Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Cancer Discov ; 14(4): 683-689, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38571435

ABSTRACT

Research on precancers, as defined as at-risk tissues and early lesions, is of high significance given the effectiveness of early intervention. We discuss the need for risk stratification to prevent overtreatment, an emphasis on the role of genetic and epigenetic aging when considering risk, and the importance of integrating macroenvironmental risk factors with molecules and cells in lesions and at-risk normal tissues for developing effective intervention and health policy strategies.


Subject(s)
Precancerous Conditions , Humans , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Risk Factors
2.
Radiology ; 310(1): e232078, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38289210

ABSTRACT

Background The natural history of colorectal polyps is not well characterized due to clinical standards of care and other practical constraints limiting in vivo longitudinal surveillance. Established CT colonography (CTC) clinical screening protocols allow surveillance of small (6-9 mm) polyps. Purpose To assess the natural history of colorectal polyps followed with CTC in a clinical screening program, with histopathologic correlation for resected polyps. Materials and Methods In this retrospective study, CTC was used to longitudinally monitor small colorectal polyps in asymptomatic adult patients from April 1, 2004, to August 31, 2020. All patients underwent at least two CTC examinations. Polyp growth patterns across multiple time points were analyzed, with histopathologic context for resected polyps. Regression analysis was performed to evaluate predictors of advanced histopathology. Results In this study of 475 asymptomatic adult patients (mean age, 56.9 years ± 6.7 [SD]; 263 men), 639 unique polyps (mean initial diameter, 6.3 mm; volume, 50.2 mm3) were followed for a mean of 5.1 years ± 2.9. Of these 639 polyps, 398 (62.3%) underwent resection and histopathologic evaluation, and 41 (6.4%) proved to be histopathologically advanced (adenocarcinoma, high-grade dysplasia, or villous content), including two cancers and 38 tubulovillous adenomas. Advanced polyps showed mean volume growth of +178% per year (752% per year for adenocarcinomas) compared with +33% per year for nonadvanced polyps and -3% per year for unresected, unretrieved, or resolved polyps (P < .001). In addition, 90% of histologically advanced polyps achieved a volume of 100 mm3 and/or volume growth rate of 100% per year, compared with 29% of nonadvanced and 16% of unresected or resolved polyps (P < .001). Polyp volume-to-diameter ratio was also significantly greater for advanced polyps. For polyps observed at three or more time points, most advanced polyps demonstrated an initial slower growth interval, followed by a period of more rapid growth. Conclusion Small colorectal polyps ultimately proving to be histopathologically advanced neoplasms demonstrated substantially faster growth and attained greater overall size compared with nonadvanced polyps. Clinical trial registration no. NCT00204867 © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Dachman in this issue.


Subject(s)
Adenocarcinoma , Colonic Polyps , Colonography, Computed Tomographic , Adult , Male , Humans , Middle Aged , Colonic Polyps/diagnostic imaging , Retrospective Studies , Physical Examination
3.
Am J Clin Exp Urol ; 11(6): 452-466, 2023.
Article in English | MEDLINE | ID: mdl-38148937

ABSTRACT

Fibroblast growth factor (FGF) is a secreted ligand that is widely expressed in embryonic tissues but its expression decreases with age. In the developing prostate, FGF5 has been proposed to interact with the Hedgehog (Hh) signaling pathway to guide mitogenic processes. In the adult prostate, the FGF/FGFR signaling axis has been implicated in prostate carcinogenesis, but focused studies on FGF5 functions in the prostate are limited. Functional studies completed in other cancer models point towards FGF5 overexpression as an oncogenic driver associated with stemness, metastatic potential, proliferative capacity, and increased tumor grade. In this review, we explore the significance of FGF5 as a therapeutic target in prostate cancer (PCa) and other malignancies; and we introduce a potential route of investigation to link FGF5 to benign prostatic hyperplasia (BPH). PCa and BPH are two primary contributors to the disease burden of the aging male population and have severe implications on quality of life, psychological wellbeing, and survival. The development of new FGF5 inhibitors could potentially alleviate the health burden of PCa and BPH in the aging male population.

4.
J Mol Diagn ; 2023 Aug 05.
Article in English | MEDLINE | ID: mdl-37544360

ABSTRACT

Microsatellite instability (MSI) is an evolving biomarker for cancer detection and treatment. MSI was first used to identify patients with Lynch syndrome, a hereditary form of colorectal cancer (CRC), but has recently become indispensable in predicting patient response to immunotherapy. To address the need for pan-cancer MSI detection, a new multiplex assay was developed that uses novel long mononucleotide repeat (LMR) markers to improve sensitivity. A total of 469 tumor samples from 20 different cancer types, including 319 from patients with Lynch syndrome, were tested for MSI using the new LMR MSI Analysis System. Results were validated by using deficient mismatch repair (dMMR) status according to immunohistochemistry as the reference standard and compared versus the Promega pentaplex MSI panel. The sensitivity of the LMR panel for detection of dMMR status by immunohistochemistry was 99% for CRC and 96% for non-CRC. The overall percent agreement between the LMR and Promega pentaplex panels was 99% for CRC and 89% for non-CRC tumors. An increased number of unstable markers and the larger size shifts observed in dMMR tumors using the LMR panel increased confidence in MSI determinations. The LMR MSI Analysis System expands the spectrum of cancer types in which MSI can be accurately detected.

5.
Gut ; 72(12): 2321-2328, 2023 Nov 24.
Article in English | MEDLINE | ID: mdl-37507217

ABSTRACT

BACKGROUND AND AIMS: The natural history of small polyps is not well established and rests on limited evidence from barium enema studies decades ago. Patients with one or two small polyps (6-9 mm) at screening CT colonography (CTC) are offered CTC surveillance at 3 years but may elect immediate colonoscopy. This practice allows direct observation of the growth of subcentimetre polyps, with histopathological correlation in patients undergoing subsequent polypectomy. DESIGN: Of 11 165 asymptomatic patients screened by CTC over a period of 16.4 years, 1067 had one or two 6-9 mm polyps detected (with no polyps ≥10 mm). Of these, 314 (mean age, 57.4 years; M:F, 141:173; 375 total polyps) elected immediate colonoscopic polypectomy, and 382 (mean age 57.0 years; M:F, 217:165; 481 total polyps) elected CTC surveillance over a mean of 4.7 years. Volumetric polyp growth was analysed, with histopathological correlation for resected polyps. Polyp growth and regression were defined as volume change of ±20% per year, with rapid growth defined as +100% per year (annual volume doubling). Regression analysis was performed to evaluate predictors of advanced histology, defined as the presence of cancer, high-grade dysplasia (HGD) or villous components. RESULTS: Of the 314 patients who underwent immediate polypectomy, 67.8% (213/314) harboured adenomas, 2.2% (7/314) with advanced histology; no polyps contained cancer or HGD. Of 382 patients who underwent CTC surveillance, 24.9% (95/382) had polyps that grew, while 62.0% (237/382) remained stable and 13.1% (50/382) regressed in size. Of the 58.6% (224/382) CTC surveillance patients who ultimately underwent colonoscopic resection, 87.1% (195/224) harboured adenomas, 12.9% (29/224) with advanced histology. Of CTC surveillance patients with growing polyps who underwent resection, 23.2% (19/82) harboured advanced histology vs 7.0% (10/142) with stable or regressing polyps (OR: 4.0; p<0.001), with even greater risk of advanced histology in those with rapid growth (63.6%, 14/22, OR: 25.4; p<0.001). Polyp growth, but not patient age/sex or polyp morphology/location were significant predictors of advanced histology. CONCLUSION: Small 6-9 mm polyps present overall low risk to patients, with polyp growth strongly associated with higher risk lesions. Most patients (75%) with small 6-9 mm polyps will see polyp stability or regression, with advanced histology seen in only 7%. The minority of patients (25%) with small polyps that do grow have a 3-fold increased risk of advanced histology.


Subject(s)
Adenoma , Colonic Polyps , Colonography, Computed Tomographic , Colorectal Neoplasms , Humans , Middle Aged , Colonic Polyps/diagnostic imaging , Colonic Polyps/surgery , Colonic Polyps/pathology , Colonoscopy , Adenoma/diagnostic imaging , Adenoma/surgery , Adenoma/pathology , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/surgery , Colorectal Neoplasms/pathology
6.
Nat Commun ; 13(1): 2971, 2022 05 27.
Article in English | MEDLINE | ID: mdl-35624112

ABSTRACT

Spatial transcriptomics is a powerful and widely used approach for profiling the gene expression landscape across a tissue with emerging applications in molecular medicine and tumor diagnostics. Recent spatial transcriptomics experiments utilize slides containing thousands of spots with spot-specific barcodes that bind RNA. Ideally, unique molecular identifiers (UMIs) at a spot measure spot-specific expression, but this is often not the case in practice due to bleed from nearby spots, an artifact we refer to as spot swapping. To improve the power and precision of downstream analyses in spatial transcriptomics experiments, we propose SpotClean, a probabilistic model that adjusts for spot swapping to provide more accurate estimates of gene-specific UMI counts. SpotClean provides substantial improvements in marker gene analyses and in clustering, especially when tissue regions are not easily separated. As demonstrated in multiple studies of cancer, SpotClean improves tumor versus normal tissue delineation and improves tumor burden estimation thus increasing the potential for clinical and diagnostic applications of spatial transcriptomics technologies.


Subject(s)
Electronic Data Processing , Transcriptome , Cluster Analysis , Models, Statistical
7.
Hum Pathol ; 119: 1-14, 2022 01.
Article in English | MEDLINE | ID: mdl-34655611

ABSTRACT

Colorectal cancer (CRC) is a leading cause of cancer death in the United States. Standard treatment for advanced-stage CRC for decades has included 5-fluorouracil-based chemotherapy. More recently, targeted therapies for metastatic CRC are being used based on the individual cancer's molecular profile. In the past few years, several different molecular subtype schemes for human CRC have been developed. The molecular subtypes can be distinguished by gene expression signatures and have the potential to be used to guide treatment decisions. However, many subtyping classification methods were developed using mRNA expression levels of hundreds to thousands of genes, making them impractical for clinical use. In this study, we assessed whether an immunohistochemical approach could be used for molecular subtyping of CRCs. We validated two previously published, independent sets of immunohistochemistry classifiers and modified the published methods to improve the accuracy of the scoring methods. In addition, we evaluated whether protein and genetic signatures identified originally in the mouse were linked to clinical outcomes of patients with CRC. We found that low DDAH1 or low GAL3ST2 protein levels in human CRCs correlate with poor patient outcomes. The results of this study have the potential to impact methods for determining the prognosis and therapy selection for patients with CRC.


Subject(s)
Adenocarcinoma/chemistry , Amidohydrolases/analysis , Biomarkers, Tumor/analysis , Colorectal Neoplasms/chemistry , Immunohistochemistry , Sulfotransferases/analysis , Adenocarcinoma/classification , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Aged , Amidohydrolases/genetics , Animals , Biomarkers, Tumor/genetics , Colorectal Neoplasms/classification , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Genes, APC , Humans , Male , Mice, Transgenic , Middle Aged , Predictive Value of Tests , Prognosis , Reproducibility of Results , Sulfotransferases/genetics , Tissue Array Analysis
8.
Cancer Rep (Hoboken) ; 5(2): e1459, 2022 02.
Article in English | MEDLINE | ID: mdl-34245130

ABSTRACT

BACKGROUND: Data are steadily accruing that demonstrate that intestinal tumors are frequently derived from multiple founding cells, resulting in tumors comprised of distinct ancestral clones that might cooperate or alternatively compete, thereby potentially impacting different phases of the disease process. AIM: We sought to determine whether tumors with a multi-ancestral architecture involving at least two distinct clones show increased tumor number, growth, progression, or resistance to drug intervention. METHODS: Mice carrying the Min allele of Apc were generated that were mosaic with only a subset of cells in the intestinal epithelium expressing an activated form of PI3K, a key regulatory kinase affecting several important cellular processes. These cells were identifiable as they fluoresced green, whereas all other cells fluoresced red. RESULTS: Cell lineage tracing revealed that many intestinal tumors from our mouse model were derived from at least two founding cells, those expressing the activated PI3K (green) and those which did not (red). Heterotypic tumors with a multi-ancestral architecture as evidenced by a mixture of green and red cells exhibited increased tumor growth and invasiveness. Clonal architecture also had an impact on tumor response to low-dose aspirin. Aspirin treatment resulted in a greater reduction of heterotypic tumors derived from multiple founding cells as compared to tumors derived from a single founding cell. CONCLUSION: These data indicate that genetically distinct tumor-founding cells can contribute to early intratumoral heterogeneity. The coevolution of the founding cells and their progeny enhances colon tumor progression and impacts the response to aspirin. These findings are important to a more complete understanding of tumorigenesis with consequences for several distinct models of tumor evolution. They also have practical implications to the clinic. Mouse models with heterogenous tumors are likely better for predicting drug efficacy as compared to models in which the tumors are highly homogeneous. Moreover, understanding how interactions among different populations in a single heterotypic tumor with a multi-ancestral architecture impact response to a single agent and combination therapies are necessary to fully develop personalized medicine.


Subject(s)
Cell Transformation, Neoplastic/genetics , Intestinal Neoplasms/genetics , Animals , Antineoplastic Agents/pharmacology , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Disease Progression , Drug Resistance, Neoplasm/genetics , Intestinal Neoplasms/drug therapy , Intestinal Neoplasms/pathology , Mice , Mice, Transgenic
9.
Int J Radiat Biol ; 97(8): 1140-1151, 2021.
Article in English | MEDLINE | ID: mdl-33720813

ABSTRACT

PURPOSE: Estimating cancer risk associated with interplanetary space travel is complicated. Human exposure data to high atomic number, high-energy (HZE) radiation is lacking, so data from low linear energy transfer (low-LET) γ-ray radiation is used in risk models, with the assumption that HZE and γ-ray radiation have comparable biological effects. This assumption has been challenged by reports indicating that HZE radiation might produce more aggressive tumors. The goal of this research is to test whether high-LET HZE radiation induced tumors are more aggressive. MATERIALS AND METHODS: Murine models of mammary and liver cancer were used to compare the impact of exposure to 0.2Gy of 300MeV/n silicon ions, 3 Gy of γ-rays or no radiation. Numerous measures of tumor aggressiveness were assessed. RESULTS: For the mammary cancer models, there was no significant change in the tumor latency or metastasis in silicon-irradiated mice compared to controls. For the liver cancer models, we observed an increase in tumor incidence but not tumor aggressiveness in irradiated mice. CONCLUSION: Tumors in the HZE-irradiated mice were not more aggressive than those arising from exposure to low-LET γ-rays or spontaneously. Thus, enhanced aggressiveness does not appear to be a uniform characteristic of all tumors in HZE-irradiated animals.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Mammary Neoplasms, Experimental/pathology , Animals , Dose-Response Relationship, Radiation , Female , Humans , Linear Energy Transfer , Mice
10.
Physiol Rep ; 8(5): e14391, 2020 03.
Article in English | MEDLINE | ID: mdl-32170841

ABSTRACT

Cancer cachexia is the loss of lean muscle mass with or without loss of fat mass that is often highlighted by a progressive loss of skeletal muscle mass and function. The mechanisms behind the cachexia-related loss of skeletal muscle are poorly understood, including cachexia-related muscle functional impairments. Existing models have revealed some potential mechanisms, but appear limited to how the cancer develops and the type of tumors that form. We studied the C57BL6/J (B6) ApcMin/+ Tg::Fabp1-Cre TG::PIK3ca* (CANCER) mouse. In this model, mice develop highly aggressive intestinal cancers. We tested whether CANCER mice develop cancer cachexia, if muscle function is altered and if sex differences are present. Both female and male mice, B6 (CONTROL) and CANCER mice, were analyzed to determine body weight, hindlimb muscle mass, protein concentration, specific force, and fatigability. Female CANCER mice had reduced body weight and hindlimb muscle mass compared with female CONTROL mice, but lacked changes in protein concentration and specific force. Male CANCER mice had reduced protein concentration and reduced specific force, but lacked altered body weight and muscle mass. There were no changes in fatigability in either group. Our study demonstrates that CANCER mice present an early stage of cachexia, have reduced specific force in male CANCER mice and develop a sex-dependent cachexia phenotype. However, CANCER mice lack certain aspects of the syndrome seen in the human scenario and, therefore, using the CANCER mice as a preclinical model does not seem sufficient in order to maximize the translation of preclinical findings to humans.


Subject(s)
Cachexia/pathology , Colorectal Neoplasms/pathology , Muscle, Skeletal/pathology , Animals , Cachexia/complications , Cachexia/physiopathology , Colorectal Neoplasms/complications , Colorectal Neoplasms/physiopathology , Disease Models, Animal , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/physiopathology , Sex Characteristics
11.
J Pathol ; 250(2): 231-242, 2020 02.
Article in English | MEDLINE | ID: mdl-31674011

ABSTRACT

We genetically engineered expression of an activated form of P110 alpha, the catalytic subunit of PI3K, in mouse prostate epithelium to create a mouse model of direct PI3K activation (Pbsn-cre4Prb;PI3KGOF/+ ). We hypothesized that direct activation would cause rapid neoplasia and cancer progression. Pbsn-cre4Prb;PI3KGOF/+ mice developed widespread prostate intraepithelial hyperplasia, but stromal invasion was limited and overall progression was slower than anticipated. However, the model produced profound and progressive stromal remodeling prior to explicit epithelial neoplasia. Increased stromal cellularity and inflammatory infiltrate were evident as early as 4 months of age and progressively increased through 12 months of age, the terminal endpoint of this study. Prostatic collagen density and phosphorylated SMAD2-positive prostatic stromal cells were expansive and accumulated with age, consistent with pro-fibrotic TGF-ß pathway activation. Few reported mouse models accumulate prostate-specific collagen to the degree observed in Pbsn-cre4Prb;PI3KGOF/+ . Our results indicate a signaling process beginning with prostatic epithelial PI3K and TGF-ß signaling that drives prostatic stromal hypertrophy and collagen accumulation. These mice afford a unique opportunity to explore molecular mechanisms of prostatic collagen accumulation that is relevant to cancer progression, metastasis, inflammation and urinary dysfunction. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/physiology , Collagen/metabolism , Prostate/enzymology , Prostatic Intraepithelial Neoplasia/enzymology , Prostatic Neoplasms/enzymology , Aging/pathology , Animals , Disease Models, Animal , Disease Progression , Epithelium/enzymology , Male , Mice, Mutant Strains , Phosphorylation , Prostate/metabolism , Prostate/pathology , Prostatic Hyperplasia/enzymology , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Signal Transduction , Smad2 Protein/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology , Transforming Growth Factor beta/physiology
12.
Radiat Res ; 193(1): 88-94, 2020 01.
Article in English | MEDLINE | ID: mdl-31738662

ABSTRACT

Radiation-induced cancer is an ongoing and significant problem, with sources that include clinics worldwide in which 3.1 billion radiology exams are performed each year, as well as a variety of other scenarios such as space travel and nuclear cleanup. These radiation exposures are typically anticipated, and the exposure is typically well below 1 Gy. When radiation-induced (actually ROS-induced) DNA mutation is prevented, then so too are downstream radiation-induced cancers. Currently, there is no protection available against the effects of such <1 Gy radiation exposures. In this study, we address whether the new PrC-210 ROS-scavenger is effective in protecting p53-deficient (p53-/-) mice against X-ray-induced accelerated tumor mortality; this is the most sensitive radiation tumorigenesis model currently known. Six-day-old p53-/- pups received a single intraperitoneal PrC-210 dose [0.5 maximum tolerated dose (MTD)] or vehicle, and 25 min later, pups received 4.0 Gy X-ray irradiation. At 5 min postirradiation, blood was collected to quantify white blood cell c-H2AX foci. Over the next 250 days, tumor-associated deaths were recorded. Findings revealed that when administered 25 min before 4 Gy X-ray irradiation, PrC-210 reduced DNA damage (c-H2AX foci) by 40%, and in a notable coincidence, caused a 40% shift in tumor latency/incidence, and the 0.5 MTD PrC210 dose had no discernible toxicities in these p53-/- mice. Essentially, the moles of PrC-210 thiol within a single 0.5 MTD PrC-210 dose suppressed the moles of ROS generated by 40% of the 4 Gy X-ray dose administered to p53-/- pups, and in doing so, eliminated the lifetime leukemia/lymphoma risk normally residing "downstream" of that 40% of the 4 Gy dose. In conclusion: 1. PrC-210 is readily tolerated by the 6-day-old p53-/- mice, with no discernible lifetime toxicities; 2. PrC-210 does not cause the nausea, emesis or hypotension that preclude clinical use of earlier aminothiols; and 3. PrC-210 significantly increased survival after 4 Gy irradiation in the p53-/- mouse model.


Subject(s)
Diamines/pharmacology , Neoplasms, Radiation-Induced/mortality , Radiation-Protective Agents/pharmacology , Sulfhydryl Compounds/pharmacology , Tumor Suppressor Protein p53/deficiency , Animals , Carcinogenesis/drug effects , Carcinogenesis/radiation effects , DNA Damage , Diamines/blood , Female , Humans , Infant, Newborn , Male , Mice , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/pathology , Neoplasms, Radiation-Induced/prevention & control , Radiation-Protective Agents/metabolism , Sulfhydryl Compounds/blood
13.
Int J Cancer ; 145(11): 3022-3032, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31018249

ABSTRACT

The normal colon epithelium is transformed into its neoplastic counterpart through a series of genetic alterations in driver genes including activating mutations in PIK3CA. Treatment often involves surgery followed by 5-fluorouracil (5-FU) based therapy, which has limited efficiency and serious side effects. We sought to determine whether fisetin, a dietary flavonoid, alone or in combination with 5-FU affected tumorigenesis in the mammalian intestine. We first determined the effect of fisetin, 5-FU or their combination on PIK3CA-mutant and PIK3CA wild-type colon cancer cells by assessing cell viability, colony formation, apoptosis and effects on PI3K/AKT/mTOR signaling. Treatment of PIK3CA-mutant cells with fisetin and 5-FU reduced the expression of PI3K, phosphorylation of AKT, mTOR, its target proteins, constituents of mTOR signaling complex and this treatment increased the phosphorylation of AMPKα. We then determined whether fisetin and 5-FU together or singly affected tumorigenesis in ApcMin/+ mice that also express constitutively active PI3K in the distal small intestine and colon. Tumor incidence was markedly lower in fisetin-treated FC1 3K1 ApcMin/+ mice that also express constitutively active PI3K in distal small intestine and colon, as compared to control animals, indicating that fisetin is a strong preventive agent. In addition, the combination of fisetin and 5-FU also reduced the total number of intestinal tumors. Fisetin could be used as a preventive agent plus an adjuvant with 5-FU for the treatment of PIK3CA-mutant colorectal cancer.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Colorectal Neoplasms/drug therapy , Flavonoids/administration & dosage , Fluorouracil/administration & dosage , Mutation , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Drug Synergism , Flavonoids/pharmacology , Flavonols , Fluorouracil/pharmacology , HCT116 Cells , HT29 Cells , Humans , Mice , Phosphorylation/drug effects , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
14.
Acad Radiol ; 26(1): 30-37, 2019 01.
Article in English | MEDLINE | ID: mdl-29566994

ABSTRACT

RATIONALE AND OBJECTIVES: To (1) apply a quantitative volumetric textural analysis (VTA) to colorectal masses at CT colonography (CTC) for the differentiation of malignant and benign lesions and to (2) compare VTA with human performance. MATERIALS AND METHODS: A validated, quantitative VTA method was applied to 63 pathologically proven colorectal masses (mean size, 4.2 cm; range, 3-8 cm) at noncontrast CTC in 59 adults (mean age, 66.5 years; range, 45.9-91.6 years). Fifty-one percent (32/63) of the masses were invasive adenocarcinoma, and the remaining 49% (31/63) were large benign adenomas. Three readers with CTC experience independently assessed the likelihood of malignancy using a 5-point scale (1 = definitely benign, 2 = probably benign, 3 = indeterminate, 4 = probably malignant, 5 = definitely malignant). Areas under the curve (AUCs) and accuracy levels were compared. RESULTS: VTA achieved optimal sensitivity of 83.6% vs 91.7% for human readers (P = .034), with specificities of 87.5% and 77.4%, respectively (P = .007). No significant difference in overall accuracy was seen between VTA and human readers (85.5% vs 84.7%, P = .753). The AUC for differentiating benign and malignant lesions was 0.936 for VTA and 0.917 for human readers. Intraclass correlation coefficient among the human readers was 0.76, indicating good to excellent agreement. CONCLUSION: VTA demonstrates excellent performance for distinguishing benign from malignant colorectal masses (≥3 cm) at CTC, comparable yet potentially complementary to experienced human performance.


Subject(s)
Adenocarcinoma/diagnostic imaging , Adenoma/diagnostic imaging , Colonography, Computed Tomographic , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/pathology , Image Processing, Computer-Assisted/methods , Adenocarcinoma/pathology , Adenoma/pathology , Aged , Aged, 80 and over , Area Under Curve , Female , Humans , Male , Middle Aged , ROC Curve
15.
Gastroenterol Clin North Am ; 47(3): 515-536, 2018 09.
Article in English | MEDLINE | ID: mdl-30115435

ABSTRACT

For decades, colorectal screening strategies have been largely driven by static features, particularly polyp size. Although cross-sectional features of polyp size, morphology, and location are important determinants of clinical relevance before histology, they lack any dynamic information on polyp growth rates. Computed tomography colonography allows for in vivo surveillance of colorectal polyps, providing volumetric growth rates that are providing new insights into tumorigenesis. In this article, existing cross-sectional and longitudinal data on colorectal polyps are reviewed, with an emphasis on how these features may affect clinical relevance and patient management.


Subject(s)
Colonic Polyps/pathology , Colorectal Neoplasms/pathology , Mass Screening/methods , Colon/pathology , Colonic Polyps/diagnosis , Colorectal Neoplasms/diagnosis , Diagnostic Techniques, Digestive System , Female , Humans , Male , Rectum/pathology
16.
PLoS One ; 12(11): e0188413, 2017.
Article in English | MEDLINE | ID: mdl-29145476

ABSTRACT

Though many methods can be used to identify cell types contained in complex tissues, most require cell disaggregation and destroy information about where cells reside in relation to their microenvironment. Here, we describe a polytomous key for cell type identification in intact sections of adult mouse prostate and prostatic urethra. The key is organized as a decision tree and initiates with one round of immunostaining for nerve, epithelial, fibromuscular/hematolymphoid, or vascular associated cells. Cell identities are recursively eliminated by subsequent staining events until the remaining pool of potential cell types can be distinguished by direct comparison to other cells. We validated our identification key using wild type adult mouse prostate and urethra tissue sections and it currently resolves sixteen distinct cell populations which include three nerve fiber types as well as four epithelial, five fibromuscular/hematolymphoid, one nerve-associated, and three vascular-associated cell types. We demonstrate two uses of this novel identification methodology. We first used the identification key to characterize prostate stromal cell type changes in response to constitutive phosphatidylinositide-3-kinase activation in prostate epithelium. We then used the key to map cell lineages in a new reporter mouse strain driven by Wnt10aem1(cre/ERT2)Amc. The identification key facilitates rigorous and reproducible cell identification in prostate tissue sections and can be expanded to resolve additional cell types as new antibodies and other resources become available.


Subject(s)
Prostate/metabolism , Urethra/metabolism , Animals , Immunohistochemistry , Male , Mice , Prostate/cytology , Urethra/cytology
17.
Expert Rev Gastroenterol Hepatol ; 11(8): 723-729, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28503955

ABSTRACT

INTRODUCTION: Tumors in the large intestine have been postulated to arise via a stepwise accumulation of mutations, a process that takes up to 20 years. Recent advances in lineage tracing and DNA sequencing, however, are revealing new evolutionary models that better explain the vast amount of heterogeneity observed within and across colorectal tumors. Areas covered: A review of the literature supporting a novel model of colorectal tumor evolution was conducted. The following commentary examines the basic science and clinical evidence supporting a modified view of tumor initiation and progression in the colon. Expert commentary: The proposed 'cancer punctuated equilibrium' model of tumor evolution better explains the variability seen within and across polyps of the colon and rectum. Small colorectal polyps (6-9mm) followed longitudinally by interval imaging with CT colonography have been reported to have multiple fates: some growing, some remaining static in size, and others regressing in size over time. This new model allows for this variability in growth behavior and supports the hypothesis that some tumors can be 'born to be bad' as originally postulated by Sottoriva and colleagues, with very early molecular events impacting tumor fitness and growth behavior in the later stages of the disease process.


Subject(s)
Biological Evolution , Cell Transformation, Neoplastic/pathology , Colonic Polyps/pathology , Colorectal Neoplasms/pathology , Animals , Biomarkers, Tumor/genetics , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Colonic Polyps/diagnostic imaging , Colonic Polyps/genetics , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/genetics , Evolution, Molecular , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Models, Biological , Phenotype , Time Factors , Tumor Burden
18.
Gut ; 66(12): 2132-2140, 2017 12.
Article in English | MEDLINE | ID: mdl-27609830

ABSTRACT

OBJECTIVE AND DESIGN: The goal of the study was to determine whether the mutational profile of early colorectal polyps correlated with growth behaviour. The growth of small polyps (6-9 mm) that were first identified during routine screening of patients was monitored over time by interval imaging with CT colonography. Mutations in these lesions with known growth rates were identified by targeted next-generation sequencing. The timing of mutational events was estimated using computer modelling and statistical inference considering several parameters including allele frequency and fitness. RESULTS: The mutational landscape of small polyps is varied both within individual polyps and among the group as a whole but no single alteration was correlated with growth behaviour. Polyps carried 0-3 pathogenic mutations with the most frequent being in APC, KRAS/NRAS, BRAF, FBXW7 and TP53. In polyps with two or more pathogenic mutations, allele frequencies were often variable, indicating the presence of multiple populations within a single tumour. Based on computer modelling, detectable mutations occurred at a mean polyp size of 30±35 crypts, well before the tumour is of a clinically detectable size. CONCLUSIONS: These data indicate that small colon polyps can have multiple pathogenic mutations in crucial driver genes that arise early in the existence of a tumour. Understanding the molecular pathway of tumourigenesis and clonal evolution in polyps that are at risk for progressing to invasive cancers will allow us to begin to better predict which polyps are more likely to progress into adenocarcinomas and which patients are at greater risk of developing advanced disease.


Subject(s)
Colonic Polyps/genetics , Colorectal Neoplasms/genetics , Mutation , Alleles , Cell Transformation, Neoplastic , Colonic Polyps/diagnostic imaging , Colonic Polyps/pathology , Colonography, Computed Tomographic , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/pathology , Disease Progression , Female , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Humans , Male , Microsatellite Instability , Middle Aged , Models, Genetic , Models, Statistical , Neoplasm Staging , Phenotype
19.
Cancer Prev Res (Phila) ; 9(8): 638-41, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27199343

ABSTRACT

Advances in DNA sequencing have created new opportunities to better understand the biology of cancers. Attention is currently focused on precision medicine: does a cancer carry a mutation that is targetable with already available drugs? But, the timing at which multiple, targetable mutations arise during the adenoma to carcinoma sequence remains unresolved. Borras and colleagues identified mutations and allelic imbalance in at-risk mucosa and early polyps in the human colon. Their analyses indicate that mutations in key genes can arise quite early during tumorigenesis and that polyps are often multiclonal with at least two clones. These results are consistent with the "Big Bang" model of tumorigenesis, which postulates that intratumoral heterogeneity is a consequence of a mutational burst in the first few cell divisions following initiation that drives divergence from a single founder with unique but related clones coevolving. Emerging questions center around the ancestry of the tumor and impact of early intratumoral heterogeneity on tumor establishment, growth, progression, and most importantly, response to therapeutic intervention. Additional sequencing studies in which samples, especially at-risk tissue and premalignant neoplasms, are analyzed from animal models and humans will further our understanding of tumorigenesis and lead to more effective strategies for prevention and treatment. Cancer Prev Res; 9(8); 638-41. ©2016 AACRSee related article by Borras, et al., Cancer Prev Res 2016;9(6):417-427.


Subject(s)
Carcinogenesis/genetics , Colon/pathology , Colonic Polyps/genetics , Colorectal Neoplasms/genetics , Precancerous Conditions/genetics , Adenoma/genetics , Adenoma/pathology , Animals , Carcinogenesis/pathology , Colonic Polyps/pathology , Colorectal Neoplasms/pathology , DNA Mutational Analysis , Genes, APC , Genetic Heterogeneity , Humans , Mice , Mutation , Precancerous Conditions/pathology
20.
Hum Pathol ; 51: 124-33, 2016 May.
Article in English | MEDLINE | ID: mdl-27067790

ABSTRACT

The purpose of this study was to objectively investigate ß-catenin and LEF1 abundance, subcellular localization, and colocalization across benign and staged prostate cancer (PCa) specimens. A tissue microarray containing tumor-adjacent histologically benign prostate tissue (BPT; n = 48 patients), high-grade prostatic intraepithelial neoplasia (HGPIN; n = 25), localized PCa (n = 42), aggressive PCa (n = 31), and metastases (n = 22) was stained using multiplexed immunohistochemistry with antibodies toward E-cadherin, ß-catenin, and LEF1. Multispectral imaging was used for quantitation, and protein expression and colocalization was evaluated across PCa progression. Stromal nuclear ß-catenin abundance was greater in HGPIN and PCa compared with BPT (P < .05 for both), and epithelial nuclear ß-catenin abundance was lower in metastatic PCa than in BPT (P < .05 for both). Epithelial and stromal nuclear LEF1 abundance was greater in HGPIN compared with BPT, whereas epithelial nuclear LEF1 was also greater in metastases. The proportion of epithelial and stromal nuclear double-positive ß-catenin(+)/LEF1(+) cells was greater in HGPIN compared with BPT. In addition, the proportion of epithelial ß-catenin(+)/LEF1(+) cells was greater in localized PCa and metastases compared with BPT. A significant amount of stromal cells were positive for LEF1 but not ß-catenin. ß-Catenin and LEF1 abundance were negatively correlated in the epithelium (P < .0001) but not the stroma (P > .05). We conclude that ß-catenin and LEF1 colocalization is increased in HGPIN and metastasis relative to BPT, suggesting a role for ß-catenin/LEF1-mediated transcription in both malignant transformation and metastasis of PCa. Furthermore, our results suggest that LEF1 abundance alone is not a reliable readout for ß-catenin activity in prostate tissues.


Subject(s)
Biomarkers, Tumor/analysis , Lymphoid Enhancer-Binding Factor 1/biosynthesis , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , beta Catenin/biosynthesis , Disease Progression , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Lymphoid Enhancer-Binding Factor 1/analysis , Male , Retrospective Studies , Tissue Array Analysis , beta Catenin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...