Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters

Publication year range
1.
Carcinogenesis ; 41(9): 1246-1252, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32525968

ABSTRACT

Castration-resistant prostate cancer (CRPC) is an incurable stage of the disease. A multivariate principal component analysis on CRPC in vitro models identified aspartyl (asparaginyl) ß hydrolase (ASPH) as the most relevant molecule associated with the CRPC phenotype. ASPH is overexpressed in various malignant neoplasms and catalyzes the hydroxylation of aspartyl and asparaginyl residues in the epidermal growth factor (EGF)-like domains of proteins like NOTCH receptors and ligands, enhancing cell motility, invasion and metastatic spread. Bioinformatics analyses of ASPH in prostate cancer (PCa) and CRPC datasets indicate that ASPH gene alterations have prognostic value both in PCa and CRPC patients. In CRPC cells, inhibition of ASPH expression obtained through specific small interfering RNA or culturing cells in hypoxic conditions, reduced cell proliferation, invasion and cyclin D1 expression through modulation of the NOTCH signaling. ASPH and HIF1α crosstalk, within a hydroxylation-regulated signaling pathway, might be transiently driven by the oxidative stress evidenced inside CRPC cells. In addition, increased phosphorylation of GSK3ß by ASPH silencing demonstrates that ASPH regulates GSK3ß activity inhibiting its interactions with upstream kinases. These findings demonstrate the critical involvement of ASPH in CRPC development and may represent an attractive molecular target for therapy.


Subject(s)
Biomarkers, Tumor/metabolism , Calcium-Binding Proteins/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 beta/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Proteins/antagonists & inhibitors , Mixed Function Oxygenases/antagonists & inhibitors , Muscle Proteins/antagonists & inhibitors , Prostatic Neoplasms, Castration-Resistant/pathology , Receptor, Notch1/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Proliferation , Glycogen Synthase Kinase 3 beta/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Prognosis , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , RNA, Small Interfering/genetics , Receptor, Notch1/genetics , Survival Rate , Tumor Cells, Cultured
3.
Int J Mol Sci ; 20(23)2019 Dec 03.
Article in English | MEDLINE | ID: mdl-31816863

ABSTRACT

BACKGROUND: Prostate cancer (PCa) is a significant health concern throughout the world. Standard therapy for advanced disease consists of anti-androgens, however, almost all prostate tumors become castration resistant (CRPC). Progression from androgen-sensitive PCa to CRPC is promoted by inflammatory signaling through cyclooxygenase-2 (COX-2) expression and ErbB family receptors/AKT activation, compensating androgen receptor inactivity. METHODS: Making use of CRPC cell lines, we investigated the effects of the anti-inflammatory drug celecoxib. Biochemical data obtained using immunoblotting, enzyme-linked immunosorbent assay (ELISA), invasion, and xenografts were further integrated by bioinformatic analyses. RESULTS: Celecoxib reduced cell growth and induced apoptosis through AKT blockade, cleavage of poly (ADP-ribose) polymerase-1 (PARP-1), and proteasomal degradation of the anti-apoptotic protein Mcl-1. Epidermal growth factor receptor (EGFR), ErbB2, and ErbB3 degradation, and heterogeneous nuclear ribonucleoprotein K (hnRNP K) downregulation, further amplified the inhibition of androgen signaling. Celecoxib reduced the invasive phenotype of CRPC cells by modulating NF-κB activity and reduced tumor growth in mice xenografts when administered in association with the anti-EGFR receptor antibody cetuximab. Bioinformatic analyses on human prostate cancer datasets support the relevance of these pathways in PCa progression. CONCLUSIONS: Signaling nodes at the intersection of pathways implicated in PCa progression are simultaneously modulated by celecoxib treatment. In combination therapies with cetuximab, celecoxib could represent a novel therapeutic strategy to curb signal transduction during CRPC progression.


Subject(s)
Celecoxib/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Signal Transduction , Amphiregulin/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Celecoxib/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cetuximab/pharmacology , Cetuximab/therapeutic use , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Epidermal Growth Factor/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Humans , Male , Mice, SCID , NF-kappa B/metabolism , Neoplasm Invasiveness , Phosphorylation/drug effects , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Cytotherapy ; 18(3): 438-51, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26857233

ABSTRACT

BACKGROUND AIMS: The amniotic fluid is a new source of multipotent stem cells with therapeutic potential for human diseases. In agreement with the regulatory requirement to reduce and possibly to avoid animal-derived reagents in the culture of cells intended for cell therapy, bovine serum, the most common supplement in the culture medium, was replaced by human platelet-derived growth factors. METHODS: We tested a new culture medium to expand monolayers of human amniotic fluid stem cells (hAFSC) for clinical use. The AFSC were isolated by c-Kit selection and expanded in media supplemented with either bovine serum or a human platelet lysate (Lyset). RESULTS: We compared proliferation kinetics, colony-forming unit percentage, multilineage differentiation, immunophenotypic characterization and inhibition of peripheral blood mononuclear cell proliferation of the two AFSC cell cultures and we found no significant differences. Moreover, the karyotype analysis of the cells expanded in the presence of the platelet lysate did not present cytogenetic abnormalities and in vitro and in vivo studies revealed no cell tumorigenicity. CONCLUSIONS: Platelet derivatives represent a rich source of growth factors that can play a safety role in the homeostasis, proliferation and remodeling of tissue healing. We propose human platelet extracts as a preferential alternative to animal serum for the expansion of stem cells for clinical applications.


Subject(s)
Amniotic Fluid/cytology , Cell Culture Techniques/methods , Cell Proliferation , Cell- and Tissue-Based Therapy , Stem Cells/cytology , Animals , Cattle , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Culture Media/metabolism , Culture Media/pharmacology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/physiology , Humans , Leukocytes, Mononuclear/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Multipotent Stem Cells/cytology , Multipotent Stem Cells/physiology , Stem Cells/drug effects
6.
Haematologica ; 99(6): 1078-87, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24561793

ABSTRACT

Hepatocyte growth factor, produced by stromal and follicular dendritic cells, and present at high concentrations in the sera of patients with chronic lymphocytic leukemia, prolongs the survival of leukemic B cells by interacting with their receptor, c-MET. It is, however, unknown whether hepatocyte growth factor influences microenvironmental cells, such as nurse-like cells, which deliver survival signals to the leukemic clone. We evaluated the expression of c-MET on nurse-like cells and monocytes from patients with chronic lymphocytic leukemia and searched for phenotypic/functional features supposed to be influenced by the hepatocyte growth factor/c-MET interaction. c-MET is expressed at high levels on nurse-like cells and at significantly higher levels than normal on monocytes from patients. Moreover, the hepatocyte growth factor/c-MET interaction activates STAT3(TYR705) phosphorylation in nurse-like cells. Indoleamine 2,3-dioxygenase, an enzyme modulating T-cell proliferation and induced on normal monocytes after hepatocyte growth factor treatment, was detected together with interleukin-10 on nurse-like cells, and on freshly-prepared patients' monocytes. Immunohistochemical/immunostaining analyses demonstrated the presence of c-MET(+) and indoleamine 2,3-dioxygenase(+) cells in lymph node biopsies, co-expressed with CD68 and vimentin. Furthermore nurse-like cells and chronic lymphocytic monocytes significantly inhibited T-cell proliferation, prevented by anti-transforming growth factor beta and interleukin-10 antibodies and indoleamine 2,3-dioxygenase inhibitors, and supported CD4(+)CD25(high+)/FOXP3(+) T regulatory cell expansion. We suggest that nurse-like cells display features of immunosuppressive type 2 macrophages: higher hepatocyte growth factor levels, produced by leukemic or other microenvironmental surrounding cells, may cooperate to induce M2 polarization. Hepatocyte growth factor may thus have a dual pathophysiological role: directly through enhancement of survival of the leukemic clone and indirectly by favoring T-cell immunosuppression.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Macrophages/immunology , Macrophages/metabolism , Proto-Oncogene Proteins c-met/genetics , Cells, Cultured , Coculture Techniques , Gene Expression , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/pharmacology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-met/metabolism , STAT3 Transcription Factor/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
7.
Cells ; 13(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38786084

ABSTRACT

Relevant advances have been made in the management of relapsed/refractory (r/r) Hodgkin Lymphomas (HL) with the use of the anti-CD30 antibody-drug conjugate (ADC) brentuximab-vedotin (Bre-Ved). Unfortunately, most patients eventually progress despite the excellent response rates and tolerability. In this report, we describe an ADC composed of the aminobisphosphonate zoledronic acid (ZA) conjugated to Bre-Ved by binding the free amino groups of this antibody with the phosphoric group of ZA. Liquid chromatography-mass spectrometry, inductively coupled plasma-mass spectrometry, and matrix-assisted laser desorption ionization-mass spectrometry analyses confirmed the covalent linkage between the antibody and ZA. The novel ADC has been tested for its reactivity with the HL/CD30+ lymphoblastoid cell lines (KMH2, L428, L540, HS445, and RPMI6666), showing a better titration than native Bre-Ved. Once the HL-cells are entered, the ADC co-localizes with the lysosomal LAMP1 in the intracellular vesicles. Also, this ADC exerted a stronger anti-proliferative and pro-apoptotic (about one log fold) effect on HL-cell proliferation compared to the native antibody Bre-Ved. Eventually, Bre-Ved-ZA ADC, in contrast with the native antibody, can trigger the proliferation and activation of cytolytic activity of effector-memory Vδ2 T-lymphocytes against HL-cell lines. These findings may support the potential use of this ADC in the management of r/r HL.


Subject(s)
Brentuximab Vedotin , Immunoconjugates , Ki-1 Antigen , Zoledronic Acid , Humans , Zoledronic Acid/pharmacology , Zoledronic Acid/therapeutic use , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Immunoconjugates/chemistry , Brentuximab Vedotin/pharmacology , Brentuximab Vedotin/therapeutic use , Ki-1 Antigen/metabolism , Ki-1 Antigen/immunology , Cell Line, Tumor , Hodgkin Disease/drug therapy , Hodgkin Disease/pathology , Hodgkin Disease/immunology , Apoptosis/drug effects , Cell Proliferation/drug effects
8.
Cancers (Basel) ; 16(7)2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38610932

ABSTRACT

Antibody--drug conjugates (ADCs) are a promising delivery system that involves linking a monoclonal antibody (mAb) to a specific drug, such as a cytotoxic agent, to target tumor cells. This new class of antitumor therapy acts as a "biological missile" that can destroy tumor cells while increasing the therapeutic index and decreasing toxicity. One of the most critical factors in ADC design is selecting a target antigen that is highly expressed on the surface of cancer cells. In this study, we conjugated Cetuximab (Cet), a monoclonal antibody that targets the epidermal growth factor receptor (EGFR), to aminobisphosphonates (N-BPs) such as ibandronate (IBA) or risedronate (RIS) or zoledronate (ZA). Cetuximab is administered to patients with metastatic colorectal carcinoma (mCRC) with a wild-type (WT) EGFR transduction pathway. Also, it is well established that N-BPs can trigger the antitumor activity of Vδ2 T cells in both in vitro and in vivo experimental models. The resulting ADCs were added in co-culture to assess the effect on CRC cell line proliferation and sensitivity to Vδ2 T antitumor lymphocytes in comparison with the native antibody. These assays have been performed both in conventional and 3D spheroid cultures. We found that all three ADCs can increase the inhibitory effect on cell proliferation of the WT-EGFR cell line Caco-2 while only Cet-RIS and Cet-ZA can increase the cytotoxicity mediated by Vδ2 T cells against both WT and EGFR-mutated CRC cell lines (Caco-2, DLD-1, and HCT-116). Also, the ADCs can trigger the cell proliferation of Vδ2 T cells present in peripheral blood and tumor specimens. Our findings indicate that anti-EGFR antibodies bound to N-BPs can improve the antitumor effects of the native antibody possibly increasing the therapeutic effect.

9.
Prostate ; 73(9): 941-51, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23334998

ABSTRACT

BACKGROUND: The role of CXCL1 in prostate cancer (PCa) progression has been poorly investigated. A limitation of previous studies is linked to the use of human PCa cell lines PC3 and DU145, producing CXCL8 at levels strongly exceeding CXCL1 levels. Moreover, in mouse models the sharing of CXCR2 receptor by both ligands makes the phenotype induced by CXCL8 and CXCL1 almost indistinguishable. To overcome this problem we used the murine TRAMP-C2 cell line, not expressing CXCL8 and expressing CXCL1 at low levels. METHODS: The effect of CXCL1 overexpression was examined by in vivo subcutaneous tumor studies and in vitro functional assays of invasion and adhesion. Biochemical modifications were evaluated by Western blotting and antibody arrays. RESULTS: Our data show that the overexpression of CXCL1 in TRAMP-C2 cells represses tumor establishment and in situ invasion. In vitro, the main action of CXCL1 expression in TRAMP cells is associated with the perturbation of molecules linked to cell adhesion and migration thus explaining in vivo data. Other in vitro findings also suggest that signaling by CXCL1 might activate a secretory network limiting in vivo tumor growth by reinforcing senescence. Immunohistochemical staining of human PCa, BPH, and normal prostate biopsies strengthen our observations on the mouse model: when expressed, CXCL1 is limited to small areas with faint staining and PCa progression does not rely on CXCL1 expression. CONCLUSION: We could speculate that CXCL1 overexpression acts as a suppressor of malignancy limiting the escape of tumor cells from the primary tumor and reinforcing growth arrest.


Subject(s)
Chemokine CXCL1/biosynthesis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Cell Growth Processes/physiology , Cellular Senescence/physiology , Chemokine CXCL1/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Prostatic Neoplasms/genetics , Tissue Array Analysis
10.
Cancers (Basel) ; 15(3)2023 Jan 28.
Article in English | MEDLINE | ID: mdl-36765755

ABSTRACT

About 15% of colorectal cancers (CRCs) are diagnosed as advanced, metastatic stage IV, a patient condition with an average survival of 2 [...].

11.
Cancers (Basel) ; 15(3)2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36765873

ABSTRACT

Colorectal Cancer (CRC) is one of most frequent malignant cancers, showing high lethality worldwide [...].

12.
Cancers (Basel) ; 15(3)2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36765872

ABSTRACT

Immune checkpoint (IC) molecules act as receptors, expressed on immune effector cells, that are able to recognize specific ligands in normal or tumor cells [...].

13.
Cancers (Basel) ; 15(2)2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36672440

ABSTRACT

Fibroblasts are incredible cells [...].

14.
Cancers (Basel) ; 15(2)2023 Jan 14.
Article in English | MEDLINE | ID: mdl-36672464

ABSTRACT

The identification and validation of simple, reliable and reproducible three dimensional (3D) in vitro culture systems represent a major challenge in the field of anticancer drug development [...].

15.
Cells ; 12(4)2023 02 20.
Article in English | MEDLINE | ID: mdl-36831331

ABSTRACT

The p38 inhibitor SB202190 is a necessary component of the medium used for normal colorectal mucosa cultures. Sato et al. suggested that the primary activity of SB202190 may be EGFR signaling stabilization, causing an increased phosphorylation of Erk1-2 sustaining organoid proliferation. However, the growth of some colorectal cancer (CRC)-derived organoid cultures is inhibited by this molecule via an unknown mechanism. We biochemically investigated SB202190 activity on a collection of 25 primary human CRC organoids, evaluating EGFR, Akt and Erk1-2 activation using Western blot. We found that Erk1-2 phosphorylation was induced by SB202190 in 20 organoid cultures and inhibited in 5 organoid cultures. A next-generation sequencing (NGS) analysis revealed that the inhibition of p-Erk1-2 signaling corresponded to the cultures with BRAF mutations (with four different hits, one being undescribed), while p-Erk1-2 induction was apparently unrelated to other mutations involving the EGFR pathway (Her2, KRAS and NRAS). We found that SB202190 mirrored the biochemical activity of the BRAF inhibitor Dabrafenib, known to induce the paradoxical activation of p-Erk1-2 signaling in BRAF wild-type cells. SB202190 was a more effective inhibitor of BRAF-mutated organoid growth in the long term than the specific BRAF inhibitors Dabrafenib and PLX8394. Overall, SB202190 can predict BRAF-activating mutations in patient-derived organoids, as well as allowing for the identification of new BRAF variants, preceding and enforcing NGS data.


Subject(s)
Colorectal Neoplasms , Proto-Oncogene Proteins B-raf , Humans , Proto-Oncogene Proteins B-raf/genetics , Protein Kinase Inhibitors/pharmacology , Mutation , Colorectal Neoplasms/genetics , ErbB Receptors/genetics , Organoids/metabolism
16.
Cancers (Basel) ; 15(3)2023 Jan 18.
Article in English | MEDLINE | ID: mdl-36765569

ABSTRACT

Tumor-associated fibroblasts (TAF) exert immunosuppressive effects in colorectal carcinoma (CRC), impairing the recognition of tumor cells by effector lymphocytes, including Vδ2 T cells. Herein, we show that CRC-derived TAF can be turned by zoledronic acid (ZA), in soluble form or as antibody-drug conjugate (ADC), into efficient stimulators of Vδ2 T cells. CRC-TAF, obtained from patients, express the epidermal growth factor receptor (EGFR) and the butyrophilin family members BTN3A1/BTN2A1. These butyrophilins mediate the presentation of the phosphoantigens, accumulated in the cells due to ZA effect, to Vδ2 T cells. CRC-TAF exposed to soluble ZA acquired the ability to trigger the proliferation of Vδ2 T cells, in part represented by effector memory cells lacking CD45RA and CD27. In turn, expanded Vδ2 T cells exerted relevant cytotoxic activity towards CRC cells and CRC-TAF when primed with soluble ZA. Of note, also the ADC made of the anti-EGFR cetuximab (Cet) and ZA (Cet-ZA), that we recently described, induced the proliferation of anti-tumor Vδ2 T lymphocytes and their activation against CRC-TAF. These findings indicate that ZA can educate TAF to stimulate effector memory Vδ2 T cells; the Cet-ZA ADC formulation can lead to the precise delivery of ZA to EGFR+ cells, with a double targeting of TAF and tumor cells.

17.
Nat Commun ; 14(1): 6951, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37907500

ABSTRACT

Identifying oncological applications for drugs that are already approved for other medical indications is considered a possible solution for the increasing costs of cancer treatment. Under the hypothesis that nutritional stress through fasting might enhance the antitumour properties of at least some non-oncological agents, by screening drug libraries, we find that cholesterol biosynthesis inhibitors (CBIs), including simvastatin, have increased activity against cancers of different histology under fasting conditions. We show fasting's ability to increase CBIs' antitumour effects to depend on the reduction in circulating insulin, insulin-like growth factor-1 and leptin, which blunts the expression of enzymes from the cholesterol biosynthesis pathway and enhances cholesterol efflux from cancer cells. Ultimately, low cholesterol levels through combined fasting and CBIs reduce AKT and STAT3 activity, oxidative phosphorylation and energy stores in the tumour. Our results support further studies of CBIs in combination with fasting-based dietary regimens in cancer treatment and highlight the value of fasting for drug repurposing in oncology.


Subject(s)
Fasting , Simvastatin , Simvastatin/pharmacology , Simvastatin/therapeutic use , Diet , Insulin , Cholesterol
18.
Int J Cancer ; 131(3): 582-90, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22015967

ABSTRACT

Current diagnostic tools cannot predict clinical failure and androgen-independent disease progression for patients with prostate cancer (PC). The survival signaling pathways of prostate cells play a central role in the progression of tumors to a neuroendocrine (NE) phenotype. NE cells demonstrate attributes that suggest that they are an integral part of the signaling cascade leading to castration-resistant PC. In this study, making use of in vitro neuroendocrine differentiation (NED) of human LNCaP and mouse TRAMP-C2 cells after androgen withdrawal, and of the transgenic adenocarcinoma of mouse prostate (TRAMP) model, we characterized a sequence of molecular events leading to NED and identified a number of markers that could be detectable by routine analyses not only in castration resistant PC but also in hormone naïve PC at the time of initial diagnosis. We found that NED associates with AKT activation that in turn regulates heterogeneous nuclear ribonucleoprotein K (hnRNP K), androgen receptor (AR) and ß-catenin levels. Addition of molecules targeting membrane-bound receptors and protein kinases blocks NE differentiation in LNCaP and TRAMP-C2 cells. The extent of AKT phosphorylation and hnRNP K, AR and ß-catenin levels may have a potential value as prognostic indicators discriminating between androgen-responsive and unresponsive cells and could be used as molecular targets to monitor the anti-tumor action of new therapeutic protocols based on antireceptor agents and/or neuroendocrine hormone antagonists.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Neuroendocrine Cells/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Signal Transduction , Androgens/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroendocrine Cells/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Androgen/metabolism , beta Catenin/metabolism
19.
Mol Med ; 18: 1292-302, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-22952060

ABSTRACT

Despite recent advances in understanding the biological basis of prostate cancer, management of the disease, especially in the phase resistant to androgen ablation, remains a significant challenge. The long latency and high incidence of prostate carcinogenesis provides the opportunity to intervene with chemoprevention to prevent or eradicate prostate malignancies. In this study, we have used human hormone-resistant prostate cancer cells, DU145 and PC3, as an in vitro model to assess the efficacy of xanthohumol (XN) against cell growth, motility and invasion. We observed that treatment of prostate cancer cells with low micromolar doses of XN inhibits proliferation and modulates focal adhesion kinase (FAK) and AKT phosphorylation leading to reduced cell migration and invasion. Oxidative stress by increased production of reactive oxygen species (ROS) was associated with these effects. Transgenic adenocarcinoma of the mouse prostate (TRAMP) transgenic mice were used as an in vivo model of prostate adenocarcinoma. Oral gavage of XN, three times per week, beginning at 4 wks of age, induced a decrease in the average weight of the urogenital (UG) tract, delayed advanced tumor progression and inhibited the growth of poorly differentiated prostate carcinoma. The ability of XN to inhibit prostate cancer in vitro and in vivo suggests that XN may be a novel agent for the management of prostate cancer.


Subject(s)
Disease Progression , Flavonoids/pharmacology , Flavonoids/therapeutic use , Propiophenones/pharmacology , Propiophenones/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Administration, Oral , Androgens/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Flavonoids/administration & dosage , G1 Phase/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Invasiveness , Neoplasm Staging , Propiophenones/administration & dosage , Reactive Oxygen Species/metabolism , Resting Phase, Cell Cycle/drug effects
20.
Cancers (Basel) ; 14(24)2022 Dec 10.
Article in English | MEDLINE | ID: mdl-36551571

ABSTRACT

Colorectal cancer (CRC) is a relatively slow-growing tumor that can be treated successfully when identified in the early stages [...].

SELECTION OF CITATIONS
SEARCH DETAIL