Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 629(8012): 679-687, 2024 May.
Article in English | MEDLINE | ID: mdl-38693266

ABSTRACT

Pancreatic intraepithelial neoplasias (PanINs) are the most common precursors of pancreatic cancer, but their small size and inaccessibility in humans make them challenging to study1. Critically, the number, dimensions and connectivity of human PanINs remain largely unknown, precluding important insights into early cancer development. Here, we provide a microanatomical survey of human PanINs by analysing 46 large samples of grossly normal human pancreas with a machine-learning pipeline for quantitative 3D histological reconstruction at single-cell resolution. To elucidate genetic relationships between and within PanINs, we developed a workflow in which 3D modelling guides multi-region microdissection and targeted and whole-exome sequencing. From these samples, we calculated a mean burden of 13 PanINs per cm3 and extrapolated that the normal intact adult pancreas harbours hundreds of PanINs, almost all with oncogenic KRAS hotspot mutations. We found that most PanINs originate as independent clones with distinct somatic mutation profiles. Some spatially continuous PanINs were found to contain multiple KRAS mutations; computational and in situ analyses demonstrated that different KRAS mutations localize to distinct cell subpopulations within these neoplasms, indicating their polyclonal origins. The extensive multifocality and genetic heterogeneity of PanINs raises important questions about mechanisms that drive precancer initiation and confer differential progression risk in the human pancreas. This detailed 3D genomic mapping of molecular alterations in human PanINs provides an empirical foundation for early detection and rational interception of pancreatic cancer.


Subject(s)
Genetic Heterogeneity , Genomics , Imaging, Three-Dimensional , Pancreatic Neoplasms , Precancerous Conditions , Single-Cell Analysis , Adult , Female , Humans , Male , Clone Cells/metabolism , Clone Cells/pathology , Exome Sequencing , Machine Learning , Mutation , Pancreas/anatomy & histology , Pancreas/cytology , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Workflow , Disease Progression , Early Detection of Cancer , Oncogenes/genetics
2.
Nat Methods ; 19(11): 1490-1499, 2022 11.
Article in English | MEDLINE | ID: mdl-36280719

ABSTRACT

A central challenge in biology is obtaining high-content, high-resolution information while analyzing tissue samples at volumes relevant to disease progression. We address this here with CODA, a method to reconstruct exceptionally large (up to multicentimeter cubed) tissues at subcellular resolution using serially sectioned hematoxylin and eosin-stained tissue sections. Here we demonstrate CODA's ability to reconstruct three-dimensional (3D) distinct microanatomical structures in pancreas, skin, lung and liver tissues. CODA allows creation of readily quantifiable tissue volumes amenable to biological research. As a testbed, we assess the microanatomy of the human pancreas during tumorigenesis within the branching pancreatic ductal system, labeling ten distinct structures to examine heterogeneity and structural transformation during neoplastic progression. We show that pancreatic precancerous lesions develop into distinct 3D morphological phenotypes and that pancreatic cancer tends to spread far from the bulk tumor along collagen fibers that are highly aligned to the 3D curves of ductal, lobular, vascular and neural structures. Thus, CODA establishes a means to transform broadly the structural study of human diseases through exploration of exhaustively labeled 3D microarchitecture.


Subject(s)
Imaging, Three-Dimensional , Pancreatic Neoplasms , Humans , Imaging, Three-Dimensional/methods , Pancreatic Neoplasms/pathology , Pancreas/pathology
3.
Stroke ; 55(1): 5-13, 2024 01.
Article in English | MEDLINE | ID: mdl-38134254

ABSTRACT

BACKGROUND: Physical inactivity in people with chronic stroke profoundly affects daily function and increases recurrent stroke risk and mortality, making physical activity improvements an important target of intervention. We compared the effects of a high-intensity walking intervention (FAST), a step activity monitoring behavioral intervention (SAM), or a combined intervention (FAST+SAM) on physical activity (ie, steps/day). We hypothesized the combined intervention would yield the greatest increase in steps/day. METHODS: This assessor-blinded multisite randomized controlled trial was conducted at 4 university/hospital-based laboratories. Participants were 21 to 85 years old, walking without physical assistance following a single, unilateral noncerebellar stroke of ≥6 months duration, and randomly assigned to FAST, SAM, or FAST+SAM for 12 weeks (2-3 sessions/week). FAST training consisted of walking-related activities at 70% to 80% heart rate reserve, while SAM received daily feedback and goal setting of walking activity (steps/day). Assessors and study statistician were masked to group assignment. The a priori-determined primary outcome and end point was a comparison of the change in steps/day between the 3 intervention groups from pre- to post-intervention. Adverse events were tracked after randomization. All randomized participants were included in the intent-to-treat analysis. RESULTS: Participants were enrolled from July 18, 2016, to November 16, 2021. Of 2385 participants initially screened, 250 participants were randomized (mean [SE] age, 63 [0.80] years; 116 females/134 males), with 89 assigned to FAST, 81 to SAM, and 80 to FAST+SAM. Steps/day significantly increased in both the SAM (mean [SE], 1542 [267; 95% CI, 1014-2069] P<0.001) and FAST+SAM group (1307 [280; 95% CI, 752-1861] P<0.001) but not in the FAST group (406 [238; 95% CI, -63 to 876] P=0.09). There were no deaths or serious study-related adverse events. CONCLUSIONS: Only individuals with chronic stroke who completed a step activity monitoring behavioral intervention with skilled coaching and goal progression demonstrated improvements in physical activity (steps/day). REGISTRATION: URL: https://www.clinicaltrials.gov; Unique identifier: NCT02835313.


Subject(s)
Stroke Rehabilitation , Stroke , Male , Female , Humans , Middle Aged , Young Adult , Adult , Aged , Aged, 80 and over , Walking/physiology , Exercise , Stroke/therapy , Exercise Therapy
4.
Cancer Metastasis Rev ; 40(3): 661-674, 2021 09.
Article in English | MEDLINE | ID: mdl-33555482

ABSTRACT

One way to understand ductal adenocarcinoma of the pancreas (pancreatic cancer) is to view it as unimaginably large numbers of evolving living organisms interacting with their environment. This "evolutionary view" creates both expected and surprising perspectives in all stages of neoplastic progression. Advances in the field will require greater attention to this critical evolutionary prospective.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/genetics , Humans , Pancreas , Pancreatic Neoplasms/genetics , Prospective Studies
5.
Ann Surg ; 275(6): 1165-1174, 2022 06 01.
Article in English | MEDLINE | ID: mdl-33214420

ABSTRACT

OBJECTIVE: This study aimed to identify risk factors for recurrence after pancreatic resection for intraductal papillary mucinous neoplasm (IPMN). SUMMARY BACKGROUND DATA: Long-term follow-up data on recurrence after surgical resection for IPMN are currently lacking. Previous studies have presented mixed results on the role of margin status in risk of recurrence after surgical resection. METHODS: A total of 126 patients that underwent resection for noninvasive IPMN were followed for a median of 9.5 years. Dedicated pathological and radiological reviews were performed to correlate clinical and pathological features (including detailed pathological features of the parenchymal margin) with recurrence after surgical resection. In addition, in a subset of 32 patients with positive margins, we determined the relationship between the margin and original IPMN using driver gene mutations identified by next-generation sequencing. RESULTS: Family history of pancreatic cancer and high-grade IPMN was identified as risk factors for recurrence in both uni- and multivariate analysis (adjusted hazard ratio 3.05 and 1.88, respectively). Although positive margin was not significantly associated with recurrence in our cohort, the size and grade of the dysplastic focus at the margin were significantly correlated with recurrence in margin-positive patients. Genetic analyses showed that the neoplastic epithelium at the margin was independent from the original IPMN in at least 9 of 32 cases (28%). The majority of recurrences (74%) occurred after 3 years, and a significant minority (32%) occurred after 5 years. CONCLUSION: Sustained postoperative surveillance for all patients is indicated, particularly those with risk factors such has family history and high-grade dysplasia.


Subject(s)
Adenocarcinoma, Mucinous , Carcinoma, Pancreatic Ductal , Carcinoma, Papillary , Pancreatic Intraductal Neoplasms , Pancreatic Neoplasms , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/pathology , Adenocarcinoma, Mucinous/surgery , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/surgery , Carcinoma, Papillary/pathology , Carcinoma, Papillary/surgery , Follow-Up Studies , Humans , Margins of Excision , Neoplasm Recurrence, Local/pathology , Pancreatectomy/methods , Pancreatic Intraductal Neoplasms/genetics , Pancreatic Intraductal Neoplasms/surgery , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Retrospective Studies
6.
J Neurol Phys Ther ; 46(4): 293-301, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35980730

ABSTRACT

BACKGROUND AND PURPOSE: Upper extremity studies suggest that implicit adaptation is less impaired than explicit learning in persons with Parkinson disease (PD). Little work has explored implicit locomotor adaptation and savings in this population, yet implicit locomotor learning is critical for everyday function. This cross-sectional study examined adaptation and savings in individuals with PD during split-belt treadmill walking. METHODS: Fourteen participants completed the following treadmill protocol: Baseline (6 minutes belts tied), Adaptation (10 minutes split), Washout (10 minutes tied), and Readaptation (10 minutes split). Step length and step symmetry index (SSI) were calculated to determine magnitude and rate of adaptation and savings. Rate was calculated as strides to reach SSI plateau during Adaptation and Readaptation. RESULTS: During Early Adaptation and Early Readaptation, SSI was perturbed from Baseline ( P < 0.001 and P = 0.002, respectively). Less perturbation in Early Readaptation ( P < 0.001) demonstrated savings. In Late Adaptation and Late Readaptation, participants returned to Baseline symmetry ( P = 0.026 and P = 0.022, respectively, with adjusted level of significance = 0.007). Adaptation was also seen in reverse asymmetry observed in Early Washout ( P = 0.003 vs Baseline). Readaptation rate was faster than in Adaptation ( P = 0.015), demonstrating savings. DISCUSSION AND CONCLUSIONS: Individuals with PD showed locomotor adaptation in an implicit sensorimotor adaptation task. They also demonstrated savings, with less perturbation and faster adaptation during the second split-belt exposure. However, performance was variable; some individuals showed minimal adaptation. Variations in learning, savings, and clinical presentation highlight the need to further explore characteristics of individuals with PD most likely to benefit from adaptation-based locomotor training.Video Abstract available for more insights from the authors (see the Video, Supplemental Digital Content 1, available at: http://links.lww.com/JNPT/A395 ).


Subject(s)
Parkinson Disease , Adaptation, Physiological , Cross-Sectional Studies , Exercise Test , Gait , Humans , Walking
7.
Gut ; 70(5): 928-939, 2021 05.
Article in English | MEDLINE | ID: mdl-33028669

ABSTRACT

OBJECTIVE: Intraductal papillary mucinous neoplasms (IPMNs) are non-invasive precursor lesions that can progress to invasive pancreatic cancer and are classified as low-grade or high-grade based on the morphology of the neoplastic epithelium. We aimed to compare genetic alterations in low-grade and high-grade regions of the same IPMN in order to identify molecular alterations underlying neoplastic progression. DESIGN: We performed multiregion whole exome sequencing on tissue samples from 17 IPMNs with both low-grade and high-grade dysplasia (76 IPMN regions, including 49 from low-grade dysplasia and 27 from high-grade dysplasia). We reconstructed the phylogeny for each case, and we assessed mutations in a novel driver gene in an independent cohort of 63 IPMN cyst fluid samples. RESULTS: Our multiregion whole exome sequencing identified KLF4, a previously unreported genetic driver of IPMN tumorigenesis, with hotspot mutations in one of two codons identified in >50% of the analyzed IPMNs. Mutations in KLF4 were significantly more prevalent in low-grade regions in our sequenced cases. Phylogenetic analyses of whole exome sequencing data demonstrated diverse patterns of IPMN initiation and progression. Hotspot mutations in KLF4 were also identified in an independent cohort of IPMN cyst fluid samples, again with a significantly higher prevalence in low-grade IPMNs. CONCLUSION: Hotspot mutations in KLF4 occur at high prevalence in IPMNs. Unique among pancreatic driver genes, KLF4 mutations are enriched in low-grade IPMNs. These data highlight distinct molecular features of low-grade and high-grade dysplasia and suggest diverse pathways to high-grade dysplasia via the IPMN pathway.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Carcinoma, Papillary/genetics , Exome Sequencing , Pancreatic Intraductal Neoplasms/genetics , Adenocarcinoma, Mucinous/pathology , Biomarkers, Tumor/genetics , Carcinoma, Papillary/pathology , Humans , Kruppel-Like Factor 4/genetics , Mutation , Neoplasm Grading , Pancreatic Intraductal Neoplasms/pathology , Retrospective Studies
8.
J Pathol ; 252(3): 252-262, 2020 11.
Article in English | MEDLINE | ID: mdl-32696980

ABSTRACT

Intraductal papillary mucinous neoplasms (IPMNs) are commonly identified non-invasive cyst-forming pancreatic neoplasms with the potential to progress into invasive pancreatic adenocarcinoma. There are few in vitro models with which to study the biology of IPMNs and their progression to invasive carcinoma. Therefore, we generated a living biobank of organoids from seven normal pancreatic ducts and ten IPMNs. We characterized eight IPMN organoid samples using whole genome sequencing and characterized five IPMN organoids and seven normal pancreatic duct organoids using transcriptome sequencing. We identified an average of 11,344 somatic mutations in the genomes of organoids derived from IPMNs, with one sample harboring 61,537 somatic mutations enriched for T→C transitions and T→A transversions. Recurrent coding somatic mutations were identified in 15 genes, including KRAS, GNAS, RNF43, PHF3, and RBM10. The most frequently mutated genes were KRAS, GNAS, and RNF43, with somatic mutations identified in six (75%), four (50%), and three (37.5%) IPMN organoid samples, respectively. On average, we identified 36 structural variants in IPMN derived organoids, and none had an unstable phenotype (> 200 structural variants). Transcriptome sequencing identified 28 genes differentially expressed between normal pancreatic duct organoid and IPMN organoid samples. The most significantly upregulated and downregulated genes were CLDN18 and FOXA1. Immunohistochemical analysis of FOXA1 expression in 112 IPMNs, 113 mucinous cystic neoplasms, and 145 pancreatic ductal adenocarcinomas demonstrated statistically significant loss of expression in low-grade IPMNs (p < 0.0016), mucinous cystic neoplasms (p < 0.0001), and pancreatic ductal adenocarcinoma of any histologic grade (p < 0.0001) compared to normal pancreatic ducts. These data indicate that FOXA1 loss of expression occurs early in pancreatic tumorigenesis. Our study highlights the utility of organoid culture to study the genetics and biology of normal pancreatic duct and IPMNs. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Mutation , Organoids , Pancreatic Intraductal Neoplasms/genetics , Biomarkers, Tumor/metabolism , Carcinogenesis/genetics , Case-Control Studies , Humans , Immunohistochemistry , Organoids/metabolism , Organoids/pathology , Pancreatic Intraductal Neoplasms/metabolism , Pancreatic Intraductal Neoplasms/pathology , Exome Sequencing , Whole Genome Sequencing
9.
World J Surg ; 45(10): 3157-3164, 2021 10.
Article in English | MEDLINE | ID: mdl-34236477

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) has a high propensity for systemic dissemination. Ovarian metastases are rare and poorly described. METHODS: We identified PDAC cases with ovarian metastasis from a prospectively maintained registry. We reported on the association between outcomes and clinicopathologic factors. Recurrence-free (RFS) and overall survival (OS) were calculated using Kaplan-Meier analysis. RESULTS: Twelve patients with PDAC and synchronous or metachronous ovarian metastases were identified. Nine patients (75%) underwent pancreatectomy for localized PDAC and developed metachronous ovarian recurrence. The median OS for all patients was 25.4 (IQR:15.4-82.9) months. For the nine patients with metachronous ovarian metastasis, the median RFS and OS were 14.2 (IQR:7.2-58.3) and 44.6 (IQR:18.6-82.9) months, respectively. Nodal disease, poor grade, vascular invasion in the pancreatic primary, and bilateral ovarian disease tended to confer worse outcomes. CONCLUSION: Patients with resected PDAC and ovarian recurrence tend to have a comparable disease course to more common patterns of recurrence. Primaries with nodal disease, poorer grade, vascular invasion, and bilateral ovarian disease were indicative of more aggressive disease biology. The ideal management remains largely unknown, and future collaborative efforts should optimize therapeutic strategies.


Subject(s)
Adenocarcinoma , Carcinoma, Pancreatic Ductal , Ovarian Neoplasms , Pancreatic Neoplasms , Adenocarcinoma/surgery , Carcinoma, Pancreatic Ductal/surgery , Female , Humans , Neoplasm Recurrence, Local , Ovarian Neoplasms/surgery , Pancreatectomy , Pancreatic Neoplasms/surgery , Prognosis , Retrospective Studies
10.
Lab Invest ; 100(7): 1003-1013, 2020 07.
Article in English | MEDLINE | ID: mdl-32005909

ABSTRACT

Intraductal tubulopapillary neoplasm (ITPN) is a distinct precancerous lesion in the pancreas with unique clinical and molecular features. Although in vitro studies in two-dimensional culture have led to numerous important insights in pancreatic cancer, such models are currently lacking for precancerous lesions. In this study, we report the generation and characterization of a cell line from a human pancreatic ITPN. Neoplastic cells were initially cultured in a three-dimensional organoid system, followed by transfer to two-dimensional culture. RNA sequencing revealed a gene expression profile consistent with pancreatic ductal origin, and whole genome sequencing identified many somatic mutations (including in genes involved in DNA repair and Wnt signaling) and structural rearrangements. In vitro characterization of the tumorigenic potential demonstrated a phenotype between that of normal pancreatic ductal cells and cancer cell lines. This cell line represents a valuable resource for interrogation of unique ITPN biology, as well as precancerous pancreatic lesions more generally.


Subject(s)
Cell Line, Tumor , Pancreatic Intraductal Neoplasms , Aged , Animals , Female , Humans , Male , Mice , Mice, Nude , Phenotype
11.
Gastroenterology ; 157(4): 1123-1137.e22, 2019 10.
Article in English | MEDLINE | ID: mdl-31175866

ABSTRACT

BACKGROUND & AIMS: Intraductal papillary mucinous neoplasms (IPMNs) are lesions that can progress to invasive pancreatic cancer and constitute an important system for studies of pancreatic tumorigenesis. We performed comprehensive genomic analyses of entire IPMNs to determine the diversity of somatic mutations in genes that promote tumorigenesis. METHODS: We microdissected neoplastic tissues from 6-24 regions each of 20 resected IPMNs, resulting in 227 neoplastic samples that were analyzed by capture-based targeted sequencing. Somatic mutations in genes associated with pancreatic tumorigenesis were assessed across entire IPMN lesions, and the resulting data were supported by evolutionary modeling, whole-exome sequencing, and in situ detection of mutations. RESULTS: We found a high prevalence of heterogeneity among mutations in IPMNs. Heterogeneity in mutations in KRAS and GNAS was significantly more prevalent in IPMNs with low-grade dysplasia than in IPMNs with high-grade dysplasia (P < .02). Whole-exome sequencing confirmed that IPMNs contained multiple independent clones, each with distinct mutations, as originally indicated by targeted sequencing and evolutionary modeling. We also found evidence for convergent evolution of mutations in RNF43 and TP53, which are acquired during later stages of tumorigenesis. CONCLUSIONS: In an analysis of the heterogeneity of mutations throughout IPMNs, we found that early-stage IPMNs contain multiple independent clones, each with distinct mutations, indicating their polyclonal origin. These findings challenge the model in which pancreatic neoplasms arise from a single clone. Increasing our understanding of the mechanisms of IPMN polyclonality could lead to strategies to identify patients at increased risk for pancreatic cancer.


Subject(s)
Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , Mutation , Pancreatic Intraductal Neoplasms/genetics , Pancreatic Neoplasms/genetics , Aged , Aged, 80 and over , Cell Transformation, Neoplastic/pathology , Chromogranins/genetics , Clonal Evolution , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Evolution, Molecular , Female , GTP-Binding Protein alpha Subunits, Gs/genetics , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Mutation Rate , Neoplasm Staging , Oncogene Proteins/genetics , Pancreatic Intraductal Neoplasms/pathology , Pancreatic Neoplasms/pathology , Phenotype , Proto-Oncogene Proteins p21(ras)/genetics , Retrospective Studies , Ubiquitin-Protein Ligases
12.
Mod Pathol ; 33(10): 2026-2034, 2020 10.
Article in English | MEDLINE | ID: mdl-32457408

ABSTRACT

Neoadjuvant chemotherapy (NAC) is often the treatment of choice for borderline resectable and locally advanced invasive pancreatic ductal adenocarcinoma (PDAC); however, most cancers only partially respond to therapy. We hypothesized that the location of residual neoplastic cells in resected specimens following NAC could provide a clue as to the mechanisms of resistance. PDAC cells invade the stroma but can also invade back into and spread via the pancreatic ducts, which has been referred to as "cancerization of ducts" (COD). We compared the responsiveness to chemotherapy between PDAC cells in the stroma and PDAC cells in the duct. Pancreatic resections from a total of 174 PDAC patients (NAC, n = 97; immediate surgery, n = 77) were reviewed. On hematoxylin and eosin sections, COD was identified at the same prevalence in both groups (NAC: 50/97 cases, 52%; immediate surgery: 39/77 cases, 51%; p = 0.879, Fisher's exact test). However, using quantitative image analysis of CK19 immunohistochemistry, we found that the proportion of cancer cells that were intraductal was significantly different between the NAC and immediate surgery groups (median; 12.7% vs. 1.99%, p < 0.0001, Mann-Whitney U test). This proportion was highest in patients with marked therapy responses (36.2%) compared with patients with moderate or poor responses (7.21 & 7.91%). In summary, our data suggest that intraductal components in PDAC are less responsive to chemotherapy than the remainder of the tumor, which could have important implications for therapeutic resistance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Chemotherapy, Adjuvant , Female , Humans , Male , Middle Aged , Neoadjuvant Therapy , Retrospective Studies , Treatment Outcome , Pancreatic Neoplasms
13.
Mod Pathol ; 33(12): 2544-2563, 2020 12.
Article in English | MEDLINE | ID: mdl-32704031

ABSTRACT

The publication of the "Pan-Cancer Atlas" by the Pan-Cancer Analysis of Whole Genomes Consortium, a partnership formed by The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC), provides a wonderful opportunity to reflect on where we stand in our understanding of the genetics of pancreatic cancer, as well as on the opportunities to translate this understanding to patient care. From germline variants that predispose to the development of pancreatic cancer, to somatic mutations that are therapeutically targetable, genetics is now providing hope, where there once was no hope, for those diagnosed with pancreatic cancer.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/genetics , Genetic Variation , Genomics/trends , Molecular Diagnostic Techniques/trends , Pancreatic Neoplasms/genetics , Animals , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Diffusion of Innovation , Forecasting , Genetic Predisposition to Disease , Humans , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Phenotype , Prognosis
14.
Mod Pathol ; 33(4): 639-647, 2020 04.
Article in English | MEDLINE | ID: mdl-31700162

ABSTRACT

Venous invasion is three times more common in pancreatic cancer than it is in other major cancers of the gastrointestinal tract, and venous invasion may explain why pancreatic cancer is so deadly. To characterize the patterns of venous invasion in pancreatic cancer, 52 thick slabs (up to 5 mm) of tissue were harvested from 52 surgically resected human ductal adenocarcinomas, cleared with a modified iDISCO method, and labeled with fluorescent-conjugated antibodies to cytokeratin 19, desmin, CD31, p53 and/or e-cadherin. Labeled three-dimensional (3D) pancreas cancer tissues were visualized with confocal laser scanning or light sheet microscopy. Multiple foci of venous and even arterial invasion were visualized. Venous invasion was detected more often in 3D (88%, 30/34 cases) than in conventional 2D slide evaluation (75%, 25/34 cases, P < 0.001). 3D visualization revealed pancreatic cancer cells crossing the walls of veins at multiple points, often at points where preexisting capillary structures bridge the blood vessels. The neoplastic cells often retained a ductal morphology (cohesive cells forming tubes) as they progressed from a stromal to intravenous location. Although immunolabeling with antibodies to e-cadherin revealed focal loss of expression at the leading edges of the cancers, the neoplastic cells within veins expressed e-cadherin and formed well-oriented glands. We conclude that venous invasion is almost universal in pancreatic cancer, suggesting that even surgically resectable PDAC has access to the venous spaces and thus the ability to disseminate widely. Furthermore, we observe that sustained epithelial-mesenchymal transition is not required for venous invasion in pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Epithelial-Mesenchymal Transition , Imaging, Three-Dimensional , Microscopy, Confocal , Pancreatic Neoplasms/pathology , Veins/pathology , Aged , Aged, 80 and over , Antigens, CD/analysis , Baltimore , Biomarkers, Tumor/analysis , Cadherins/analysis , Carcinoma, Pancreatic Ductal/chemistry , Carcinoma, Pancreatic Ductal/surgery , Desmin/analysis , Female , Fluorescent Antibody Technique, Indirect , Germany , Humans , Keratin-19/analysis , Male , Middle Aged , Neoplasm Invasiveness , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/surgery , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Tumor Suppressor Protein p53/analysis , Veins/chemistry
15.
J Pathol ; 247(3): 347-356, 2019 03.
Article in English | MEDLINE | ID: mdl-30430578

ABSTRACT

Intraductal papillary mucinous neoplasms (IPMNs) are precursors to pancreatic cancer; however, little is known about genetic heterogeneity in these lesions. The objective of this study was to characterize genetic heterogeneity in IPMNs at the single-cell level. We isolated single cells from fresh tissue from ten IPMNs, followed by whole genome amplification and targeted next-generation sequencing of pancreatic driver genes. We then determined single-cell genotypes using a novel multi-sample mutation calling algorithm. Our analyses revealed that different mutations in the same driver gene frequently occur in the same IPMN. Two IPMNs had multiple mutations in the initiating driver gene KRAS that occurred in unique tumor clones, suggesting the possibility of polyclonal origin or an unidentified initiating event preceding this critical mutation. Multiple mutations in later-occurring driver genes were also common and were frequently localized to unique tumor clones, raising the possibility of convergent evolution of these genetic events in pancreatic tumorigenesis. Single-cell sequencing of IPMNs demonstrated genetic heterogeneity with respect to early and late occurring driver gene mutations, suggesting a more complex pattern of tumor evolution than previously appreciated in these lesions. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Genetic Heterogeneity , Pancreatic Intraductal Neoplasms/genetics , Aged , Aged, 80 and over , DNA Mutational Analysis/methods , Female , Genes, Neoplasm/genetics , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Mutation , Pancreatic Intraductal Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics
16.
Adv Anat Pathol ; 26(1): 31-39, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30256228

ABSTRACT

Although pathologic lesions in the pancreas are 3-dimensional (3D) complex structures, we currently use thin 2D hematoxylin and eosin stained slides to study and diagnose pancreatic pathology. Two technologies, tissue clearing and advanced microscopy, have recently converged, and when used together they open the remarkable world of 3D anatomy and pathology to pathologists. Advances in tissue clearing and antibody penetration now make even dense fibrotic tissues amenable to clearing, and light sheet and confocal microscopies allow labeled cells deep within these cleared tissues to be visualized. Clearing techniques can be categorized as solvent-based or aqueous-based techniques, but both clearing methods consist of 4 fundamental steps, including pretreatment of specimens, permeabilization and/or removal of lipid, immunolabeling with antibody penetration, and clearing by refractive index matching. Specialized microscopes, including the light sheet microscope, the 2-photon microscope, and the confocal microscope, can then be used to visualize and evaluate the 3D histology. Both endocrine and exocrine pancreas pathology can then be visualized. The application of labeling and clearing to surgically resected human pancreatic parenchyma can provide detailed visualization of the complexities of normal pancreatic anatomy. It also can be used to characterize the 3D architecture of disease processes ranging from precursor lesions, such as pancreatic intraepithelial neoplasia lesions and intraductal papillary mucinous neoplasms, to infiltrating pancreatic ductal adenocarcinomas. The evaluation of 3D histopathology, including pathology of the pancreatic lesions, will provide new insights into lesions that previously were seen, and thought of, only in 2 dimensions.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Imaging, Three-Dimensional , Microscopy, Confocal , Pancreas/pathology , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Pancreatic Ductal/diagnosis , Humans , Imaging, Three-Dimensional/methods , Microscopy, Confocal/methods , Pancreatic Neoplasms/diagnosis , Parenchymal Tissue/pathology , Pancreatic Neoplasms
17.
J Appl Biomech ; 35(1): 32­36, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30080433

ABSTRACT

The Balance Error Scoring System (BESS) is a human-scored, field-based balance test used in cases of suspected concussion. Recently developed instrumented alternatives to human scoring carry substantial advantages over traditional testing, but thus far report relatively abstract outcomes which may not be useful to clinicians or coaches. In contrast, the Automated Assessment of Postural Stability (AAPS) is a computerized system that tabulates error events in accordance with the original description of the BESS. This study compared AAPS and human-based BESS scores. Twenty-five healthy adults performed the modified BESS. Tests were scored twice each by human raters (3) and the computerized system. Interrater (between-human) and inter-method (AAPS vs. human) agreement (ICC(2,1)) were calculated alongside Bland-Altman limits of agreement (LOA). Interrater analyses were significant (p<0.005) and demonstrated good to excellent agreement. Inter-method agreement analyses were significant (p<0.005), with agreement ranging from poor to excellent. Computerized scores were equivalent across rating occasions. LOA ranges for AAPS vs. the Human Average exceeded the average LOA ranges between human raters. Coaches and clinicians may consider a system such as AAPS to automate balance testing while maintaining the familiarity of human-based scoring, although scores should not yet be considered interchangeable with those of a human rater.

18.
Gut ; 66(5): 794-801, 2017 05.
Article in English | MEDLINE | ID: mdl-26801886

ABSTRACT

OBJECTIVE: Recent data supports a significant role for immune checkpoint inhibitors in the treatment of solid tumours. Here, we evaluate gastric and gastro-oesophageal junction (G/GEJ) adenocarcinomas for their expression of programmed death-ligand 1 (PD-L1), infiltration by CD8+ T cells and the relationship of both factors to patient survival. DESIGN: Thirty-four resections of primary invasive G/GEJ were stained by immunohistochemistry for PD-L1 and CD8 and by DNA in situ hybridisation for Epstein-Barr virus (EBV). CD8+ T cell densities both within tumours and at the tumour-stromal interface were analysed using whole slide digital imaging. Patient survival was evaluated according to PD-L1 status and CD8 density. RESULTS: 12% of resections showed tumour cell membranous PD-L1 expression and 44% showed expression within the immune stroma. Two cases (6%) were EBV positive, with one showing membranous PD-L1 positivity. Increasing CD8+ densities both within tumours and immune stroma was associated with increasing percentage of tumour (p=0.027) and stromal (p=0.005) PD-L1 expression. Both tumour and immune stromal PD-L1 expression and high intratumoral or stromal CD8+ T cell density (>500/mm2) were associated with worse progression-free survival (PFS) and overall survival (OS). CONCLUSIONS: PD-L1 is expressed on both tumour cells and in the immune stroma across all stages and histologies of G/GEJ. Surprisingly, we demonstrate that increasing CD8 infiltration is correlated with impaired PFS and OS. Patients with higher CD8+ T cell densities also have higher PD-L1 expression, indicating an adaptive immune resistance mechanism may be occurring. Further characterisation of the G/GEJ immune microenvironment may highlight targets for immune-based therapy.


Subject(s)
Adenocarcinoma/chemistry , B7-H1 Antigen/analysis , CD8-Positive T-Lymphocytes/immunology , DNA, Viral/analysis , Esophagogastric Junction/chemistry , Herpesvirus 4, Human , Stomach Neoplasms/chemistry , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma/virology , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/genetics , Disease-Free Survival , Esophagogastric Junction/immunology , Esophagogastric Junction/pathology , Esophagogastric Junction/virology , Female , Humans , Lymphocytes, Tumor-Infiltrating , Male , Middle Aged , Neoplasm Invasiveness , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Stomach Neoplasms/virology , Survival Rate , Young Adult
19.
Mod Pathol ; 30(11): 1551-1560, 2017 11.
Article in English | MEDLINE | ID: mdl-28731046

ABSTRACT

Tumor-infiltrating lymphocytes and immune checkpoint proteins such as PD-L1 are potential prognostic factors and therapeutic targets in breast cancer. Most studies characterizing the breast tumor immune microenvironment have focused on ductal carcinomas. Here we investigate the tumor microenvironment of primary invasive lobular carcinomas. Previously constructed tissue microarrays of 47 lobular carcinomas were labeled by immunohistochemistry for PD-L1, CD8, CD20, and FoxP3. The stromal immune infiltrate density was qualitatively scored as a percentage of tumor area: 1+ (<5%); 2+ (5-10%); 3+ (10-15%); or 4+ (>50%). The average immune cell subtype per high-power field was quantitatively scored. The percentage PD-L1 labeling on tumor-infiltrating lymphocytes was scored as none, focal (<5%), moderate (10-24%), or diffuse (50-100%). The percentage of membranous carcinoma cell PD-L1 labeling was also recorded, with <5% considered negative. All lobular carcinomas contained PD-L1+ tumor-infiltrating lymphocytes with the majority showing 1+ immune infiltrates with focal-moderate PD-L1 labeling. PD-L1 was expressed by tumor cells in 17% of lobular carcinomas. In contrast to ductal carcinomas, there was no correlation between the immune infiltrate density, the PD-L1 expression by lobular carcinoma cells, tumor grade, or the expression of estrogen receptor or human epidermal growth factor receptor-2. However, both the tumor-infiltrating lymphocyte density and the average CD8+ T-cell counts correlated with immune cell PD-L1 status (P=0.004 and 0.03, respectively). Similar to breast ductal carcinomas, PD-L1+ lobular breast carcinomas had higher numbers of PD-L1+ tumor-infiltrating lymphocytes (63%) than PD-L1- lobular carcinomas (23%; P=0.04). These data show that a subset of primary breast lobular carcinomas both express PD-L1 on tumor cells and contain PD-L1+ tumor-infiltrating lymphocytes, suggesting the possibility of both constitutive and adaptive PD-L1 expression. Together, these results support immunotherapy as a potential treatment for a subset of patients with primary invasive lobular breast carcinomas.


Subject(s)
B7-H1 Antigen/immunology , Breast Neoplasms/immunology , Carcinoma, Lobular/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Tumor Microenvironment/immunology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/immunology , Female , Humans , Middle Aged
20.
Arch Pathol Lab Med ; 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38386006

ABSTRACT

CONTEXT.­: Macroscopic precursor lesions of the pancreas represent a complex clinical management problem. Molecular characterization of pancreatic cysts has helped to confirm and refine clinical and pathologic classifications of these lesions, inform our understanding of tumorigenesis in the pancreas, and provide opportunities for preoperative diagnosis. OBJECTIVE.­: To review the pathologic classification of macroscopic cystic lesions of the pancreas: intraductal papillary mucinous neoplasms (IPMNs), mucinous cystic neoplasms (MCNs), intraductal oncocytic papillary neoplasms (IOPNs), and intraductal tubulopapillary neoplasms (ITPNs), and to describe our current state of understanding of their molecular underpinnings, relationship to invasive carcinomas, and implications for diagnosis and prognostication. DATA SOURCES.­: We assessed the current primary literature and current World Health Organization Classification of Digestive System Tumours. CONCLUSIONS.­: Macroscopic cystic lesions of the pancreas are morphologically and molecularly diverse. IPMNs and MCNs share mucinous cytoplasm with papillae. MCNs are defined by ovarian-type stroma. IOPNs have granular eosinophilic cytoplasm, prominent nucleoli, and complex, arborizing papillae. ITPNs demonstrate complex, back-to-back tubules and anastomosing papillae and lack prominent intracellular mucin. IPMNs and MCNs are characterized by driver mutations in KRAS/GNAS (IPMNs) and KRAS (MCNs), with later driver events in RNF43, CDKN2A, SMAD4, and TP53. In contrast, IOPNs and ITPNs have recurrent rearrangements in PRKACA/PRKACB and MAPK-associated genes, respectively. The recurrent alterations described in cysts provide an opportunity for diagnosis using aspirated cyst fluid. Molecular characterization of IPMNs shows a striking spatial and mutational heterogeneity, challenging traditional models of neoplastic development and creating challenges to interpretation of cyst fluid sequencing results.

SELECTION OF CITATIONS
SEARCH DETAIL