ABSTRACT
Epithelial ovarian cancer (EOC) is often diagnosed in advanced stage with peritoneal dissemination. Recent studies indicate that aberrant accumulation of collagen fibers in tumor stroma has a variety of effects on tumor progression. We refer to remodeled fibrous stroma with altered expression of collagen molecules, increased stiffness, and highly oriented collagen fibers as tumor-associated fibrosis (TAF). TAF contributes to EOC cell invasion and metastasis in the intraperitoneal cavity. However, an understanding of molecular events involved is only just beginning to emerge. Further development in this field will lead to new strategies to treat EOC. In this review, we focus on the recent findings on how the TAF contributes to EOC malignancy. Furthermore, we will review the recent initiatives and future therapeutic strategies for targeting TAF in EOC.
Subject(s)
Fibrosis , Ovarian Neoplasms , Peritoneal Neoplasms , Humans , Female , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/metabolism , Animals , Carcinoma, Ovarian Epithelial/pathology , Carcinoma, Ovarian Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/etiology , Neoplasm MetastasisABSTRACT
BACKGROUND: Thymic epithelial tumours (TETs) are rare tumours comprised of thymomas and thymic carcinoma. Novel therapies are needed, especially in thymic carcinoma where the 5-year survival rate hovers at 30%. Mesothelin (MSLN), a surface glycoprotein that is cleaved to produce mature MSLN (mMSLN) and megakaryocyte potentiating factor (MPF), is expressed in limited tissues. However, its expression is present in various cancers, including thymic carcinoma, where it is expressed in 79% of cases. METHODS: We utilised flow cytometry, in vitro cytotoxicity assays, and an in vivo xenograft model in order to demonstrate the ability of the MSLN targeting antibody-drug conjugate (ADC) anetumab ravtansine (ARav) in inhibiting the growth of thymic carcinoma. RESULTS: Thymoma and thymic carcinoma cell lines express MSLN, and anetumab, the antibody moiety of ARav, was capable of binding MSLN expressing thymic carcinoma cells and internalising. ARav was effective at inhibiting the growth of thymic carcinoma cells stably transfected with mMSLN in vitro. In vivo, 15 mg/kg ARav inhibited T1889 xenograft tumour growth, while combining 7.5 mg/kg ARav with 4 mg/kg cisplatin yielded an additive effect on inhibiting tumour growth. CONCLUSIONS: These data demonstrate that anetumab ravtansine inhibits the growth of MSLN positive thymic carcinoma cells in vitro and in vivo.
Subject(s)
Immunoconjugates/administration & dosage , Maytansine/analogs & derivatives , Mesothelin/genetics , Mesothelin/metabolism , Neoplasms, Glandular and Epithelial/drug therapy , Thymoma/drug therapy , Thymus Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/administration & dosage , Cisplatin/pharmacology , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , HT29 Cells , Humans , Immunoconjugates/pharmacology , Maytansine/administration & dosage , Maytansine/pharmacology , Mice , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/metabolism , Thymoma/genetics , Thymoma/metabolism , Thymus Neoplasms/genetics , Thymus Neoplasms/metabolism , Up-Regulation/drug effects , Xenograft Model Antitumor AssaysABSTRACT
The Hedgehog (Hh) morphogen regulates growth and patterning. Since Hh signaling is also implicated in carcinogenesis, it is conceivable that de novo Hh-secreting organizers, if formed in association with oncogenic hit could be tumor-cooperative. Here we validate this hypothesis using the Drosophila model of cooperative epithelial carcinogenesis. We generate somatic clones with simultaneous loss of tumor suppressor, Lgl, and gain of the posterior compartment selector, Engrailed (En), known to induce synthesis of Hh. We show that lgl UAS-en clones in the anterior wing compartment trigger Hh signaling cascade via cross-talk with their Ci-expressing wild type cell neighbors. Hh-Dpp signaling from clone boundaries of such ectopically formed de novo organizers in turn drive lgl carcinogenesis. By contrast, Ci-expressing lgl clones transform by autocrine and/or juxtracine activation of Hh signaling in only the posterior compartment. We further show that sequestration of the Hh ligand or loss of Dpp receptor, Tkv, in these Hh-sending or -receiving lgl clones arrested their carcinogenesis. Our results therefore reveal a hitherto unrecognized mechanism of tumor cooperation by developmental organizers, which are induced fortuitously by oncogenic hits.
Subject(s)
Drosophila Proteins/metabolism , Hedgehog Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Hedgehog Proteins/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/geneticsABSTRACT
Proteins involved in the organizing of lipid rafts can be found in exosomes, as shown for caveolin-1, and they could contribute to exosomal cargo sorting, as shown for flotillins. Stomatin belongs to the same stomatin/prohibitin/flotillin/HflK/C family of lipid rafts proteins, but it has never been studied in exosomes except for extracellular vesicles (EVs) originating from blood cells. Here we first show the presence of stomatin in exosomes produced by epithelial cancer cells (non-small cell lung cancer, breast, and ovarian cancer cells) as well as in EVs from biological fluids, including blood plasma, ascitic fluids, and uterine flushings. A high abundance of stomatin in EVs of various origins and its enrichment in exosomes make stomatin a promising exosomal marker. Comparison with other lipid raft proteins and exosomal markers showed that the level of stomatin protein in exosomes from different sources corresponds well to that of CD9, while it differs essentially from flotillin-1 and flotillin-2 homologs, which in turn are present in exosomes in nearly equal proportions. In contrast, the level of vesicular caveolin-1 as well as its EV-to-cellular ratio vary drastically depending on cell type.
Subject(s)
Biomarkers, Tumor/metabolism , Caveolin 1/metabolism , Exosomes/metabolism , Membrane Proteins/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ascitic Fluid/chemistry , Body Fluids/chemistry , Female , Humans , Neoplasms, Glandular and Epithelial/metabolism , Uterus/chemistryABSTRACT
PURPOSE: To evaluate the distribution of the PAX8 transcription factor protein in ocular tissues and to investigate if immunohistochemical stains for this biomarker are useful in the diagnosis of intraocular tumors. DESIGN: Observational case series. PARTICIPANTS: Excision and cytologic analysis specimens of 6 ciliary body epithelial neoplasms, 2 iris epithelial neoplasms, 3 retinal pigment epithelial neoplasms, 3 intraocular medulloepitheliomas, 15 uveal melanomas, and 5 uveal melanocytomas. METHODS: Hematoxylin-eosin and PAX8 immunohistochemical stains were performed on all specimens. In appropriate cases, bleached preparations and other immunohistochemical stains, including AE1/AE3 cytokeratin, Lin28A, and CD45, were performed. MAIN OUTCOME MEASURES: Distribution of PAX8 expression in normal and neoplastic tissue. RESULTS: Strong nuclear PAX8 expression was observed in the normal corneal epithelium, iris sphincter pupillae muscle, iris pigment epithelium and dilator muscle complex, nonpigmented and pigmented epithelia of the ciliary body, lens epithelium, and a subset of retinal neurons. The normal retinal pigment epithelium and uveal melanocytes did not stain for PAX8. The ciliary body epithelial and neuroepithelial tumors (adenoma, adenocarcinoma, and medulloepithelioma) showed uniform strong nuclear PAX8 immunoreactivity. All melanocytic tumors (iris melanoma, ciliary-choroidal melanoma, and melanocytoma) and retinal pigment epithelial neoplasms showed negative results for PAX8. A subset of tumor-associated lymphocytes, most prominent in uveal melanoma, showed positive results for PAX8. The uniformity of the PAX8 staining was superior to the variable cytokeratin staining in the ciliary epithelial neoplasms and the variable Lin28A staining in malignant medulloepithelioma. The veracity of PAX8 staining was equally as robust on cytologic analysis and open-flap biopsy specimens of ciliary epithelial and iris epithelial neoplasms, melanocytoma, and melanoma. CONCLUSIONS: PAX8 has proven to be a very useful diagnostic marker in a select group of adult intraocular tumors, and we highly recommend its inclusion in diagnostic antibody panels of morphologically challenging intraocular neoplasms.
Subject(s)
Biomarkers, Tumor/metabolism , Eye Neoplasms/diagnosis , Eye Neoplasms/metabolism , PAX8 Transcription Factor/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Ciliary Body/metabolism , Ciliary Body/pathology , Female , Humans , Immunohistochemistry , Iris Neoplasms/diagnosis , Iris Neoplasms/metabolism , Keratins/metabolism , Leukocyte Common Antigens/metabolism , Male , Melanoma/diagnosis , Melanoma/metabolism , Middle Aged , Neoplasms, Glandular and Epithelial/diagnosis , Neoplasms, Glandular and Epithelial/metabolism , RNA-Binding Proteins/metabolism , Retinal Neoplasms/diagnosis , Retinal Neoplasms/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Uveal Neoplasms/diagnosis , Uveal Neoplasms/metabolismABSTRACT
Large-scale genomic studies have shown that half of epithelial ovarian cancers (EOCs) have alterations in genes regulating homologous recombination (HR) repair. Loss of HR accounts for the genomic instability of EOCs and for their cellular hyper-dependence on alternative poly-ADP ribose polymerase (PARP)-mediated DNA repair mechanisms. Previous studies have implicated the DNA polymerase θ (Polθ also known as POLQ, encoded by POLQ) in a pathway required for the repair of DNA double-strand breaks, referred to as the error-prone microhomology-mediated end-joining (MMEJ) pathway. Whether Polθ interacts with canonical DNA repair pathways to prevent genomic instability remains unknown. Here we report an inverse correlation between HR activity and Polθ expression in EOCs. Knockdown of Polθ in HR-proficient cells upregulates HR activity and RAD51 nucleofilament assembly, while knockdown of Polθ in HR-deficient EOCs enhances cell death. Consistent with these results, genetic inactivation of an HR gene (Fancd2) and Polq in mice results in embryonic lethality. Moreover, Polθ contains RAD51 binding motifs and it blocks RAD51-mediated recombination. Our results reveal a synthetic lethal relationship between the HR pathway and Polθ-mediated repair in EOCs, and identify Polθ as a novel druggable target for cancer therapy.
Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA-Directed DNA Polymerase/metabolism , Homologous Recombination , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Amino Acid Motifs , Animals , Carcinoma, Ovarian Epithelial , Cell Cycle , Cell Death , Cell Line, Tumor , DNA End-Joining Repair/genetics , DNA Replication , DNA-Directed DNA Polymerase/deficiency , Embryo Loss , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Genomic Instability , Homologous Recombination/genetics , Humans , Mice , Molecular Targeted Therapy , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Protein Binding , Rad51 Recombinase/antagonists & inhibitors , Rad51 Recombinase/metabolism , Recombinational DNA Repair/genetics , DNA Polymerase thetaABSTRACT
Cancer testis antigens (CTAs) are detected in cancer cells but not in healthy normal tissues, with the exception of gametogenic tissues. However, to our knowledge, expression of the antigens in thymic epithelial tumors has not been examined yet. We examined the immunohistochemical expression of five CTAs (MAGE-A, NY-ESO-1, MAGE-C1, SAGE and GAGE7) in 192 cases of thymic epithelial tumor. The CTAs were variably expressed in the thymic epithelial tumors. Type B component of type AB thymomas, type B1/B2/B3 thymomas, and thymic carcinomas showed a generally positive correlation between the malignancy grades and positive expression rates in four CTAs other than MAGE-C1. In thymic squamous cell carcinomas (SqCCs), four antigens except for MAGE-C1 showed high expression rates ranging from 23.1% to 43.6%. In the prognostic analysis, a positive expression of SAGE (P = 0.0485) and GAGE7 (P = 0.0289) were associated with a shorter overall survival in type B2/B3 thymomas, respectively. In thymic SqCC, a positive MAGE-A expression was significantly associated with an increased level of programmed death ligand in tumor-infiltrating lymphocytes (P = 0.0181). We showed (i) a frequent CTA expression, (ii) a general correlation of CTA expression with tumor malignancy grades and (iii) a prognostic impact in some of the CTAs.
Subject(s)
Antigens, Neoplasm/metabolism , Neoplasms, Glandular and Epithelial , Thymus Neoplasms , Adult , Aged , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Prognosis , Testis/metabolism , Thymoma/metabolism , Thymoma/pathology , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathologyABSTRACT
The current statistics on cancer show that 90% of all human cancers originate from epithelial cells. Breast and prostate cancer are examples of common tumors of epithelial origin that would benefit from improved drug treatment strategies. About 90% of preclinically approved drugs fail in clinical trials, partially due to the use of too simplified in vitro models and a lack of mimicking the tumor microenvironment in drug efficacy testing. This review focuses on the origin and mechanism of epithelial cancers, followed by experimental models designed to recapitulate the epithelial cancer structure and microenvironment, such as 2D and 3D cell culture models and animal models. A specific focus is put on novel technologies for cell culture of spheroids, organoids, and 3D-printed tissue-like models utilizing biomaterials of natural or synthetic origins. Further emphasis is laid on high-content imaging technologies that are used in the field to visualize in vitro models and their morphology. The associated technological advancements and challenges are also discussed. Finally, the review gives an insight into the potential of exploiting nanotechnological approaches in epithelial cancer research both as tools in tumor modeling and how they can be utilized for the development of nanotherapeutics.
Subject(s)
Bioprinting , Breast Neoplasms , Models, Biological , Neoplasms, Glandular and Epithelial , Organoids , Printing, Three-Dimensional , Prostatic Neoplasms , Animals , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Male , Nanotechnology , Neoplasms, Glandular and Epithelial/diagnostic imaging , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Organoids/diagnostic imaging , Organoids/metabolism , Organoids/pathology , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Tissue EngineeringABSTRACT
AIM: To describe four novel primary epithelial tumours of the sella with papillary architecture and Thyroid Transcription Factor 1 (TTF-1) expression. METHODS: Paraffin-embedded tissue from the four cases and recurrence of patient 1 was investigated with haematoxylin-eosin, special histochemical stains, immunohistochemistry with a broad panel of antibodies and next-generation sequencing. The ultrastructure of one tumour was studied in tissue retrieved from paraffin. RESULTS: The lesions occurred in three females aged 20, 26 and 42 years and a male aged 49 years. They presented with signs and symptoms secondary to pituitary stalk compression. Preoperative neuroimaging documented mixed solid and cystic, enhancing sellar masses with suprasellar extension. Histologically, the tumours showed thin papillae lined by a single layer of cytokeratin and TTF-1-positive cuboidal and cylindrical cells with mildly atypical nucleus. Next-generation sequencing performed in three cases did not identify any mutations. The main differential diagnosis included metastasis from lung or thyroid carcinoma, extraventricular choroid plexus papilloma and sellar ependymoma. CONCLUSION: We suggest the descriptive term of primary papillary epithelial tumour of the sella (PPETS) for this entity and propose that it could represent the intracranial equivalent of thyroid-like low-grade nasopharyngeal papillary adenocarcinoma. The cell of origin of PPETS remains undetermined although the intense and ubiquitous expression of TTF-1 may suggest a derivation from the infundibulum or ventricular recess. Our study expands the spectrum of sellar TTF-1-positive tumour and challenges the view that they all derive from pituicytes.
Subject(s)
Carcinoma, Papillary/pathology , Pituitary Neoplasms/pathology , Thyroid Nuclear Factor 1/metabolism , Adult , Biomarkers, Tumor/metabolism , Carcinoma, Papillary/metabolism , Female , Humans , Male , Middle Aged , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Pituitary Neoplasms/metabolism , Young AdultABSTRACT
OBJECTIVES: To understand underlying changes in pretreatment serum inflammatory markers associated with thymic epithelial tumors (TETs) development. METHODS: A retrospective analysis of 113 TETs patients who underwent 18-fluorine fluorodeoxyglucose (18F-FDG) positron emission tomography combined computed tomography (PET/CT) one to two weeks before tumor resection or biopsy was performed. Pretreatment serum neutrophil, monocyte, platelet, and lymphocyte counts, and fibrinogen and C-reaction protein (CRP) concentrations were measured one day before surgery or biopsy. Neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR), and platelet-to-lymphocyte ratio (PLR) were calculated by dividing corresponding cells counts by lymphocyte counts, respectively. The maximum standard uptake value (SUVmax) of 18F-FDG of primary TETs was applied to reflect tumor glycolytic activity. The student's t-test, one-way ANOVA analysis, Chi-square test, receiver operating characteristic curve analysis, and Logistic regression analysis were used for statistical analysis. RESULTS: The serum NLR and MLR were significantly higher in TETs patients than in healthy volunteers (P both ≤ 0.001). High serum NLR and MLR were related to the thymic carcinomas (TCs) subtype, elevated Masaoka-Koga (M-K) tumor stage, and metastasis of TETs (P all < 0.005). High serum NLR and MLR were also associated with high SUVmax values of TETs (P all < 0.005), with increasingly differences between groups as the cut-off values defining low-SUVmax and high-SUVmax groups increased. With the medium cutoff of NLR, MLR, and SUVmax of 3.07, 0.25, and 8.00 respectively, the high NLR and MLR levels were significantly associated with high SUVmax level of TETs (P both < 0.005). Moreover, the incidences of co-high SUVmax/NLR and co-high SUVmax/MLR were higher in TETs patients older than 55 years, with TCs, in M-K stage IV, and with metastasis (P all < 0.05). Both the co-high SUVmax/NLR and co-high SUVmax/MLR increased the risk of TETs metastasis (P both < 0.001), while the co-high SUVmax/MLR was also an independent risk factor for TETs metastasis (odds ratio: 3.92, 95% confidence interval: 1.02-15.12, P = 0.047). CONCLUSION: Pretreatment serum NLR and MLR of TETs patients are two tumor-progression- and tumor-glycolysis-related inflammatory markers. Enhanced tumor glycolytic activity and associated systemic inflammatory reaction may play a synergistic role in TETs metastasis.
Subject(s)
Biomarkers, Tumor/metabolism , Inflammation/pathology , Lymphocytes/pathology , Monocytes/pathology , Neoplasms, Glandular and Epithelial/pathology , Neutrophils/pathology , Thymus Neoplasms/pathology , Adult , Aged , Blood Platelets/metabolism , Blood Platelets/pathology , C-Reactive Protein/metabolism , Case-Control Studies , Female , Humans , Inflammation/metabolism , Leukocyte Count/methods , Lymphocyte Count/methods , Lymphocytes/metabolism , Male , Middle Aged , Monocytes/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Neutrophils/metabolism , Positron Emission Tomography Computed Tomography/methods , ROC Curve , Retrospective Studies , Thymus Neoplasms/metabolism , Young AdultABSTRACT
AIMS: Special AT-rich sequence-binding protein 2 (SATB2) is a transcriptional regulator with critical roles in brain, craniofacial and skeletal development. It has emerged as a key marker of lower gastrointestinal (GI) tract columnar epithelial and osteoblastic differentiation. Transcription factor immunohistochemistry is useful in assigning site of origin in well-differentiated neuroendocrine tumours (NETs), and has had a limited role in poorly differentiated neuroendocrine carcinomas (NECs). This study sought to evaluate the role of SATB2 in assigning site of origin in neuroendocrine epithelial neoplasms. METHODS AND RESULTS: Tissue microarrays were constructed from the following: 317 NETs (37 thyroid, 46 lung, 16 stomach, 12 duodenum, 70 pancreas, 106 jejunoileum, 24 appendix, and six rectosigmoid), 44 phaeochromocytomas/paragangliomas, and 79 NECs (29 Merkel cell, 30 lung, and 20 extrapulmonary visceral); nine appendiceal and 19 rectal NETs were examined in whole sections. SATB2 immunohistochemistry was scored for extent (%) and intensity (0-3+), with an H-score being calculated. SATB2 was expressed by 96% of rectosigmoid NETs, 79% of appendiceal NETs, and only 7% of other well-differentiated neoplasms (P < 0.0001). Expression in lower GI tract NETs (median H-score of 255) was stronger than in other positive tumours (median H-score of 7) (P < 0.0001). Any SATB2 expression was 86% sensitive/93% specific for lower GI tract origin. SATB2 was expressed by 79% of Merkel cell carcinomas (median H-score of 300), 33% of lung NECs (median H-score of 23), and 60% of extrapulmonary visceral NECs (median H-score of 110), with stronger expression in Merkel cell carcinoma (P < 0.001). At an H-score cutoff of ≥150, SATB2 was 69% sensitive/90% specific for Merkel cell carcinoma. CONCLUSIONS: SATB2 is frequently and strongly expressed by lower GI tract NETs; we have adopted it as our rectal NET marker. Relatively frequent and strong expression in Merkel cell carcinoma may have value in assigning NEC site of origin.
Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Merkel Cell/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Neuroendocrine Tumors/metabolism , Rectal Neoplasms/metabolism , Transcription Factors/metabolism , Carcinoma, Merkel Cell/diagnosis , Carcinoma, Merkel Cell/pathology , Carcinoma, Neuroendocrine/diagnosis , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Cell Differentiation , Cohort Studies , Diagnosis, Differential , Humans , Immunohistochemistry , Iowa , Lower Gastrointestinal Tract/metabolism , Lower Gastrointestinal Tract/pathology , Neoplasms, Glandular and Epithelial/diagnosis , Neoplasms, Glandular and Epithelial/pathology , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/pathology , Rectal Neoplasms/diagnosis , Rectal Neoplasms/pathology , Tissue Array AnalysisABSTRACT
Epithelial ovarian cancer (EOC) is a deadly cancer, and its prognosis has not been changed significantly during several decades. To seek new therapeutic targets for EOC, we performed an in vivo dropout screen in human tumor xenografts using a pooled shRNA library targeting thousands of druggable genes. Then, in follow-up studies, we performed a second screen using a genome-wide CRISPR/Cas9 library. These screens identified 10 high-confidence drug targets that included well-known oncogenes such as ERBB2 and RAF1, and novel oncogenes, notably KPNB1, which we investigated further. Genetic and pharmacological inhibition showed that KPNB1 exerts its antitumor effects through multiphase cell cycle arrest and apoptosis induction. Mechanistically, proteomic studies revealed that KPNB1 acts as a master regulator of cell cycle-related proteins, including p21, p27, and APC/C. Clinically, EOC patients with higher expression levels of KPNB1 showed earlier recurrence and worse prognosis than those with lower expression levels of KPNB1. Interestingly, ivermectin, a Food and Drug Administration-approved antiparasitic drug, showed KPNB1-dependent antitumor effects on EOC, serving as an alternative therapeutic toward EOC patients through drug repositioning. Last, we found that the combination of ivermectin and paclitaxel produces a stronger antitumor effect on EOC both in vitro and in vivo than either drug alone. Our studies have thus identified a combinatorial therapy for EOC, in addition to a plethora of potential drug targets.
Subject(s)
Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , beta Karyopherins/genetics , beta Karyopherins/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Ovarian Epithelial , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor/methods , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Ivermectin/pharmacology , Loss of Function Mutation/genetics , Neoplasms, Glandular and Epithelial/metabolism , Oncogenes , Ovarian Neoplasms/metabolism , Paclitaxel/pharmacology , RNA, Small Interfering/geneticsABSTRACT
Immunotherapies, particularly checkpoint inhibitors, have set off a revolution in cancer therapy by releasing the power of the immune system. However, only little is known about the antigens that are essentially presented on cancer cells, capable of exposing them to immune cells. Large-scale HLA ligandome analysis has enabled us to exhaustively characterize the immunopeptidomic landscape of epithelial ovarian cancers (EOCs). Additional comparative profiling with the immunopeptidome of a variety of benign sources has unveiled a multitude of ovarian cancer antigens (MUC16, MSLN, LGALS1, IDO1, KLK10) to be presented by HLA class I and class II molecules exclusively on ovarian cancer cells. Most strikingly, ligands derived from mucin 16 and mesothelin, a molecular axis of prognostic importance in EOC, are prominent in a majority of patients. Differential gene-expression analysis has allowed us to confirm the relevance of these targets for EOC and further provided important insights into the relationship between gene transcript levels and HLA ligand presentation.
Subject(s)
Antigen Presentation/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , CA-125 Antigen/immunology , Carcinoma, Ovarian Epithelial , Female , GPI-Linked Proteins/immunology , Galectin 1/immunology , Gene Expression Regulation, Neoplastic , HLA-DR Antigens/immunology , HLA-DR Antigens/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Immunotherapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Kallikreins/immunology , Ligands , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Membrane Proteins/immunology , Mesothelin , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , VaccinationABSTRACT
AIMS: Low-grade inflammation, measured by elevated plasma concentrations of high-sensitive C-reactive protein (CRP), is a risk factor for cardiovascular disease (CVD). There is evidence that low-grade inflammation is also related to a higher risk of cancer. The present prospective cohort study evaluates the relation between low-grade systemic inflammation and risk of cancer in patients with stable CVD. METHODS AND RESULTS: In total, 7178 patients with stable CVD and plasma CRP levels ≤10 mg/L were included. Data were linked to the Dutch national cancer registry. Cox regression models were fitted to study the relation between CRP and incident CVD and cancer. After a median follow-up time of 8.3 years (interquartile range 4.6-12.3) 1072 incident cancer diagnoses were observed. C-reactive protein concentration was related to total cancer [hazard ratio (HR) 1.35; 95% confidence interval (CI) 1.10-1.65] comparing last quintile to first quintile of CRP. Especially lung cancer, independent of histopathological subtype, was related to CRP (HR 3.39; 95% CI 2.02-5.69 comparing last to first quintile of CRP). Incidence of epithelial neoplasms and especially squamous cell neoplasms were related to CRP concentration, irrespective of anatomical location. Sensitivity analyses after excluding patients with a cancer diagnosis within 1, 2, and 5 years of follow-up showed similar results. No effect modification was observed by smoking status or time since smoking cessation (P-values for interaction > 0.05). CONCLUSION: Chronic systemic low-grade inflammation, measured by CRP levels ≤10 mg/L, is a risk factor for incident cancer, markedly lung cancer, in patients with stable CVD. The relation between inflammation and incident cancer is seen in former and current smokers and is uncertain in never smokers.
Subject(s)
Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Inflammation/complications , Neoplasms/etiology , Aged , C-Reactive Protein/metabolism , Case-Control Studies , Humans , Incidence , Inflammation/blood , Lung Neoplasms/epidemiology , Lung Neoplasms/pathology , Middle Aged , Neoplasms/epidemiology , Neoplasms, Glandular and Epithelial/epidemiology , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Neoplasms, Squamous Cell/epidemiology , Neoplasms, Squamous Cell/metabolism , Neoplasms, Squamous Cell/pathology , Netherlands/epidemiology , Proportional Hazards Models , Prospective Studies , Risk Factors , Smoking/epidemiologyABSTRACT
Epithelial ovarian cancer (EOC) frequently metastasises to the omentum, a process that requires pro-angiogenic activation of human omental microvascular endothelial cells (HOMECs) by tumour-secreted factors. We have previously shown that ovarian cancer cells secrete a range of factors that induce pro-angiogenic responses e.g. migration, in HOMECs including the lysosomal protease cathepsin D (CathD). However, the cellular mechanism by which CathD induces these cellular responses is not understood. The aim of this study was to further examine the pro-angiogenic effects of CathD in HOMECs i.e. proliferation and migration, to investigate whether these effects are dependent on CathD catalytic activity and to delineate the intracellular signalling kinases activated by CathD. We report, for the first time, that CathD significantly increases HOMEC proliferation and migration via a non-proteolytic mechanism resulting in activation of ERK1/2 and AKT. These data suggest that EOC cancer secreted CathD acts as an extracellular ligand and may play an important pro-angiogenic, and thus pro-metastatic, role by activating the omental microvasculature during EOC metastasis to the omentum.
Subject(s)
Cathepsin D/physiology , Cell Movement/genetics , Cell Proliferation/genetics , Endothelial Cells/physiology , Omentum/cytology , Carcinoma, Ovarian Epithelial , Cathepsin D/genetics , Cells, Cultured , Endothelial Cells/cytology , Female , Humans , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Metastasis , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Omentum/blood supply , Omentum/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/secondary , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiologyABSTRACT
Submucosal glands (SMGs) present throughout human esophagus with clusters at either the upper third or lower third of the organ. SMGs tend to atrophy with age, and neoplasms arising in these glands are rare. In order to bring convenience to diagnosis, we summarize the histopathologic characteristics of all esophageal submucosal gland tumors (SGTs). Due to the morphological similarity, the nomenclature of salivary tumors is adopted for SGTs. However, there is great confusion about the definition and histogenesis of these tumors, especially the malignant subtypes. In the literature, esophageal mucoepidermoid carcinoma and adenoid cystic carcinoma usually adjoin the surface squamous epithelium and coexist with intraepithelial neoplasia or invasive squamous cell carcinoma (SCC). In addition, the typical gene alterations of salivary tumors have not been reported in these SGTs. Therefore, we propose to apply stringent diagnostic criteria to esophageal SGTs so as to exclude mimickers that are SCCs with various degree of SMG differentiation.
Subject(s)
Esophageal Neoplasms/pathology , Esophagus/pathology , Neoplasms, Glandular and Epithelial/pathology , Aged, 80 and over , Atrophy/pathology , Carcinoma in Situ/pathology , Carcinoma, Adenoid Cystic/diagnosis , Carcinoma, Mucoepidermoid/diagnosis , Carcinoma, Squamous Cell/diagnosis , Humans , Keratins/metabolism , Male , Mucin-5B/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Retrospective StudiesABSTRACT
Epithelial ovarian cancer (EOC) is the fifth leading cause of cancer deaths among women in the United States, but its pathogenesis is poorly understood. Some epithelial cancers are known to occur in transitional zones between two types of epithelium, whereas others have been shown to originate in epithelial tissue stem cells. The stem cell niche of the ovarian surface epithelium (OSE), which is ruptured and regenerates during ovulation, has not yet been defined unequivocally. Here we identify the hilum region of the mouse ovary, the transitional (or junction) area between the OSE, mesothelium and tubal (oviductal) epithelium, as a previously unrecognized stem cell niche of the OSE. We find that cells of the hilum OSE are cycling slowly and express stem and/or progenitor cell markers ALDH1, LGR5, LEF1, CD133 and CK6B. These cells display long-term stem cell properties ex vivo and in vivo, as shown by our serial sphere generation and long-term lineage-tracing assays. Importantly, the hilum cells show increased transformation potential after inactivation of tumour suppressor genes Trp53 and Rb1, whose pathways are altered frequently in the most aggressive and common type of human EOC, high-grade serous adenocarcinoma. Our study supports experimentally the idea that susceptibility of transitional zones to malignant transformation may be explained by the presence of stem cell niches in those areas. Identification of a stem cell niche for the OSE may have important implications for understanding EOC pathogenesis.
Subject(s)
Epithelium/pathology , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Ovary/pathology , Stem Cell Niche , Animals , Biomarkers/analysis , Biomarkers/metabolism , Carcinoma, Ovarian Epithelial , Cell Lineage , Cell Separation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Epithelium/metabolism , Female , Male , Mice , Neoplasms, Glandular and Epithelial/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/metabolism , Ovary/metabolism , Receptors, G-Protein-Coupled/metabolism , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolismABSTRACT
The crosstalk between cancer cells and host cells is a crucial prerequisite for tumor growth and progression. The cells from both the innate and adaptive immune systems enter into a perverse relationship with tumor cells to create a tumor-promoting and immunosuppressive tumor microenvironment (TME). Epithelial ovarian cancer (EOC), the most lethal of all gynecological malignancies, is characterized by a unique TME that paves the way to the formation of metastasis and mediates therapy resistance through the deregulation of immune surveillance. A characteristic feature of the ovarian cancer TME is the ascites/peritoneal fluid, a malignancy-associated effusion occurring at more advanced stages, which enables the peritoneal dissemination of tumor cells and the formation of metastasis. The standard therapy for EOC involves a combination of debulking surgery and platinum-based chemotherapy. However, most patients experience disease recurrence. New therapeutic strategies are needed to improve the prognosis of patients with advanced EOC. Harnessing the body's natural immune defenses against cancer in the form of immunotherapy is emerging as an innovative treatment strategy. NK cells have attracted attention as a promising cancer immunotherapeutic target due to their ability to kill malignant cells and avoid healthy cells. Here, we will discuss the recent advances in the clinical application of NK cell immunotherapy in EOC.
Subject(s)
Immunomodulation , Immunotherapy , Killer Cells, Natural/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Biomarkers, Tumor , Combined Modality Therapy , Female , Humans , Immunomodulation/drug effects , Killer Cells, Natural/metabolism , Molecular Targeted Therapy , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Neoplasms, Glandular and Epithelial/therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Tumor Escape/immunologyABSTRACT
Rac GTPase activating protein 1 (RacGAP1) can regulate cytokinesis and cell differentiation. The oncogenic role of RacGAP1 has been partially studied in gastric cancer, colorectal cancer, and breast cancer. In the present study, we endeavor to evaluate its expression and functions in epithelial ovarian cancer (EOC). We retrospectively collected the clinicopathological information of 117 patients who underwent curative surgery for EOC. Expression of RacGAP1 protein in primary tumor tissues was evaluated by immunohistochemistry, which was significantly associated with tumor pathological grade, tumor stage, and lymph node metastasis. Patients with lower RacGAP1 level had a longer survival time and lower recurrence risk. Multivariate results identified the independent prognostic role of RacGAP1 for both recurrence and survival in EOC patients. Cellular studies showed that RacGAP1 can positively regulate the activation of RhoA and Erk proteins. In addition, wound healing assay and Transwell assay found that RacGAP1 can up-regulate the migration and invasion process of EOC cells, respectively. In all, our results not only confirmed the prognostic role of RacGAP1 for recurrence and survival in EOC patients, but also highlighted its possible potency for drug development.
Subject(s)
GTPase-Activating Proteins/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Proliferation , Disease Progression , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Neoplasm Grading , Neoplasm Invasiveness , Prognosis , Retrospective Studies , Survival Analysis , rhoA GTP-Binding Protein/metabolismABSTRACT
The ovary is surrounded by a whitish layer of mesodermally derived ovarian surface epithelium (OSE) that lines the intraembryonic celom and comprises simple squamous to cuboidal to low pseudostratified columnar epithelial cells. Its integrity is maintained by simple desmosomes, incomplete tight junctions, several integrins and cadherins. Recent research has found that ovarian stem cells (OSCs) exist within the OSE and may be responsible for both neo-oogenesis and ovarian cancer during adult life. The factors determining whether OSCs undergo neo-oogenesis or ovarian cancer are of great interest to researchers and clinicians. Accumulating evidence suggests the mechanism for the decision of ovarian surface epithelial stem cells to undergo either neo-oogenesis or ovarian cancer transformation may comprise both internal and external factors. Here, we review recent progress on how the internal factors, including genes, signaling pathways and lncRNA: OSE stem cells mediate the development and progression of ovarian cancer through various genes such as p53, KRAS, BRAF, and PTEN, and mutations in PIK3CA, and through various signaling pathways, including TGF-B pathway, Wnt signaling pathway, Notch signaling pathway, NF-kB signal transducer and transcriptional activator 3 (STAT3) pathway and Hedghog (HH) pathway. A series of expressions of IncRNA have changed in epithelial ovarian cancer tissues and cell lines compared to normal ovarian tissues and cell lines. As well as external factors, including incessant ovulation, gonadotropin and chronicinflammation: Frequent ovulation, without long-term dormancy, increases the risk of illness, because repeated rupture and repair at the ovulation site provides an opportunity for the accumulation of genetic aberrations; FSH affects all aspects of ovarian cancer metastasis, such as inhibition of apoptosis, through Induction of increased expression of VEGFA (VEGF) to support tumor growth, promote vascular growth, and possibly alter certain oncogenic pathways, thereby promoting proliferation and invasive phenotypic inflammation contributes to tumorigenesis, which help determine whether OSCs undergo neo-oogenesis or ovarian tumorigenesis. Understanding this issue is critical for developing novel strategies for premature ovarian failure and ovarian cancer prevention and therapy.