separately regarding proliferation, cell cycle, genetic mutations, invasiveness, chemosensitivity, tumorigenesis, epithelial-mesenchymal transition (EMT) status, and proteomics. Later passage cells accelerated their doubling time and colony formation, and were more concentrated in the G0/G1 phase and less in the G2/M checkpoint phase. Later passage cells were more sensitive to gemcitabine and 5-fluorouracil than earlier passage cells, but all four new cell lines were more chemo-resistant compared with commercial ATCC cell lines. EMT induction was observed when establishing and passaging cell lines in vitro and furthermore by growing them as subcutaneous tumors in vivo. This study demonstrates a novel approach to the establishment of PDAC cell lines and observes a process by which newly established cell lines undergo phenotypic changes during in vitro culture and in vivo tumorigenesis. This may help explain differences of treatment effects often observed between experiments conducted in vitro, in vivo, and in human clinical trials.


Subject(s)
Cell Culture Techniques/methods , Epithelial-Mesenchymal Transition/physiology , Pancreatic Neoplasms/physiopathology , Phenotype , Animals , Blotting, Western , Cell Cycle/physiology , Cell Proliferation/physiology , Colony-Forming Units Assay , Deoxycytidine/analogs & derivatives , Fluorouracil , Heterografts/cytology , Heterografts/physiology , Humans , Immunohistochemistry , Mice , Neoplasm Invasiveness/physiopathology , Protein Array Analysis , Proteomics/methods , Tumor Cells, Cultured , Gemcitabine , Pancreatic Neoplasms
16.
Ann Surg Oncol ; 22(6): 1884-92, 2015.
Article in English | MEDLINE | ID: mdl-25404477

ABSTRACT

BACKGROUND: Recurrence after resection of pancreatic ductal adenocarcinoma (PDAC) is common, thus postoperative surveillance is critical for detection and treatment of recurrent disease. The development of biologically based techniques for early recurrence detection may enable more timely and effective treatment of such recurrences. METHODS: Tumor fragments derived from patients who underwent potentially curative resection of PDAC were heterotopically implanted into NOD/SCID mice. Engraftment success rates and growth parameters were matched to clinicopathologic data, preoperative treatment status, and oncologic outcomes to correlate disease-free survival (DFS) and overall survival. RESULTS: Seventy patients consented to participate with 56 (80 %) developing a mouse PDAC tumorgraft. Patients with successful engraftment had a shorter median DFS compared with patients whose tumorgrafts failed to engraft (9.8 vs. 40.9 mo, respectively; p < 0.01). Fifty patients received preoperative therapy with 36 (72 %) successful tumorgrafts from this cohort. On multivariate analysis, lymph node metastasis (hazard ratio [HR] 3, 95 % CI 1.4-6.7, p < 0.01) and successful engraftment (HR 5.8, 95 % CI 2-16.9, p < 0.01) were predictive of a shorter DFS in the preoperative therapy cohort. In patients who recurred, tumorgraft formation was identified at a median of 134.5 days before standard methods of radiographic recurrence detection (p < 0.01). CONCLUSIONS: Patient-derived tumorgrafts from resected PDAC may potentially predict recurrence months before currently available surveillance modalities. This lead-time advantage may allow for earlier implementation or changes in therapy as successful engraftment, particularly in those having undergone preoperative therapy, may indicate a more biologically aggressive disease.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Neoplasm Recurrence, Local/pathology , Pancreatectomy/mortality , Pancreatic Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Animals , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/surgery , Female , Follow-Up Studies , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Invasiveness , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/surgery , Neoplasm Staging , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/surgery , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Ann Surg Oncol ; 22(4): 1168-75, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25352267

ABSTRACT

BACKGROUND: The use of neoadjuvant therapy (NAC) for the treatment of potentially resectable pancreatic cancer remains controversial. In this study, we sought to evaluate cancer-specific endpoints in patients undergoing a NAC versus a surgery-first (SF) approach with specific emphasis on lymph node metastases. METHODS: A total of 222 patients who underwent NAC and 85 patients who underwent SF were identified from 1990 to 2008 and compared for cancer-related endpoints. Peripancreatic lymph nodes from 135 neoadjuvant therapy patients were evaluated for histologic tumor regression. RESULTS: Patients who underwent NAC followed by surgery had improved overall survival and time to local recurrence compared with the SF approach. NAC patients were less likely to have lymph node metastases (p = 0.001), lymphovascular invasion (LVI), and had smaller tumors. On multivariate analysis, lymph node positivity was associated with SF, tumor size, and the presence of LVI. NAC patients with N0 disease had equivalent outcomes to patients with a low-LNR (0.01-0.15), whereas patients with a LNR >0.15 had reduced survival, and time to local and distant recurrence. Ten of 135 (7.4 %) NAC patients had evidence of tumor regression in at least one lymph node. CONCLUSIONS: Patients with potentially resectable PDAC selected to undergo NAC had improved survival and longer time to recurrence. Although some of these differences may be related to improvements in multimodality therapy completion rates, tumor regression in lymph node metastases exists and may demonstrate a biologic benefit of NAC compared with a SF approach.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/therapy , Lymph Nodes/pathology , Neoadjuvant Therapy , Neoplasm Recurrence, Local/therapy , Pancreatic Neoplasms/therapy , Adenocarcinoma/mortality , Adenocarcinoma/secondary , Adult , Aged , Aged, 80 and over , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/secondary , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Pancreatectomy , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis , Survival Rate
19.
Gastroenterology ; 145(6): 1449-58, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23958541

ABSTRACT

BACKGROUND & AIMS: Obesity is a risk factor for pancreatic ductal adenocarcinoma (PDAC), but it is not clear how obesity contributes to pancreatic carcinogenesis. The oncogenic form of KRAS is expressed during early stages of PDAC development and is detected in almost all of these tumors. However, there is evidence that mutant KRAS requires an additional stimulus to activate its full oncogenic activity and that this stimulus involves the inflammatory response. We investigated whether the inflammation induced by a high-fat diet, and the accompanying up-regulation of cyclooxygenase-2 (COX2), increases Kras activity during pancreatic carcinogenesis in mice. METHODS: We studied mice with acinar cell-specific expression of KrasG12D (LSL-Kras/Ela-CreERT mice) alone or crossed with COX2 conditional knockout mice (COXKO/LSL-Kras/Ela-CreERT). We also studied LSL-Kras/PDX1-Cre mice. All mice were fed isocaloric diets with different amounts of fat, and a COX2 inhibitor was administered to some LSL-Kras/Ela-CreERT mice. Pancreata were collected from mice and analyzed for Kras activity, levels of phosphorylated extracellular-regulated kinase, inflammation, fibrosis, pancreatic intraepithelial neoplasia (PanIN), and PDACs. RESULTS: Pancreatic tissues from LSL-Kras/Ela-CreERT mice fed high-fat diets (HFDs) had increased Kras activity, fibrotic stroma, and numbers of PanINs and PDACs than LSL-Kras/Ela-CreERT mice fed control diets; the mice fed the HFDs also had shorter survival times than mice fed control diets. Administration of a COX2 inhibitor to LSL-Kras/Ela-CreERT mice prevented these effects of HFDs. We also observed a significant reduction in survival times of mice fed HFDs. COXKO/LSL-Kras/Ela-CreERT mice fed HFDs had no evidence for increased numbers of PanIN lesions, inflammation, or fibrosis, as opposed to the increases observed in LSL-Kras/Ela-CreERT mice fed HFDs. CONCLUSIONS: In mice, an HFD can activate oncogenic Kras via COX2, leading to pancreatic inflammation and fibrosis and development of PanINs and PDAC. This mechanism might be involved in the association between risk for PDAC and HFDs.


Subject(s)
Adenocarcinoma/physiopathology , Carcinoma, Pancreatic Ductal/physiopathology , Cyclooxygenase 2/physiology , Diet, High-Fat/adverse effects , Pancreatic Neoplasms/physiopathology , Proto-Oncogene Proteins p21(ras)/physiology , Adenocarcinoma/pathology , Adiposity/physiology , Animals , Carcinoma, Pancreatic Ductal/pathology , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , Disease Models, Animal , Fibrosis , Gene Expression Regulation, Neoplastic/physiology , Mice , Mice, Knockout , Mice, Transgenic , Pancreas/pathology , Pancreatic Neoplasms/pathology
20.
Phys Biol ; 11(6): 065002, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25427073

ABSTRACT

There is substantial heterogeneity in the clinical behavior of pancreatic cancer and in its response to therapy. Some of this variation may be due to differences in delivery of cytotoxic therapies between patients and within individual tumors. Indeed, in 12 patients with resectable pancreatic cancer, we previously demonstrated wide inter-patient variability in the delivery of gemcitabine as well as in the mass transport properties of tumors as measured by computed tomography (CT) scans. However, the variability of drug delivery and transport properties within pancreatic tumors is currently unknown. Here, we analyzed regional measurements of gemcitabine DNA incorporation in the tumors of the same 12 patients to understand the degree of intra-tumoral heterogeneity of drug delivery. We also developed a volumetric segmentation approach to measure mass transport properties from the CT scans of these patients and tested inter-observer agreement with this new methodology. Our results demonstrate significant heterogeneity of gemcitabine delivery within individual pancreatic tumors and across the patient cohort, with gemcitabine DNA incorporation in the inner portion of the tumors ranging from 38 to 74% of the total. Similarly, the CT-derived mass transport properties of the tumors had a high degree of heterogeneity, ranging from minimal difference to almost 200% difference between inner and outer portions of the tumor. Our quantitative method to derive transport properties from CT scans demonstrated less than 5% difference in gemcitabine prediction at the average CT-derived transport value across observers. These data illustrate significant inter-patient and intra-tumoral heterogeneity in the delivery of gemcitabine, and highlight how this variability can be reproducibly accounted for using principles of mass transport. With further validation as a biophysical marker, transport properties of tumors may be useful in patient selection for therapy and prediction of therapeutic outcome.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Deoxycytidine/analogs & derivatives , Drug Delivery Systems , Pancreatic Neoplasms/metabolism , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/metabolism , Biological Transport , DNA Adducts/metabolism , DNA, Neoplasm/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/metabolism , Deoxycytidine/pharmacokinetics , Humans , Injections, Intravenous , Pancreatectomy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Tissue Distribution , Tomography Scanners, X-Ray Computed , Tumor Microenvironment , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL