289 results on '"Festuccia, Claudio"'
Search Results
252. Crocetin and Crocin from Saffron in Cancer Chemotherapy and Chemoprevention.
- Author
-
Colapietro A, Mancini A, D'Alessandro AM, and Festuccia C
- Subjects
- Animals, Chemoprevention, Humans, Neoplasms metabolism, Neoplasms pathology, Vitamin A analogs & derivatives, Antineoplastic Agents, Phytogenic pharmacology, Antioxidants pharmacology, Carotenoids pharmacology, Crocus chemistry, Neoplasms drug therapy
- Abstract
Introduction: Cancer is a disorder which has a powerful impact on the quality life and life expectancy despite the increase in drugs and treatments available for cancer patients. Moreover, many new therapeutic options are known to have adverse reactions without any improvement in outcome than before. Nowadays, natural products or plant derivatives are used as chemoprevention drugs and chemotherapy is the new approach that uses specific cell premalignant transformation in the malignant form. Natural substances derived from plants, such as polyphenols, flavonoids, carotenoids, alkaloids and others, can be biologically active and have a wide spectrum of effects. The protective effects of Saffron carotenoids (crocin and crocetin) have been extensively studied mainly for their antioxidant properties, however, they have various other biological activities including tumor growth inhibition with the induction of cell death., Methods: The relevant information on Saffron and its carotenoids was collected from scientific databases (such as PubMed, Web of Science, Science Direct). To identify all published articles in relation to saffron, crocin and crocetin, in different types of cancer, no language restriction has been used., Results: To date, crossing the words saffron and cancer, approximately 150 articles can be found. If crossing is made between crocin and cancer, approximately 60 articles can be found. With the crossing between crocetin and cancer, the number is approximately 55, while between carotenoids and cancer, the number exceeds 16.000 reports. In all the papers published to date, there are evidences that saffron and its carotenoids exert chemopreventive activity through anti-oxidant activity, cancer cells apoptosis, inhibition of cell proliferation, enhancement of cell differentiation, modulation of cell cycle progression and cell growth, modulation of tumor metabolism, stimulation of cell-to-cell communication and immune modulation., Conclusion: Here, we have tried to offer an up-to-date overview of pre-clinical experimental investigations on the potential use of the main carotenoids of saffron in tumor models and focus the attention on the molecular mechanisms involved., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.net.)
- Published
- 2019
- Full Text
- View/download PDF
253. Immunolocalization of Advanced Glycation End Products, Mitogen Activated Protein Kinases, and Transforming Growth Factor-β/Smads in Pelvic Organ Prolapse.
- Author
-
Vetuschi A, Pompili S, Gallone A, D'Alfonso A, Carbone MG, Carta G, Festuccia C, Gaudio E, Colapietro A, and Sferra R
- Subjects
- Female, Humans, Immunohistochemistry, Glycation End Products, Advanced analysis, Mitogen-Activated Protein Kinases analysis, Pelvic Organ Prolapse pathology, Smad Proteins analysis, Transforming Growth Factor beta analysis
- Abstract
Collagen and matrix metalloproteinases (MMP) play a pivotal role in the pathophysiology of Pelvic Organ Prolapse (POP) as a switch between type I and III collagen together with a simultaneous activation of MMPs have been observed in the vaginal wall. The aim of this study was to evaluate the Advanced Glycation End (AGE) products, ERK1/2 and transforming growth factor (TGF)-β/Smad pathway expression in muscularis propria in women with POP compared with control patients. We examined 20 patients with POP and 10 control patients treated for uterine fibromatosis. Immunohistochemical analysis using AGE, RAGE, ERK1/2, Smads-2/3, Smad-7, MMP-3, and collagen I-III, TIMP, and α-SMA were performed. Smad-2/3, Smad-7, AGE, ERK1/2, p-ERK, and p-Smad3 were also evaluated using Western-blot analysis. POP samples from the anterior vaginal wall showed disorganization of the normal muscularis architecture. In POP samples, AGE, ERK1/2, Smad-2/3, MMP-3, and collagen III were upregulated in muscularis whereas in controls, Smad-7 and collagen I were increased. The receptor for AGEs (RAGE) was mild or absent both in controls and prolapse. We demonstrated the involvement of these markers in women with POP but further studies are required to elucidate if the overexpression of these molecules could play a crucial role in the pathophysiology of POP disease.
- Published
- 2018
- Full Text
- View/download PDF
254. UniPR1331, a small molecule targeting Eph/ephrin interaction, prolongs survival in glioblastoma and potentiates the effect of antiangiogenic therapy in mice.
- Author
-
Festuccia C, Gravina GL, Giorgio C, Mancini A, Pellegrini C, Colapietro A, Delle Monache S, Maturo MG, Sferra R, Chiodelli P, Rusnati M, Cantoni A, Castelli R, Vacondio F, Lodola A, and Tognolini M
- Abstract
Glioblastoma multiforme (GBM) is the most malignant brain tumor, showing high resistance to standard therapeutic approaches that combine surgery, radiotherapy, and chemotherapy. As opposed to healthy tissues, EphA2 has been found highly expressed in specimens of glioblastoma, and increased expression of EphA2 has been shown to correlate with poor survival rates. Accordingly, agents blocking Eph receptor activity could represent a new therapeutic approach. Herein, we demonstrate that UniPR1331, a pan Eph receptor antagonist, possesses significant in vivo anti-angiogenic and anti-vasculogenic properties which lead to a significant anti-tumor activity in xenograft and orthotopic models of GBM. UniPR1331 halved the final volume of tumors when tested in xenografts (p<0.01) and enhanced the disease-free survival of treated animals in the orthotopic models of GBM both by using U87MG cells (40 vs 24 days of control, p<0.05) or TPC8 cells (52 vs 16 days, p<0.01). Further, the association of UniPR1331 with the anti-VEGF antibody Bevacizumab significantly increased the efficacy of both monotherapies in all tested models. Overall, our data promote UniPR1331 as a novel tool for tackling GBM., Competing Interests: CONFLICTS OF INTEREST The authors declare no conflicts of interest.
- Published
- 2018
- Full Text
- View/download PDF
255. Dual PI3 K/mTOR inhibition reduces prostate cancer bone engraftment altering tumor-induced bone remodeling.
- Author
-
Mancini A, Colapietro A, Pompili S, Del Fattore A, Delle Monache S, Biordi LA, Angelucci A, Mattei V, Liang C, Gravina GL, and Festuccia C
- Subjects
- Animals, Bone Remodeling physiology, Cell Differentiation drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Disease-Free Survival, Humans, Male, Mice, Mice, Nude, Prostatic Neoplasms drug therapy, Proto-Oncogene Proteins c-akt biosynthesis, RAW 264.7 Cells, Xenograft Model Antitumor Assays, Antineoplastic Agents therapeutic use, Bone Neoplasms drug therapy, Bone Neoplasms prevention & control, Bone Neoplasms secondary, Organic Chemicals pharmacology, Phosphoinositide-3 Kinase Inhibitors, Prostatic Neoplasms pathology, Protein Kinase Inhibitors therapeutic use, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Morbidity in advanced prostate cancer patients is largely associated with bone metastatic events. The development of novel therapeutic strategies is imperative in order to effectively treat this incurable stage of the malignancy. In this context, Akt signaling pathway represents a promising therapeutic target able to counteract biochemical recurrence and metastatic progression in prostate cancer. We explored the therapeutic potential of a novel dual PI3 K/mTOR inhibitor, X480, to inhibit tumor growth and bone colonization using different in vivo prostate cancer models including the subcutaneous injection of aggressive and bone metastatic (PC3) and non-bone metastatic (22rv1) cell lines and preclinical models known to generate bone lesions. We observed that X480 both inhibited the primary growth of subcutaneous tumors generated by PC3 and 22rv1 cells and reduced bone spreading of PCb2, a high osteotropic PC3 cell derivative. In metastatic bone, X480 inhibited significantly the growth and osteolytic activity of PC3 cells as observed by intratibial injection model. X480 also increased the bone disease-free survival compared to untreated animals. In vitro experiments demonstrated that X480 was effective in counteracting osteoclastogenesis whereas it stimulated osteoblast activity. Our report provides novel information on the potential activity of PI3 K/Akt inhibitors on the formation and progression of prostate cancer bone metastases and supports a biological rationale for the use of these inhibitors in castrate-resistant prostate cancer patients at high risk of developing clinically evident bone lesions.
- Published
- 2018
- Full Text
- View/download PDF
256. The first-in-class alkylating deacetylase inhibitor molecule tinostamustine shows antitumor effects and is synergistic with radiotherapy in preclinical models of glioblastoma.
- Author
-
Festuccia C, Mancini A, Colapietro A, Gravina GL, Vitale F, Marampon F, Delle Monache S, Pompili S, Cristiano L, Vetuschi A, Tombolini V, Chen Y, and Mehrling T
- Subjects
- Animals, Antineoplastic Agents, Alkylating pharmacology, Bendamustine Hydrochloride pharmacology, Bendamustine Hydrochloride therapeutic use, Benzimidazoles pharmacology, Brain Neoplasms pathology, Cell Line, Tumor, Female, Glioblastoma pathology, Histone Deacetylase Inhibitors pharmacology, Humans, Mice, Vorinostat pharmacology, Vorinostat therapeutic use, Antineoplastic Agents, Alkylating therapeutic use, Benzimidazoles therapeutic use, Brain Neoplasms drug therapy, Brain Neoplasms radiotherapy, Glioblastoma drug therapy, Glioblastoma radiotherapy, Histone Deacetylase Inhibitors therapeutic use
- Abstract
Background: The use of alkylating agents such as temozolomide in association with radiotherapy (RT) is the therapeutic standard of glioblastoma (GBM). This regimen modestly prolongs overall survival, also if, in light of the still dismal prognosis, further improvements are desperately needed, especially in the patients with O6-methylguanine-DNA-methyltransferase (MGMT) unmethylated tumors, in which the benefit of standard treatment is less. Tinostamustine (EDO-S101) is a first-in-class alkylating deacetylase inhibitor (AK-DACi) molecule that fuses the DNA damaging effect of bendamustine with the fully functional pan-histone deacetylase (HDAC) inhibitor, vorinostat, in a completely new chemical entity., Methods: Tinostamustine has been tested in models of GBM by using 13 GBM cell lines and seven patient-derived GBM proliferating/stem cell lines in vitro. U87MG and U251MG (MGMT negative), as well as T98G (MGMT positive), were subcutaneously injected in nude mice, whereas luciferase positive U251MG cells and patient-derived GBM stem cell line (CSCs-5) were evaluated the orthotopic intra-brain in vivo experiments., Results: We demonstrated that tinostamustine possesses stronger antiproliferative and pro-apoptotic effects than those observed for vorinostat and bendamustine alone and similar to their combination and irrespective of MGMT expression. In addition, we observed a stronger radio-sensitization of single treatment and temozolomide used as control due to reduced expression and increased time of disappearance of γH2AX indicative of reduced signal and DNA repair. This was associated with higher caspase-3 activation and reduction of RT-mediated autophagy. In vivo, tinostamustine increased time-to-progression (TTP) and this was additive/synergistic to RT. Tinostamustine had significant therapeutic activity with suppression of tumor growth and prolongation of DFS (disease-free survival) and OS (overall survival) in orthotopic intra-brain models that was superior to bendamustine, RT and temozolomide and showing stronger radio sensitivity., Conclusions: Our data suggest that tinostamustine deserves further investigation in patients with glioblastoma.
- Published
- 2018
- Full Text
- View/download PDF
257. Episode-like pulse testosterone supplementation induces tumor senescence and growth arrest down-modulating androgen receptor through modulation of p-ERK1/2, pAR ser81 and CDK1 signaling: biological implications for men treated with testosterone replacement therapy.
- Author
-
Gravina GL, Marampon F, Sanità P, Festuccia C, Forcella C, Scarsella L, Jitariuc A, Vetuschi A, Sferra R, Colapietro A, Carosa E, Dolci S, Lenzi A, and Jannini EA
- Abstract
Despite the growing body of knowledge showing that testosterone (T) may not significantly affect tumor progression in hypogonadal patients treated for prostate cancer (Pca), the use of this hormone in this population still remains controversial. The effects of continuous or pulsed T stimulation were tested in vitro and in vivo on androgen-sensitive Pca cell lines in order to assess the differential biological properties of these two treatment modalities. Pulsed T treatment resulted in a greater inhibition than continuous T supplementation of tumor growth in vitro and in vivo . The effects of pulsed T treatment on tumor growth inhibition, G0/G1 cell cycle arrest, and tumor senescence was more pronounced than those obtained upon continuous T treatments. Mechanistic studies revealed that G0/G1 arrest and tumor senescence upon pulsed T treatment were associated with a marked decrease in cyclin D1, c-Myc and SKp2, CDK4 and p-Rb levels and upregulation of p27 and p-ERK1/2. Pulsed, but not continuous, T supplementation decreased the expression levels of AR, p-AR
ser81 and CDK1 in both cellular models. The in vitro results were confirmed in an in vivo xenografts, providing evidence of a greater inhibitory activity of pulsed supraphysiological T supplementation than continuous treatment, both in terms of tumor volume and decreased AR, p-ARser81 , PSA and CDK1 staining. The rapid cycling from hypogonadal to physiological or supra-physiological T intraprostatic concentrations results in cytostatic and senescence effects in preclinical models of androgen-sensitive Pca. Our preclinical evidence provides relevant new insights in the biology of Pca response to pulsed T supplementation., Competing Interests: CONFLICTS OF INTEREST The authors declare no conflicts of interest.- Published
- 2017
- Full Text
- View/download PDF
258. Pharmacological treatment with inhibitors of nuclear export enhances the antitumor activity of docetaxel in human prostate cancer.
- Author
-
Gravina GL, Mancini A, Colapietro A, Marampon F, Sferra R, Pompili S, Biordi LA, Iorio R, Flati V, Argueta C, Landesman Y, Kauffman M, Shacham S, and Festuccia C
- Abstract
Background and Aims: Docetaxel (DTX) modestly increases patient survival of metastatic castration-resistant prostate cancer (mCRPC) due to insurgence of pharmacological resistance. Deregulation of Chromosome Region Maintenance (CRM-1)/ exportin-1 (XPO-1)-mediated nuclear export may play a crucial role in this phenomenon., Material and Methods: Here, we evaluated the effects of two Selective Inhibitor of Nuclear Export (SINE) compounds, selinexor (KPT-330) and KPT-251, in association with DTX by using 22rv1, PC3 and DU145 cell lines with their. DTX resistant derivatives., Results and Conclusions: We show that DTX resistance may involve overexpression of β-III tubulin (TUBB3) and P-glycoprotein as well as increased cytoplasmic accumulation of Foxo3a. Increased levels of XPO-1 were also observed in DTX resistant cells suggesting that SINE compounds may modulate DTX effectiveness in sensitive cells as well as restore the sensitivity to DTX in resistant ones. Pretreatment with SINE compounds, indeed, sensitized to DTX through increased tumor shrinkage and apoptosis by preventing DTX-induced cell cycle arrest. Basally SINE compounds induce FOXO3a activation and nuclear accumulation increasing the expression of FOXO-responsive genes including p21, p27 and Bim causing cell cycle arrest. SINE compounds-catenin and survivin supporting apoptosis. βdown-regulated Cyclin D1, c-myc, Nuclear sequestration of p-Foxo3a was able to reduce ABCB1 and TUBB3 H2AX levels, prolonged γ expression. Selinexor treatment increased DTX-mediated double strand breaks (DSB), and reduced the levels of DNA repairing proteins including DNA PKc and Topo2A. Our results provide supportive evidence for the therapeutic use of SINE compounds in combination with DTX suggesting their clinical use in mCRPC patients., Competing Interests: CONFLICTS OF INTEREST Yosef Landesman, Christian Argueta, Michael G Kauffman and Sharon Shacham are employees of Karyopharm Therapeutics, Newton, MA, USA. Other authors declare that they have no competing interests.
- Published
- 2017
- Full Text
- View/download PDF
259. Editorial: New Therapeutic Approaches for the Treatment of Glioblastoma.
- Author
-
Festuccia C and Gravina GL
- Subjects
- Animals, Biomarkers, Tumor blood, Biomarkers, Tumor genetics, Brain Neoplasms blood, Brain Neoplasms genetics, Brain Neoplasms pathology, Genetic Predisposition to Disease, Glioblastoma blood, Glioblastoma genetics, Glioblastoma pathology, Humans, Prognosis, Brain Neoplasms therapy, Glioblastoma therapy
- Published
- 2017
- Full Text
- View/download PDF
260. Cyclin D1 silencing suppresses tumorigenicity, impairs DNA double strand break repair and thus radiosensitizes androgen-independent prostate cancer cells to DNA damage.
- Author
-
Marampon F, Gravina GL, Ju X, Vetuschi A, Sferra R, Casimiro MC, Pompili S, Festuccia C, Colapietro A, Gaudio E, Di Cesare E, Tombolini V, and Pestell RG
- Published
- 2016
- Full Text
- View/download PDF
261. Dual PI3K/mTOR inhibitor, XL765 (SAR245409), shows superior effects to sole PI3K [XL147 (SAR245408)] or mTOR [rapamycin] inhibition in prostate cancer cell models.
- Author
-
Gravina GL, Mancini A, Scarsella L, Colapietro A, Jitariuc A, Vitale F, Marampon F, Ricevuto E, and Festuccia C
- Subjects
- Apoptosis, Cell Cycle, Cell Line, Tumor drug effects, Cell Proliferation, Forkhead Box Protein O1 metabolism, Glycogen Synthase Kinase 3 beta metabolism, Humans, Inhibitory Concentration 50, Male, RNA, Small Interfering metabolism, Receptors, Androgen metabolism, Drug Resistance, Neoplasm, Phosphoinositide-3 Kinase Inhibitors, Prostatic Neoplasms metabolism, Quinoxalines chemistry, Sirolimus chemistry, Sulfonamides chemistry, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Deregulation of phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway contributes to prostate cancer development and progression. Here, we compared the in vitro effects of the dual PI3K/mTOR inhibitor (XL765) with those observed with the sole PI3K (XL147) or mTOR (rapamycin) inhibition in 2 non-tumor prostate epithelial cell lines, 8 prostate cancer cell lines, and 11 prostate cancer cell derivatives. We demonstrated that the XL765 treatment showed superior and proliferative effects of XL147 or rapamycin. The XL765 effects were associated to increasing the chromosome region maintenance 1 (CRM1)-mediated nuclear localization of glycogen synthase kinase 3 beta (GSK3β) and Foxo-1a with higher induction of apoptosis when compared to those observed in XL147 and rapamycin treatments. IC50 values were calculated in phosphatase and tensin homologue deleted on chromosome 10 (PTEN)-positive and PTEN-negative cell lines as well as after PTEN transfection or PTEN downmodulation by siRNA strategy revealing that the presence of this protein was associated with reduced sensitivity to PI3K and mTOR inhibitors. The comparison of IC50 values was also calculated for androgen-dependent and -independent cell lines as well as after androgen receptor (AR) transfection or the AR downmodulation by siRNA strategy revealing that androgen independence was associated with enhanced responsiveness. Our results provide a rationale to use the dual PI3K/Akt/mTOR inhibitors in hormone-insensitive prostate cancer models due to the overactivity of PI3K/Akt/mTOR in this disease condition.
- Published
- 2016
- Full Text
- View/download PDF
262. Editorial: Antitarget Therapies: New Frontiers in the Treatment of Cancer.
- Author
-
Festuccia C, Lodola A, and Gravina GL
- Subjects
- Cell Death drug effects, Cell Proliferation drug effects, Humans, Neoplasms pathology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Molecular Targeted Therapy, Neoplasms drug therapy, Neoplasms metabolism
- Published
- 2016
- Full Text
- View/download PDF
263. Editorial: New Drug Targets for Treatment of Recurrent/Metastatic Prostate Cancer.
- Author
-
Festuccia C, Negri-Cesi P, and Gravina GL
- Subjects
- Antineoplastic Agents pharmacology, Bone Neoplasms secondary, Drug Design, Drug Resistance, Neoplasm, Drug Screening Assays, Antitumor, Humans, Male, Tumor Microenvironment drug effects, Antineoplastic Agents therapeutic use, Bone Neoplasms drug therapy, Neoplasm Recurrence, Local drug therapy, Prostatic Neoplasms drug therapy
- Published
- 2016
- Full Text
- View/download PDF
264. CXCR4 pharmacogical inhibition reduces bone and soft tissue metastatic burden by affecting tumor growth and tumorigenic potential in prostate cancer preclinical models.
- Author
-
Gravina GL, Mancini A, Muzi P, Ventura L, Biordi L, Ricevuto E, Pompili S, Mattei C, Di Cesare E, Jannini EA, and Festuccia C
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antiviral Agents pharmacology, Benzylamines, Blotting, Western, Cell Adhesion, Cell Movement, Chemokine CXCL12 metabolism, Coculture Techniques, Cyclams, Disease Models, Animal, Enzyme-Linked Immunosorbent Assay, Heterografts, Humans, Immunohistochemistry, Lymph Nodes pathology, Lymphatic Metastasis, Male, Mice, Mice, Nude, Prostatic Neoplasms metabolism, Receptors, CXCR4 metabolism, Tomography, X-Ray Computed, Tumor Cells, Cultured, Vascular Endothelial Growth Factor A metabolism, Bone Neoplasms secondary, Heterocyclic Compounds pharmacology, Peptides pharmacology, Prostatic Neoplasms pathology, Receptors, CXCR4 antagonists & inhibitors
- Abstract
Background: The majority of prostate cancer (Pca) patient morbidity can be attributed to bone metastatic events, which poses a significant clinical obstacle. Therefore, a better understanding of this phenomenon is imperative and might help to develop novel therapeutic strategies. Stromal cell-derived factor 1α (SDF-1α) and its receptor CXCR4 have been implicated as regulators of bone resorption and bone metastatic development, suggesting that agents able to suppress this signaling pathway may be used as pharmacological treatments. In this study we studied if two CXCR4 receptor antagonists, Plerixafor and CTE9908, may affect bone metastatic disease induced by Pca in preclinical experimental models, Methods: To verify the hypothesis that CXCR4 inhibition affects Pca metastatic disease, selective CXCR4 compounds, Plerixafor, and CTE9908, were tested in preclinical models known to generate bone lesions. Additionally, the expression levels of CXCR4 and SDF-1α were analyzed in a number of human tissues derived from primary tumors, lymph-nodes and osseous metastases of Pca as well as in a wide panel of human Pca cell lines to non-tumorigenic and tumorigenic phenotype., Results: Bone-derived Pca cells express higher CXCR4 levels than other Pca cell lines. This differential expression was also observed in human Pca samples. In vitro evidence supports the hypothesis that factors produced by bone microenvironment differentially sustain CXCR4 and SDF1-α expression with respect to prostate microenvironment determining increased efficacy toward Plerixafor. The use of SDF1-α neutralizing antibodies greatly reduced the increase of CXCR4 expression in cells co-cultured with bone stromal cells (BMSc) and to a lesser extent in cells co-cultured with prostate stromal cells (HPSc) and partially reduced SDF1-α Plerixafor efficacy. SDF-1α induced tumor cell migration and invasion, as well as MMP-9, MMP-2, and uPA expression, which were reduced by Plerixafor. The incidence of X-ray detectable bone lesions was significantly reduced following Plerixafor and CTE9908 treatment Kaplan-Meier probability plots showed a significant improvement in the overall survival of mice treated with Plerixafor and CTE9908. The reduced intra-osseous growth of PC3 and PCb2 tumor cells after intratibial injection, as a result of Plerixafor and CTE9908 treatment, correlated with decreased osteolysis and serum levels of both mTRAP and type I collagen fragments (CTX), which were significantly lower with respect to controls., Conclusions: Our report provides novel information on the potential activity of CXCR4 inhibitors on the formation and progression of Pca bone and soft tissue metastases and supports a biological rationale for the use of these inhibitors in men at high risk to develop clinically evident bone lesions., (© 2015 Wiley Periodicals, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
265. SRC family kinase (SFK) inhibition reduces rhabdomyosarcoma cell growth in vitro and in vivo and triggers p38 MAP kinase-mediated differentiation.
- Author
-
Casini N, Forte IM, Mastrogiovanni G, Pentimalli F, Angelucci A, Festuccia C, Tomei V, Ceccherini E, Di Marzo D, Schenone S, Botta M, Giordano A, and Indovina P
- Subjects
- Animals, Apoptosis drug effects, Blotting, Western, Cell Differentiation physiology, Cell Movement drug effects, Cell Proliferation drug effects, Cell Survival drug effects, Fluorescent Antibody Technique, Humans, Mice, Polymerase Chain Reaction, Xenograft Model Antitumor Assays, p38 Mitogen-Activated Protein Kinases metabolism, Antineoplastic Agents pharmacology, Pyrazoles pharmacology, Pyrimidines pharmacology, Rhabdomyosarcoma pathology, src-Family Kinases antagonists & inhibitors
- Abstract
Recent data suggest that SRC family kinases (SFKs) could represent potential therapeutic targets for rhabdomyosarcoma (RMS), the most common soft-tissue sarcoma in children. Here, we assessed the effect of a recently developed selective SFK inhibitor (a pyrazolo[3,4-d]pyrimidine derivative, called SI221) on RMS cell lines. SI221, which showed to be mainly effective against the SFK member YES, significantly reduced cell viability and induced apoptosis, without affecting non-tumor cells, such as primary human skin fibroblasts and differentiated C2C12 cells. Moreover, SI221 decreased in vitro cell migration and invasion and reduced tumor growth in a RMS xenograft model. SFK inhibition also induced muscle differentiation in RMS cells by affecting the NOTCH3 receptor-p38 mitogen-activated protein kinase (MAPK) axis, which regulates the balance between proliferation and differentiation. Overall, our findings suggest that SFK inhibition, besides reducing RMS cell growth and invasive potential, could also represent a differentiation therapeutic strategy for RMS.
- Published
- 2015
- Full Text
- View/download PDF
266. Torc1/Torc2 inhibitor, Palomid 529, enhances radiation response modulating CRM1-mediated survivin function and delaying DNA repair in prostate cancer models.
- Author
-
Gravina GL, Marampon F, Sherris D, Vittorini F, Di Cesare E, Tombolini V, Lenzi A, Jannini EA, and Festuccia C
- Subjects
- Animals, Benzopyrans therapeutic use, Cell Line, Tumor, DNA Repair drug effects, DNA Repair physiology, Humans, Inhibitor of Apoptosis Proteins metabolism, Karyopherins metabolism, Male, Mechanistic Target of Rapamycin Complex 1, Mechanistic Target of Rapamycin Complex 2, Mice, Mice, Nude, Multiprotein Complexes metabolism, Prostatic Neoplasms drug therapy, Prostatic Neoplasms radiotherapy, Radiation-Sensitizing Agents therapeutic use, Receptors, Cytoplasmic and Nuclear metabolism, Survivin, TOR Serine-Threonine Kinases metabolism, Up-Regulation drug effects, Xenograft Model Antitumor Assays methods, Exportin 1 Protein, Benzopyrans pharmacology, Inhibitor of Apoptosis Proteins physiology, Karyopherins physiology, Multiprotein Complexes antagonists & inhibitors, Prostatic Neoplasms metabolism, Radiation-Sensitizing Agents pharmacology, Receptors, Cytoplasmic and Nuclear physiology, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Background: P529, a Torc1/Torc2 inhibitor, has demonstrated its potential as a radiosensitizer. However the molecular mechanisms underlying this phenomenon still need to be elucidated. Aim of this study is to dissect molecular mechanisms regulating the radiosensitizing properties of P529 in a wide panel of prostate cancer models., Methods: Six tumor cell lines and xenograft models were used for in vitro and in vivo studies. Clonogenic survival, apoptotic, autophagic, and senescence assays were used to examine the effects of ionizing radiation (IR) alone and in combination with P529. CRM1, survivin, GSK-3β, and DNA-DSBs expression and modulation, upon P529 and RT, were monitored by western blot. In vivo treatment response upon P529, irradiation or combination of P529 with IR was monitored by tumor volume, time to progression (TTP), and immunohistochemical analysis., Results: P529 treatment induced significantly more apoptosis and DNA double-strand break (DSB) when combined with radiotherapy resulting in cellular radiosensitization and growth delay of irradiated tumor xenografts. Upon P529 treatment Rad51, DNA-PKcs, and Ku70 protein expression was downregulated, indicating delayed DNA double-strand damage repair. The radiosensitizing properties of P529 were partially linked to GSK-3β, cyclin-D1, and c-myc modulation with associated inhibition of CRM1-mediated nuclear export of survivin. Importantly, autophagy and tumor senescence were involved in the enhanced P529 radioresponse., Conclusions: Impaired DNA double-strand damage repair, inhibition of CRM1-mediated nuclear export of survivin, modulation of cyclin-D1 and c-myc with associated pro-apoptotic and autophagic and senescent events explain the radiosensitizing properties of P529 in preclinical models of prostate cancer., (© 2014 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
267. Close correlation between MEK/ERK and Aurora-B signaling pathways in sustaining tumorigenic potential and radioresistance of gynecological cancer cell lines.
- Author
-
Marampon F, Gravina GL, Popov VM, Scarsella L, Festuccia C, La Verghetta ME, Parente S, Cerasani M, Bruera G, Ficorella C, Ricevuto E, Tombolini V, Di Cesare E, and Zani BM
- Subjects
- Butadienes pharmacology, Cell Line, Tumor, Cell Proliferation drug effects, DNA Repair drug effects, Female, Genital Neoplasms, Female pathology, Genital Neoplasms, Female radiotherapy, HeLa Cells, Humans, Nitriles pharmacology, Phosphorylation, Prognosis, Radiation Tolerance genetics, Signal Transduction drug effects, Aurora Kinase B genetics, Carcinogenesis, Genital Neoplasms, Female genetics, MAP Kinase Signaling System genetics
- Abstract
Both Aurora-A and -B kinases have been implicated in tumorigenesis; and as such, they represent an attractive therapeutic target. Recent studies found that Aurora-A is a downstream target of mitogen-activated protein kinase 1/ERK2, while Aurora-B has been found to be a prognostic/predictive therapeutic target for epithelial cancer. In a wide range of human cancers, the Ras/Raf/MEK/ERK/MAP kinase pathway is enhanced and the cellular response to growth signals is known to increase. The purpose of this study was to investigate whether the MEK/ERK cascade regulates tumorigenic signaling and radioresistance via the Aurora-B-mediated pathway in a panel of gynecological cancer cell lines. Exponentially growing human endometrial (Ishikawa), cervical (HeLa), cervical (CASKI) and vulva (SiHa) cancer cells were used in culture treated with either control or MEK/ERK inhibitor or AZD1152 before and after irradiation. Western blotting, ERK1/2 siRNA transfection, growth assay in modified monolayer, Annexin V and migration/invasion assays were performed. The specific MEK/ERK inhibitor U0126 decreased the tumorigenic potential and improved the radiation response in all cellular models. The modulation of radioresponse upon U0126 treatment positively correlated with the inhibition of phospho-ERKs and the reduction of Aurora-B kinase expression. In addition, upon U0126 treatment DNA-PKcs protein expression was found to be downregulated, indicating that the improved radiation response may be caused by decreased DNA double-strand damage repair mechanisms. The knockdown of ERK by siRNA confirmed the MEK/ERK-dependent Aurora-B kinase expression. The use of AZD1152, a selective Aurora-B inhibitor, counteracted tumorigenic potential and radioresistance phenotype by highly increasing apoptotic mechanisms in all gynecological cancer cell lines used. Evidence from our experiments show that tumorigenic potential and radiation response in gynecological cancer cells may ensue from a MEK/ERK or Aurora-B inhibition. Together with the close correlation of MEK/ERK and Aurora-B protein expression, this study underlines the potential role of a MEK/ERK/Aurora-B axis whose interruption recovers the antitumor effects of radiotherapy.
- Published
- 2014
- Full Text
- View/download PDF
268. Increased levels of DNA methyltransferases are associated with the tumorigenic capacity of prostate cancer cells.
- Author
-
Gravina GL, Ranieri G, Muzi P, Marampon F, Mancini A, Di Pasquale B, Di Clemente L, Dolo V, D'Alessandro AM, and Festuccia C
- Subjects
- Antimetabolites, Antineoplastic pharmacology, Azacitidine pharmacology, Cell Line, Tumor, DNA (Cytosine-5-)-Methyltransferase 1, DNA Methyltransferase 3A, Epithelial Cells enzymology, Gene Expression drug effects, Glutathione S-Transferase pi genetics, Glutathione S-Transferase pi metabolism, Humans, Male, Phenotype, Prostate enzymology, Prostatic Hyperplasia enzymology, DNA Methyltransferase 3B, Carcinogenesis metabolism, DNA (Cytosine-5-)-Methyltransferases metabolism, Prostatic Neoplasms enzymology
- Abstract
DNA methylation might be the earliest somatic genome changes in prostate cancer that also play an important role in the process of tumor invasion, growth and metastasis. In recent years, several inhibitors of DNA methyltransferases (DNMTis) have been developed and evaluated in pre-clinical models and in clinical trials. While these compounds are effective in the treatment of hematological conditions, clinical trials in solid tumors and in prostate cancer have shown limited or no efficacy. This may be attributed to inappropriate dose regimens leading to toxicity-related adverse events. As with other anti-target compounds, one of the obstacles encountered with DNMTis in prostate cancer could be the inability to select patients for the clinical studies as well as the inability to monitor the efficacy of the drug if not the conclusion of the study. Primary cultures derived from human prostatic tissues harvested from patients with benign prostatic hyperplasia (BPH) and prostate cancer (PCa) as well as neoplastic and non-neoplastic prostate cell lines were tested for DNMT expression/activity and to monitor azacitidine molecular efficacy. We observed that in primary cultures the levels of DNMT activity as well as the protein levels of DNMT1, DNMT3a and DNMT3b were higher in cultures derived from PCa compared to BPH tissue samples and significantly higher in cultures derived from PCa with Gleason scores ≥7 compared to those observed in cultures derived from Gleason scores <7. In addition, DNMT activity as well as DNMT1, DNMT3a and DNMT3b levels were higher in PCa cell lines compared to their non-neoplastic counterparts. Although DNMT activity was higher in high tumorigenic/aggressive PCa cell lines compared to low tumorigenic/aggressive cell lines, only the levels of DNMT3a and DNMT3b were significantly higher in the first group of cells, suggesting that DNMT1 activity is related to the transition to non-neoplastic versus neoplastic phenotype whereas the de novo methylation enzymes were mainly related to progression. Nevertheless, the comparison in the more aggressive PC3 cell derivatives (PC3-LN4 cells) also possessed higher levels of DNMT1 compared to PC3 and PC3M from which these cells were derived. Collectively, our results confirm previous data on the increased methylation in more aggressive tumors supporting the use of DNMTis in advanced prostate cancer. In addition, since glutathione S-transferase-π (GSTP1) was re-expressed or its protein levels were increased after treatment with non-toxic azacitidine doses and since GSTP1 can easily be measured in patient sera, the monitoring of this protein may aide in the evaluation of therapy in future clinical trials.
- Published
- 2013
- Full Text
- View/download PDF
269. Strategies for imaging androgen receptor signaling pathway in prostate cancer: implications for hormonal manipulation and radiation treatment.
- Author
-
Gravina GL, Festuccia C, Bonfili P, Di Staso M, Franzese P, Ruggieri V, Popov VM, Tombolini V, Masciocchi C, Carosa E, Lenzi A, Jannini EA, and Di Cesare E
- Subjects
- Antineoplastic Agents, Hormonal therapeutic use, Humans, Male, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Receptors, Androgen genetics, Receptors, Androgen therapeutic use, Signal Transduction genetics, Treatment Outcome, Molecular Imaging methods, Prostatic Neoplasms drug therapy, Prostatic Neoplasms radiotherapy, Translational Research, Biomedical
- Abstract
Prostate cancer (Pca) is a heterogeneous disease; its etiology appears to be related to genetic and epigenetic factors. Radiotherapy and hormone manipulation are effective treatments, but many tumors will progress despite these treatments. Molecular imaging provides novel opportunities for image-guided optimization and management of these treatment modalities. Here we reviewed the advances in targeted imaging of key biomarkers of androgen receptor signaling pathways. A computerized search was performed to identify all relevant studies in Medline up to 2013. There are well-known limitations and inaccuracies of current imaging approaches for monitoring biological changes governing tumor progression. The close integration of molecular biology and clinical imaging could ease the development of new molecular imaging agents providing novel tools to monitor a number of biological events that, until a few years ago, were studied by conventional molecular assays. Advances in translational research may represent the next step in improving the oncological outcome of men with Pca who remain at high risk for systemic failure. This aim may be obtained by combining the anatomical properties of conventional imaging modalities with biological information to better predict tumor response to conventional treatments.
- Published
- 2013
- Full Text
- View/download PDF
270. Differential effects of PXD101 (belinostat) on androgen-dependent and androgen-independent prostate cancer models.
- Author
-
Gravina GL, Marampon F, Giusti I, Carosa E, Di Sante S, Ricevuto E, Dolo V, Tombolini V, Jannini EA, and Festuccia C
- Subjects
- Acetylation drug effects, Animals, Apoptosis drug effects, Cell Cycle Checkpoints drug effects, Cell Division drug effects, Cell Line, Cell Line, Tumor, Cell Proliferation drug effects, Epithelial Cells drug effects, Epithelial Cells metabolism, G2 Phase drug effects, Gene Expression Regulation, Neoplastic drug effects, Histones metabolism, Humans, Male, Mice, Mice, Nude, Neoplasms, Hormone-Dependent metabolism, Prostatic Neoplasms metabolism, Receptors, Androgen metabolism, Sulfonamides, Transfection methods, Xenograft Model Antitumor Assays methods, Androgens metabolism, Histone Deacetylase Inhibitors pharmacology, Hydroxamic Acids pharmacology, Neoplasms, Hormone-Dependent drug therapy, Prostatic Neoplasms drug therapy
- Abstract
Histone deacetylase inhibitors (HDACi) are promising epigenetic cancer chemotherapeutics rapidly approaching clinical use. In this study, we tested using in vitro and in vivo models the differential biological effects of a novel HDAC inhibitor [belinostat (PXD101)], in a wide panel of androgen-sensitive and androgen-independent tumor cells. Belinostat significantly increased acetylation of histones H3 and H4. Belinostat potently inhibited the growth of prostate cancer cell lines (IC50 range from 0.5 to 2.5 µM) with cytotoxic activity preferentially against tumor cells. This agent induced G2/M arrest and increased significantly the percentage of apoptosis mainly in androgen-sensitive tumor cells confirming its growth-inhibitory effects. The cell death mechanisms were studied in three different prostate cancer cell lines with different androgen dependence and expression of androgen receptor; LAPC-4 and 22rv1 (androgen-dependent and expressing androgen receptor) and PC3 (androgen-independent not expressing androgen receptor). Belinostat induced the expression of p21 and p27, acetylation of p53 and G2/M arrest associated with Bcl2 and Bcl-Xl downmodulation and significant reduction of survivin, IAPs and Akt/pAkt and increased caspase-8 and -9 expression/activity. Belinostat effectiveness was dependent on the androgen receptor (AR), since the stable transfection of AR greatly increased the efficacy of this HDAC inhibitor. These observations were correlated using in vivo models. We demonstrated that belinostat preferentially resulted in antitumor effect in androgen-dependent tumor cells expressing AR. Our findings provide evidence that belinostat may be a promising anticancer drug for prostate cancer expressing AR, supporting its clinical role in prostate cancer.
- Published
- 2012
- Full Text
- View/download PDF
271. Hormonal therapy promotes hormone-resistant phenotype by increasing DNMT activity and expression in prostate cancer models.
- Author
-
Gravina GL, Marampon F, Piccolella M, Motta M, Ventura L, Pomante R, Popov VM, Zani BM, Pestell RG, Tombolini V, Jannini EA, and Festuccia C
- Subjects
- Anilides therapeutic use, Cell Line, DNA (Cytosine-5-)-Methyltransferases genetics, DNA Methyltransferase 3A, Drug Resistance, Epigenomics, Gene Expression Regulation, Neoplastic drug effects, Hormones therapeutic use, Humans, Male, Nitriles therapeutic use, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Tosyl Compounds therapeutic use, DNA Methyltransferase 3B, Anilides pharmacology, DNA (Cytosine-5-)-Methyltransferases metabolism, Hormones pharmacology, Nitriles pharmacology, Prostatic Neoplasms drug therapy, Tosyl Compounds pharmacology
- Abstract
We hypothesized that hormonal therapy favors the development of the hormone-resistant phenotype through epigenetic mechanisms. Human prostate cancer tissues and in vitro and in vivo models were used to verify this hypothesis. We demonstrated that tumor cells continuously treated with bicalutamide (BCLT) or cultured in androgen-depleted medium progressively acquire higher DNA methyltransferase (DNMT) activity and expression than cells cultured in standard condition. Increased DNMT expression and activity also paralleled the up-regulation of truncated AR isoforms, which favors the development of the hormone-resistant phenotype. After androgen stimulation with 10(-12) m dihydrotestosterone, DNMT activity was significantly reduced in comparison with hormonal therapy. Consistent with these observations, the silencing of DNMT3a and DNMT3b significantly decreased the DNMT activity levels. These findings were also directly correlated with phosphatase and tensin homolog down-regulation and activation of ERK and phosphatidylinositol 3-kinases/AKT8 virus oncogene cellular homolog pathways. The use of a pan-DNMT inhibitor (5-Azacitidine) greatly reduced the development of the hormone-resistant phenotype induced by long-term BCLT treatment, and this finding correlated with low DNMT activity. The regulation of DNMT activity was, in some measure, dependent on the androgen receptor, as small interfering RNA treatment targeting the androgen receptor greatly decreased the modulation of DNMT activity under androgenic and antiandrogenic stimulation. These observations were correlated in vivo in patients, as demonstrated by immunohistochemistry. Patients treated by BCLT before surgery had higher DNMT3a and DNMT3b expression than patients who had not undergone this treatment. Our findings provide evidence of a relationship between the castration-resistant phenotype and DNMT expression and activity in human prostate cancer.
- Published
- 2011
- Full Text
- View/download PDF
272. Antitumor effects of carnertinib in castration resistant prostate cancer models: a comparative study with erlotinib.
- Author
-
Gravina GL, Marampon F, Piccolella M, Biordi L, Ficorella C, Motta M, Jannini EA, Tombolini V, and Festuccia C
- Subjects
- Androgen Antagonists pharmacology, Anilides pharmacology, Animals, Apoptosis drug effects, Cell Division drug effects, Cell Line, Cell Line, Tumor, Disease Models, Animal, Drug Resistance, Neoplasm, Epithelial Cells cytology, Erlotinib Hydrochloride, Humans, Male, Mice, Nude, Nitriles pharmacology, Orchiectomy, Prostatic Neoplasms, Castration-Resistant pathology, Protein Kinase Inhibitors pharmacology, Tosyl Compounds pharmacology, Xenograft Model Antitumor Assays, Morpholines pharmacology, Prostate cytology, Prostatic Neoplasms, Castration-Resistant drug therapy, Quinazolines pharmacology
- Abstract
Background and Purpose: Although preclinical results suggest that the inhibition of erb-B1 or erb-B2 can be an useful tool to castration resistant prostate cancer (CRPC), neither inhibitor demonstrated to provide benefit in this category of patient. Here, we compared the effects of erlotinib, a specific EGFR inhibitor, with those observed with Carnertinib, an orally available pan-erbB receptor inhibitor, in a wide panel of hormone sensitive and independent prostate cancer cell lines., Materials and Methods: Variation in proliferation rate, cell cycle, and apoptosis after erlotinib and carnertinib treatments will be evaluated in vitro. In vivo experiments were performed using two models of CRPC, 22rv1 (AR expressing), and PC3 (AR negative) cell lines grown in nude mice. Intact nude mice bearing 22rv1 cells also received bicalutamide (BCLT) in combination with anti-target agents., Results: Here, we found that Erlotinib and carnertinib effectiveness was positively related to expression and activation levels of Her2, whereas erlotinib effectiveness was influenced to the EGFR/Her2 ratio resulting more effective when EGFR levels were significantly higher of Her2. Overall, in vitro carnertinib efficacy was higher than those observed with erlotinib. The combination between erlotinib and androgen deprivation therapy or BCLT showed no significant effects when compared to single treatments whereas carnertinib was active in presence of any anti-hormone manipulation., Conclusions: Erlotinib efficacy was higher in androgen-sensitive PCa cells when we compare to the effects evident in CRPC cells, whereas the carnertinib efficacy may have therapeutical significance in Her2 overexpressing AR+ CRPC models in combination with hormone manipulation., (Copyright © 2011 Wiley-Liss, Inc.)
- Published
- 2011
- Full Text
- View/download PDF
273. Ozarelix, a fourth generation GnRH antagonist, induces apoptosis in hormone refractory androgen receptor negative prostate cancer cells modulating expression and activity of death receptors.
- Author
-
Festuccia C, Dondi D, Piccolella M, Locatelli A, Gravina GL, Tombolini V, and Motta M
- Subjects
- Antineoplastic Agents pharmacology, Caspase 3 metabolism, Cell Cycle drug effects, Cell Division drug effects, Drug Synergism, Enzyme Activation drug effects, Extracellular Signal-Regulated MAP Kinases antagonists & inhibitors, Gefitinib, Humans, Male, Prostatic Neoplasms drug therapy, Quinazolines pharmacology, Tumor Cells, Cultured, p38 Mitogen-Activated Protein Kinases antagonists & inhibitors, Antineoplastic Agents therapeutic use, Apoptosis drug effects, Gonadotropin-Releasing Hormone antagonists & inhibitors, Prostatic Neoplasms pathology
- Abstract
Background and Aims: Antagonistic or agonistic analogues of gonadotropin-releasing hormone are extensively used for the treatment of advanced hormone-dependent prostate cancer. However, the majority of recurrent prostate tumors is androgen independent. This study explored the in vitro effects on DU145 and PC3 cell lines, two models of androgen-independent prostate cancer, of a fourth generation GnRH antagonist (Ozarelix)., Methods: Ozarelix was added to cultures and toxicity, cell cycle modifications, cell viability and caspase activity were investigated., Results: Ozarelix showed antiproliferative effects and produced an accumulation of cells in G2/M cell cycle phase. Apoptosis was related with caspase-8-dependent caspase 3 activation with down-regulation of c-FLIP (L) and a sensitization to TRAIL-induced apoptosis linked also to increased expression and activity of death receptors DR4/5 and Fas., Conclusions: TRAIL-resistant cancer cells can be sensitized to TRAIL by Ozarelix. This effect may be achieved by the activation of apoptotic pathway improving the therapeutic effects in androgen independent tumor cell lines. However, a better understanding of molecular mechanisms by which GnRH antagonists may act in androgen independent models is necessary., ((c) 2010 Wiley-Liss, Inc.)
- Published
- 2010
- Full Text
- View/download PDF
274. 5-Azacitidine restores and amplifies the bicalutamide response on preclinical models of androgen receptor expressing or deficient prostate tumors.
- Author
-
Gravina GL, Marampon F, Di Staso M, Bonfili P, Vitturini A, Jannini EA, Pestell RG, Tombolini V, and Festuccia C
- Subjects
- Androgen Antagonists administration & dosage, Androgen Receptor Antagonists, Anilides administration & dosage, Animals, Apoptosis drug effects, Azacitidine administration & dosage, Cell Growth Processes drug effects, Cell Line, Tumor, Drug Synergism, Humans, Male, Mice, Mice, Nude, Neoplasms, Experimental, Neoplasms, Hormone-Dependent metabolism, Neoplasms, Hormone-Dependent pathology, Nitriles administration & dosage, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Random Allocation, Tosyl Compounds administration & dosage, Androgen Antagonists pharmacology, Anilides pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Azacitidine pharmacology, Neoplasms, Hormone-Dependent drug therapy, Nitriles pharmacology, Prostatic Neoplasms drug therapy, Receptors, Androgen biosynthesis, Tosyl Compounds pharmacology
- Abstract
Background: Epigenetic modifications play a key role in the in prostate cancer (Pca) progression to a hormone refractory state (HRPC) and the current use of agents targeting epigenetic changes has become a topic of intense interest in cancer research. In this regard, 5-Azacitine (5-Aza) represents a promising epigenetic modulator. This study tested the hypothesis that 5-Aza may restore and enhance the responsiveness of HRPC cells to anti-hormonal therapy on Androgen receptor (AR) expressing (22rv1) and AR-deficient (PC3) cells., Methods: The effects were studied in vitro and in vivo models. This sequential treatment induced in vitro cell cycle arrest and apoptosis both in 22rv1 and PC3 tumor cell lines., Results: This combined treatment up-regulated the expression of FasL, phospho-FADD, p16(INKA), Bax, Bak, and p21(WAF1), and inhibited FLIP, Bcl-2, and Bcl-XL expression. The re-activation of hormonal response of AR-negative PC3 cell line was partially due to the AR re-expression mediated by 5-Aza treatment. In contrast, the increase in the response to anti-androgenic therapy in 22rv1 did not correlate with AR expression levels. Furthermore, xenograft studies revealed that the combined treatment of 5-Aza with AR-antagonist Bicalutamide had additive/synergistic effects in repressing tumor growth in vivo and the underlying mechanisms responsible for these effects seem to be in part mediated by induction of apoptosis., Conclusions: So, this study strongly suggests a therapeutic potential of 5-Aza in combination with anti-androgen therapy in patients with in AR expressing and AR-deficient HRPC., ((c) 2010 Wiley-Liss, Inc.)
- Published
- 2010
- Full Text
- View/download PDF
275. Epigenetic modulation of PTEN expression during antiandrogenic therapies in human prostate cancer.
- Author
-
Gravina GL, Biordi L, Martella F, Flati V, Ricevuto E, Ficorella C, Tombolini V, and Festuccia C
- Subjects
- Apoptosis drug effects, Azacitidine therapeutic use, Blotting, Western, Cells, Cultured, Humans, Male, PTEN Phosphohydrolase genetics, Polymerase Chain Reaction, RNA, Messenger analysis, Reverse Transcriptase Polymerase Chain Reaction, Valproic Acid therapeutic use, Androgen Antagonists therapeutic use, Epigenesis, Genetic drug effects, Gene Expression drug effects, PTEN Phosphohydrolase biosynthesis, Prostatic Neoplasms drug therapy, Prostatic Neoplasms genetics
- Abstract
Although the tumor-suppressor phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is frequently mutated or deleted in a wide variety of solid tumors, some malignancies, including prostate cancer, exhibit undetectable PTEN protein without loss of PTEN gene. Aim of this study was to evaluate whether the PTEN downmodulation, observed during bicalutamide treatment, was due to epigentic events. We analyzed the expression of PTEN in presence or absence of azacitidine or valproic acid in a panel of 50 primary cultures derived from naive (UNT, 23 ptz) and bicalutamide-based neoadjuvant hormone therapy-treated patients (NHT, 27 pts). Results showed that Western blot and PCR analyses showed that 54 and 68% of primary cultures displayed detectable amounts of PTEN protein and mRNA, respectively. Treatment with azacitidine increased the percentage of PTEN-positive cultures up to 72 and 80% for PTEN protein and mRNA determination, respectively. Treatment with valproic acid was able to increase the percentage of PTEN-positive cultures up to 80 and 74% for PTEN protein and mRNA determination, respectively. The percentage of cultures with undetectable levels of PTEN protein was significatively higher in cultures derived NHT patients respect to cultures derived from UNT men (P=0.020). Valproic acid reduced significantly the percentage of cultures PTEN-negative only at protein level and only in NHT (P=0.029) group. In conclusion, our data suggests that antiandrogenic therapy reduced PTEN expression by epigenetic mechanisms suggesting that epigenetic drugs, upmodulating PTEN expression, can reduce Akt activity and probably enhance the efficacy of antiandrogenic therapy.
- Published
- 2009
- Full Text
- View/download PDF
276. Effects of EGFR tyrosine kinase inhibitor erlotinib in prostate cancer cells in vitro.
- Author
-
Festuccia C, Gravina GL, Biordi L, D'Ascenzo S, Dolo V, Ficorella C, Ricevuto E, and Tombolini V
- Subjects
- Apoptosis drug effects, Cell Cycle Proteins metabolism, Cell Division drug effects, Cell Line, Tumor, ErbB Receptors metabolism, Erlotinib Hydrochloride, Gene Expression Regulation, Neoplastic, Humans, In Vitro Techniques, Male, PTEN Phosphohydrolase genetics, PTEN Phosphohydrolase metabolism, RNA, Small Interfering, Receptors, Androgen genetics, Receptors, Androgen metabolism, Signal Transduction drug effects, ErbB Receptors antagonists & inhibitors, Prostatic Neoplasms drug therapy, Prostatic Neoplasms pathology, Protein Kinase Inhibitors pharmacology, Quinazolines pharmacology
- Abstract
Background: Erlotinib is a small-molecule tyrosine kinase inhibitor targeted EGFR, known to be overexpressed in a variety of cancers, including prostate cancer. Clinical trials showed insignificant clinical benefit in patients with castration resistant prostate cancer both when EGFR inhibitors were administered as monotherapy or in association with antiandrogens or chemotherapeutics. Why, differently to other tumors, have EGFR inhibitors been so ineffective in human prostate cancer? This is the question that we have set in this report., Methods: For this purpose, the effectiveness of erlotinib, a selective EGFR inhibitor, in a wide range of prostate cancer cells (wild type or engineered to overexpress peculiar proteins including androgen receptor and PTEN)., Results: We demonstrated that the effectiveness of erlotinib was inversely correlated to the EGFR/Her2 ratio rather than EGFR/p-EGFR or Her2/p-Her2 levels. Chronic treatment with bicalutamide induced overexpression of Her2 and reduction of EGFR/Her2ratio and this was associated with increased Akt and Erk activity. In these conditions of treatment a reduced efficacy of erlotinib was observed. At the same time, an increased efficacy versus erlotinib was documented in cancer cells chronically exposed to DHT. In these culture conditions low levels of Her2 and increased EGFR/Her2 ratio were evidenced., Conclusions: Taken together, our results seem to suggest that a low EGFR/Her2 ratio and PTEN absence are the main factors responsible of erlotinib inefficacy. Therefore the inhibition of EGFR could have important antitumor effects in hormone-naive rather than in hormonally treated patients.
- Published
- 2009
- Full Text
- View/download PDF
277. Bicalutamide demonstrates biologic effectiveness in prostate cancer cell lines and tumor primary cultures irrespective of Her2/neu expression levels.
- Author
-
Gravina GL, Festuccia C, Millimaggi D, Tombolini V, Dolo V, Vicentini C, and Bologna M
- Subjects
- Cell Line, Tumor, Drug Screening Assays, Antitumor, Epidermal Growth Factor pharmacology, Flow Cytometry, Humans, Male, Prostate-Specific Antigen metabolism, Prostatic Neoplasms metabolism, Protein Kinase Inhibitors pharmacology, Receptor, ErbB-2 antagonists & inhibitors, Tumor Cells, Cultured, Androgen Antagonists pharmacology, Anilides pharmacology, Antineoplastic Agents pharmacology, Nitriles pharmacology, Prostatic Neoplasms pathology, Receptor, ErbB-2 metabolism, Tosyl Compounds pharmacology
- Abstract
Objectives: To evaluate the role of Her2/neu as a molecular marker predictive of the treatment response to bicalutamide in prostate cancer (PCa)., Methods: Four PCa cell lines with graded Her2/neu expression levels and 63 primary tumor cultures derived from men with PCa and selected according to Her2/neu tumor levels were used. Primary tumor cultures and PCa cell lines were treated with bicalutamide (0.1-10 microM) in the presence of dehydrotestosterone (10(-12) M) for 4 days. The presence of a significant correlation between Her2/nue expression and drug efficacy was used to define the role of Her2/neu as molecular predictor of bicalutamide effectiveness. As an indicator of drug effectiveness we used the concentration that inhibits 50% values determined after bicalutamide treatment., Results: After bicalutamide treatment, no significant differences in the concentration that inhibits 50% were found among the different tumor cell lines (P = .081). In this experimental model, the correlation analysis suggested that the effectiveness of this drug was not influenced by Her2/neu levels (r = 0.053, P = .823). In primary cultures with high Her2/neu levels (43 tumor cultures), the mean value of the concentration that inhibits 50% for bicalutamide was 0.43 +/- 0.13 microM, and in cultures with low Her2/neu levels (20 tumor cultures), the same parameter was 0.5 +/- 0.16 microM (P = .14). The correlation analysis suggested that the effectiveness of this drug was not influenced by Her2/neu levels (r = 0.33, P = .101)., Conclusions: Our biologic data seem to indicate that the antitumor effect of bicalutamide is independent of Her2/neu levels in preclinical models of PCa. Bicalutamide could be configured as a pharmacologic option to treat patients with high or low levels of Her2/neu.
- Published
- 2009
- Full Text
- View/download PDF
278. Downmodulation of dimethyl transferase activity enhances tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis in prostate cancer cells.
- Author
-
Festuccia C, Gravina GL, D'Alessandro AM, Millimaggi D, Di Rocco C, Dolo V, Ricevuto E, Vicentini C, and Bologna M
- Subjects
- Azacitidine pharmacology, Blotting, Western, Caspase 8 drug effects, Caspase 8 metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Decitabine, Dose-Response Relationship, Drug, Down-Regulation, Flow Cytometry, Humans, Male, Prostatic Neoplasms pathology, Receptors, TNF-Related Apoptosis-Inducing Ligand metabolism, TNF-Related Apoptosis-Inducing Ligand metabolism, Antimetabolites, Antineoplastic pharmacology, Azacitidine analogs & derivatives, Drug Resistance, Neoplasm physiology, Prostatic Neoplasms enzymology, TNF-Related Apoptosis-Inducing Ligand pharmacology
- Abstract
One of the major obstacles in curing prostate cancer is the development of drug resistance. It is not only imperative to discover the molecular basis of resistance but also to find therapeutic agents that can disrupt the resistant pathways. Tumor necrosis factor TNF-related apoptosis-inducing ligand TRAIL-like ligands or agonist TRAIL-receptor monoclonal antibodies have entered phase I and II clinical trials with a very limited cytotoxic profile when used systemically in a variety of cancers. Therefore, TRAIL-receptor agonists are new proapoptotic pharmaceutical agents with great potential as new cancer therapeutic agents. Although many cancer cells undergo TRAIL-mediated apoptosis, some are resistant to TRAIL. Therefore, we have been investigating mechanisms to overcome TRAIL resistance in cancer cells so that TRAIL-associated compounds can be used effectively in clinical trials. Epigenetic inactivation of proapoptotic genes, or activation of survival signaling, can cause cross-resistance to several anti-tumor therapies and to immune cytotoxic lymphocytes. We hypothesize that 5-aza-2 deoxycytidine aza-dCR, decitabine may render TRAIL-resistant prostate cancer cells sensitive to caspase-8-mediated apoptosis and may, therefore, be therapeutically efficient. We evaluated the antiproliferative effects of decitabine on the following four prostate cancer cell lines: well-differentiated AR positive LnCaP p53(+), PTEN- and 22rv1 p53(+) and PTEN(+)]; poorly-differentiated AR negative PC3 p53-, PTEN- and DU145 p53 mutant, PTEN(+). Here, we provide evidence that treatment with sub-optimal concentrations of decitabine are additive to TRAIL effects in well-differentiated PCa cells whereas the same treatment shows synergistic effects in poorly-differentiated PCa cells through increased caspase-8 expression, down-modulation of Akt activation and through the expression of certain anti-apoptotic molecules including FLIP, PED/PEA-15, survivin and c-IAP-1. Our findings demonstrate that decitabine at relatively low concentrations restores caspase-8 expression and sensitises resistant PCa cells to TRAIL-induced apoptosis leading to important implications in novel therapeutic strategies targeting defective apoptosis pathways in advanced prostate tumors.
- Published
- 2008
279. Effects of dutasteride on prostate carcinoma primary cultures: a comparative study with finasteride and MK386.
- Author
-
Festuccia C, Gravina GL, Muzi P, Pomante R, Angelucci A, Vicentini C, and Bologna M
- Subjects
- Aged, Cell Proliferation drug effects, Dutasteride, Humans, Male, Prostatic Neoplasms pathology, Tumor Cells, Cultured, 5-alpha Reductase Inhibitors, Azasteroids therapeutic use, Finasteride therapeutic use, Prostatic Neoplasms drug therapy
- Abstract
Purpose: The profound decrease in serum dihydrotestosterone observed with the dual 5alpha-reductase inhibitor dutasteride makes it an attractive agent for prostate cancer therapy. To our knowledge we compared for the first time the antitumor effect of dutasteride with that of the specific 5alpha-reductase-1 inhibitor MK386 and the specific 5alpha-reductase-2 inhibitor finasteride in human prostate primary cultures., Materials and Methods: Biochemical markers of the cellular response to 5alpha-reductase inhibitors were evaluated in primary cultures of prostate epithelial cancer cells from 54 patients with prostate carcinoma., Results: In our cohort of 54 patients prostate cancer cell growth decreased with dutasteride in 42 (about 78%), whereas in 21 (39%) it decreased with finasteride or MK386 alone. We observed a relationship between the levels of 5alpha-reductase enzymes in cell culture extracts and those revealed by immunohistochemistry in sections of samples from which we established primary cultures. Finasteride effects depended on 5alpha-reductase-2 levels and they were higher when the 5alpha-reductase-1:2 ratio was low. However, dutasteride effects were related to 5alpha-reductase-1 and 2 levels, and were not influenced by the 5alpha-reductase-1:2 ratio. Conversely the effects of MK386 were related to 5alpha-reductase-1 levels and they were higher when the 5alpha-reductase-1:2 ratio was high., Conclusions: Our data may provide a rationale for the use of a dual 5alpha-reductase inhibitor rather than a mono specific inhibitor for the prevention or treatment of early prostate cancer. This finding appears to confirm some preliminary clinical results and it could be due to the simultaneous presence of each 5alpha-reductase isoenzyme in prostate tumor cells.
- Published
- 2008
- Full Text
- View/download PDF
280. Akt down-modulation induces apoptosis of human prostate cancer cells and synergizes with EGFR tyrosine kinase inhibitors.
- Author
-
Festuccia C, Gravina GL, Muzi P, Millimaggi D, Dolo V, Vicentini C, and Bologna M
- Subjects
- Apoptosis physiology, Blotting, Western, Caspases metabolism, Cell Cycle drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Chromones pharmacology, Drug Synergism, Enzyme Activation, Epidermal Growth Factor immunology, Erlotinib Hydrochloride, Flow Cytometry, Humans, Male, Morpholines pharmacology, Neoplasms, Hormone-Dependent drug therapy, Neoplasms, Hormone-Dependent enzymology, PTEN Phosphohydrolase metabolism, Phosphorylcholine pharmacology, Prostatic Neoplasms drug therapy, Prostatic Neoplasms enzymology, Quinazolines pharmacology, Apoptosis drug effects, Neoplasms, Hormone-Dependent pathology, Phosphorylcholine analogs & derivatives, Prostatic Neoplasms pathology, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-akt antagonists & inhibitors
- Abstract
Background: PTEN is a well-characterized tumor suppressor that negatively regulates cell growth and survival through the modulation of PI3K/Akt pathway., Methods: In this paper, we investigated the effects of an PI3K/Akt inhibitor, perifosine, in human prostate cancer (PCa) cells analyzing cell proliferation, apoptosis, and the synergy with EGFR inhibitors., Results: Clinically achievable concentrations of perifosine, as well as Akt gene knockdown, induced a G0/G1 arrest and apoptosis in PTEN defective PCa cells. Although PTEN introduction was able to restore the control of Akt activity and to reduce cell proliferation, the manipulation of PTEN gene was not able alone to influence apoptosis. Perifosine induced apoptotic program also in PTEN positive cells when Akt activity was augmented by EGF suggesting the possibility that this drug could be used in combination with EGFR inhibitors. The combination treatment between erlotinib and pharmacological or molecular Akt knockdown, indeed, showed synergistic effects. This is the first demonstration that a pharmacological compound against Akt activity can restore the efficacy against EGFR inhibitors in PCa and has important therapeutic fallout since EGFR inhibitors have demonstrated very low effectiveness in PCa patients., Conclusions: Taken together our data have an important clinical relevance in the treatment of advanced prostate tumors. However, further studies in the setting of combination therapies in advanced PCas are necessary.
- Published
- 2008
- Full Text
- View/download PDF
281. Surgical and biologic outcomes after neoadjuvant bicalutamide treatment in prostate cancer.
- Author
-
Gravina GL, Festuccia C, Galatioto GP, Muzi P, Angelucci A, Ronchi P, Costa AM, Bologna M, and Vicentini C
- Subjects
- Adult, Aged, ErbB Receptors metabolism, Humans, Male, Middle Aged, Prostatic Neoplasms drug therapy, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Receptor, ErbB-2 metabolism, Up-Regulation, Androgen Antagonists therapeutic use, Anilides therapeutic use, Antineoplastic Agents, Hormonal therapeutic use, Neoadjuvant Therapy, Nitriles therapeutic use, Prostatectomy, Prostatic Neoplasms surgery, Tosyl Compounds therapeutic use
- Abstract
Objectives: To perform a randomized, prospective, controlled, intention-to-treat study to determine the usefulness of bicalutamide as a neoadjuvant hormonal therapy regimen to surgery in reducing positive surgical margins and modulating epidermal growth factor receptor (EGFR) member's in men with prostate cancer., Methods: From April 2002 to December 2003, 430 men were diagnosed with prostate cancer. Of these men, 119 with clinical Stage T2-T3a were enrolled. Of the 119 men, 61 were assigned to receive 150 mg/day bicalutamide for 120 days before radical prostatectomy (bicalutamide plus surgery group) and 58 to radical prostatectomy alone (surgery group). Logistic regression analysis was performed to determine the relationship between bicalutamide and EGFR/Her2/neu levels. P <0.05 was considered to indicate significance., Results: Patients treated with bicalutamide had a 3.5-fold increase in negative surgical margins (odds ratio [OR] 3.5; 95% confidence interval [CI] 1.4 to 8.74; P = 0.011). In particular, in Stage pT3a tumors, bicalutamide treatment was associated with a fivefold increase in negative surgical margins (OR 5.4; 95% CI 1.9 to 15.5; P = 0.002). In those with Stage pT2, no difference for this surgical outcome was noted. Immunohistochemical analysis revealed that bicalutamide increased EGFR levels 2.8-fold (OR 2.8; 95% CI 1.3 to 6.2; P = 0.014) and of 2.7-fold Her2/neu (OR 2.7; 95% CI 1.2 to 5.8; P = 0.022). When EGFR and Her2/neu were overexpressed, they were also active. In this regard, bicalutamide increased p-EGFR levels 3.3-fold (OR 3.3; 95% CI 1.3 to 8.2; P = 0.0016) and increased p-Her2/neu 2.8-fold (OR 2.8; 95% CI 1.2 to 6.3; P = 0.025)., Conclusions: Bicalutamide appears to reduce the prevalence of positive surgical margins. The upregulation of Her2/neu and EGFR and their phosphorylated forms was an early event after bicalutamide treatment. We hypothesized that the benefits of neoadjuvant hormonal therapy might be overwhelmed by the capacity of the residual tumor to acquire compensatory survival pathways and to grow and progress.
- Published
- 2007
- Full Text
- View/download PDF
282. In vitro and in vivo effects of bicalutamide on the expression of TrkA and P75 neurotrophin receptors in prostate carcinoma.
- Author
-
Festuccia C, Gravina GL, Muzi P, Pomante R, Ventura L, Ricevuto E, Vicentini C, and Bologna M
- Subjects
- Adult, Aged, Androgen Antagonists pharmacology, Androgen Antagonists therapeutic use, Anilides therapeutic use, Antineoplastic Agents therapeutic use, Blotting, Western, Carbazoles pharmacology, Carcinoma metabolism, Carcinoma pathology, Carcinoma surgery, Cell Line, Tumor, Cohort Studies, Dihydrotestosterone pharmacology, Furans, Humans, Immunohistochemistry, Indoles pharmacology, Male, Middle Aged, Neoadjuvant Therapy, Neoplasms, Hormone-Dependent metabolism, Neoplasms, Hormone-Dependent pathology, Nitriles therapeutic use, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Prostatic Neoplasms surgery, Protein Kinase Inhibitors pharmacology, Receptor, trkC biosynthesis, Tosyl Compounds therapeutic use, Anilides pharmacology, Antineoplastic Agents pharmacology, Carcinoma drug therapy, Neoplasms, Hormone-Dependent drug therapy, Nitriles pharmacology, Prostatic Neoplasms drug therapy, Receptor, Nerve Growth Factor biosynthesis, Receptor, trkA biosynthesis, Tosyl Compounds pharmacology
- Abstract
Rationale: Neurotrophine tyrosine kinase receptors (NTR) are expressed in prostate carcinoma (PCa), and their distribution seems to be related to disease malignancy., Material and Methods: In this article we analyzed the expression of NTRK1 (TrkA), NTRK2 (TrkB), NTRK3 (TrkC), and p75NTR in a 102 patient cohort with clinically localized tumors, which had been surgically treated with radical prostatectomy (RP). Among these, 61 patients received RP as sole treatment, and 41 patients received neoadjuvant hormone therapy (NHT) for 120 days with bicalutamide 150 mg/day. In addition, we analyzed the NTR expression in vitro in the androgen receptor positive, androgen-sensitive cell strains derived from CWR22R., Results and Conclusions: We demonstrated that: (i) TrkA and TrkC levels were significantly upmodulated, whereas (ii) p75NTR seemed to be reduced, and (iii) TrkB expression seemed to be not affected by NHT. TrkA were constitutively activated when its levels were very high. In vitro studies showed that the dehydrotestosterone (DHT) was able to maintain low TrkA and TrkC protein levels. Conversely, DHT was able to maintain p75NTR at high levels. Bicalutamide treatment induced TrkA and TrkC and reduced p75NTR expression. Antiproliferative effects of CEP701 were dependent to TrkA levels. A therapeutical effect of CEP701 was seen in all culture conditions, and bicalutamide seemed to sensitize prostate cancer cells to the effects of a pan TrkA inhibitor CEP701, suggesting that a sequential therapy between these drugs could further increase the efficacy of Trk inhibition.
- Published
- 2007
- Full Text
- View/download PDF
283. PPARgamma-dependent effects of conjugated linoleic acid on the human glioblastoma cell line (ADF).
- Author
-
Cimini A, Cristiano L, Colafarina S, Benedetti E, Di Loreto S, Festuccia C, Amicarelli F, Canuto RA, and Cerù MP
- Subjects
- Antineoplastic Agents pharmacology, Cell Adhesion drug effects, Cell Death drug effects, Cell Differentiation drug effects, Cell Division drug effects, Cell Line, Tumor, Cell Movement drug effects, Fluorescent Antibody Technique, Humans, Immunoblotting, Neoplasm Invasiveness, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Transcriptional Activation, Glioblastoma pathology, Linoleic Acids, Conjugated pharmacology, PPAR gamma physiology
- Abstract
Conjugated linoleic acid (CLA) has been shown to exert beneficial effects against carcinogenesis, atherosclerosis and diabetes. It has been demonstrated that CLA modulates lipid metabolism through the activation of peroxisome proliferator-activated receptors (PPARs). The PPAR family comprises 3 closely related gene products, PPAR alpha, beta/delta and gamma, differing for tissue distribution, developmental expression and ligand specificity. It has also been demonstrated that activated PPARgamma results in growth inhibition and differentiation of transformed cells. These observations stimulated a great interest toward PPARgamma ligands as potential anticancer drugs to be used in a differentiation therapy. Glioblastomas are the most commonly diagnosed primary tumors of the brain in humans. The prognosis of patients with high-grade gliomas is poor and only marginally improved by chemotherapy. The aim of this work was to study the effects of CLA and of a specific synthetic PPARgamma ligand on cell growth, differentiation and death of a human glioblastoma cell line as well as on parameters responsible for the metastatic behavior of this tumor. We demonstrate here that CLA and PPARgamma agonist strongly inhibit cell growth and proliferation rate and induce apoptosis. Moreover, both treatments decrease cell migration and invasiveness. The results obtained show that CLA acts, directly or indirectly, as a PPARgamma activator, strongly suggesting that this naturally occurring fatty acid may be used as brain antitumor drug and as a chemopreventive agent. Moreover, the gamma-agonist, once experimented and validated on man, may represent a useful coadjuvant in glioblastoma therapy and in the prevention of recurrences., (Copyright 2005 Wiley-Liss, Inc)
- Published
- 2005
- Full Text
- View/download PDF
284. Additive antitumor effects of the epidermal growth factor receptor tyrosine kinase inhibitor, gefitinib (Iressa), and the nonsteroidal antiandrogen, bicalutamide (Casodex), in prostate cancer cells in vitro.
- Author
-
Festuccia C, Gravina GL, Angelucci A, Millimaggi D, Muzi P, Vicentini C, and Bologna M
- Subjects
- Androgen Antagonists toxicity, Animals, Antineoplastic Agents toxicity, Cell Division drug effects, Cell Line, Tumor, Dihydrotestosterone pharmacology, Epidermal Growth Factor pharmacology, ErbB Receptors antagonists & inhibitors, Gefitinib, Humans, Male, Nitriles, Tosyl Compounds, Transplantation, Heterologous, Anilides toxicity, Prostatic Neoplasms pathology, Quinazolines toxicity
- Abstract
Progression from an androgen-dependent to an androgen-independent state often occurs in patients with prostate cancer (PCa) who undergo hormonal therapy. We have investigated whether inhibition of the epidermal growth factor receptor (EGFR) signaling pathway affects the antitumor effect of a nonsteroidal antiandrogen. Gefitinib (Iressa), an EGFR tyrosine kinase inhibitor, and bicalutamide (Casodex), a nonsteroidal antiandrogen [androgen receptor (AR) antagonist], were administered alone and in combination to AR-positive human PCa cell lines. FACS analysis showed lower EGFR expression levels on AR-positive cells (LNCaP, CWR22, CWR22R 2152 and AR-transfected DU145 cell lines) compared with AR-negative cells (DU145, PC3 and TSU-Pr1). Moreover, in AR-transfected DU145 cells, chronic treatment with bicalutamide increased EGFR expression to levels similar to androgen-independent DU145 cells. All AR-positive PCa cell lines were sensitive to gefitinib (IC50 = 0.1-0.6 microM), whereas higher concentrations of bicalutamide were needed to reduce AR-positive PCa cell line proliferation (IC50 = 0.8-2.0 microM). Low doses of gefitinib increased the antitumor effects of bicalutamide by strongly reducing the IC50 of bicalutamide (approximately 10-fold). Similarly, bicalutamide increased the antiproliferative effects of gefitinib by reducing the IC50 of gefitinib (approximately 5-fold). Taken together, our data suggest that in androgen-dependent cell lines, addition of gefitinib in combination with bicalutamide results in concurrent dual inhibition of AR and EGFR/HER2 pathways. This causes a significant delay in the onset of EGFR-driven androgen independence., (Copyright 2005 Wiley-Liss, Inc.)
- Published
- 2005
- Full Text
- View/download PDF
285. Effects of 5 alpha reductase inhibitors on androgen-dependent human prostatic carcinoma cells.
- Author
-
Festuccia C, Angelucci A, Gravina GL, Muzi P, Vicentini C, and Bologna M
- Subjects
- 5-alpha Reductase Inhibitors, Aged, Cell Line, Tumor, Cell Proliferation drug effects, Humans, Male, Neoplasm Staging, Prostatic Hyperplasia drug therapy, Prostatic Hyperplasia metabolism, Prostatic Neoplasms enzymology, Prostatic Neoplasms pathology, Androgens metabolism, Antineoplastic Agents, Hormonal pharmacology, Azasteroids pharmacology, Cholestenone 5 alpha-Reductase antagonists & inhibitors, Enzyme Inhibitors pharmacology, Finasteride pharmacology, Prostatic Neoplasms drug therapy, Prostatic Neoplasms metabolism
- Abstract
Purpose: To investigate the effects of MK906, a selective 5 alpha reductase (5alphaR) type 2 (5alphaR2) inhibitor, and of MK386, a specific 5alphaR1 inhibitor, on the cellular proliferation of androgen-dependent human prostatic cancer (PCa) cells in cultures of cells derived from bioptic and surgical tissues., Methods: In this study we tested the effects of MK906 and MK386 in 30 cultures derived from PCa, 6 from PIN and 10 from benign prostatic hyperplasia specimens., Results: Prostate primary cultures under short-term conditions (with <4 subcultures) represent a mixture of epithelial and stromal cells. Epithelial cells require testosterone (T) for optimal growth, but were not able to grow in the presence of T under long-term conditions even if DHT was able to induce cellular proliferation to a similar extent in both conditions, suggesting that 5alphaR can be lost in long-term cultures. Therefore, our studies were performed under short-term conditions. Both 5alphaR inhibitors decreased cell proliferation significantly and dose-dependently in all the samples tested. MK906 was more efficient than MK386 in 7 out of 10 cultures derived from BPH tissues, in 4 out of 6 cultures derived from PIN and in 18 out of 30 cultures derived from PCa. In 3 out of 10 BPH, in 2 out of 6 PIN and in 5 out of 30 PCa-derived cultures, both inhibitors presented similar efficacy, whereas in 1 out of 10 BPH and 7 out of 30 PCa-derived cultures MK386 was more efficient than MK906. In addition, MK386 was more efficient than MK906 in 4 out of 15 non-metastatic PCa and 2 out of 7 metastatic PCa-derived cultures., Conclusions: Considering that 5alphaR1 (responsible primarily for androgenic catabolism) is mostly expressed in epithelial cells and that 5alphaR2 (responsible for local DHT synthesis and release) is expressed in the stromal cells (which provides several paracrine growth factors and DHT itself to the epithelial cells), our experiments suggest that the inhibition of both 5alphaR1 and 5alphaR2 by MK386 and MK906, respectively, may have therapeutic potential in order to reduce the growth and progression of human prostatic cancers, through the inhibition of autocrine or paracrine mechanisms involving the stromal cell compartment. In addition, some effects of 5alphaR inhibitors could be mediated by estrogens, which are synthesized by the aromatase enzyme present in the epithelial cells. These aspects could be considered in order to improve the therapeutical management of PCa and for future clinical trials.
- Published
- 2005
- Full Text
- View/download PDF
286. Osteopontin enhances the cell proliferation induced by the epidermal growth factor in human prostate cancer cells.
- Author
-
Angelucci A, Festuccia C, Gravina GL, Muzi P, Bonghi L, Vicentini C, and Bologna M
- Subjects
- Cell Adhesion, Cytokines, Disease Progression, Humans, Male, Osteopontin, Phosphoproteins, Bone Neoplasms physiopathology, Bone Neoplasms secondary, Cell Division drug effects, Epidermal Growth Factor pharmacology, ErbB Receptors physiology, Prostatic Neoplasms pathology, Sialoglycoproteins pharmacology, Stromal Cells physiology
- Abstract
Background: Susceptibility to extracellular matrix and growth factors has been demonstrated to play a critical role in the development of prostate cancer (PCa) metastases. The aim of this study was to elucidate some mechanisms by which stroma controls tumor progression., Methods: In our study we tested the growth ability of the LNCaP human prostatic cell line in steroid-free culture conditions in response to osteopontin (OPN), a non-collageneous matrix protein, localized in large amounts in the bone., Results: In the LNCaP cell model, OPN stimulates cell proliferation in serum-free medium and colony growth at high dilution but this effect is visible only in presence of epidermal growth factor (EGF). Proliferation induced by OPN is accompanied by a sustained activation of EGF receptor (EGFR) whose phosphorylation is detectable up to 12 hr after treatment in association with EGF. The colocalization of integrin beta1, a ligand of OPN, and of EGFR on the cellular membrane, suggests that the association of these cell surface receptors may be the principal mechanism involved in the long-term activation of the EGFR., Conclusions: Our data describe a new possible mechanism involved in the establishment of bone metastases which may also account for the formation of androgen-independent cellular clones, frequently responsible of the clinical progression of PCa., (Copyright 2004 Wiley-Liss, Inc.)
- Published
- 2004
- Full Text
- View/download PDF
287. Prostate cancer cell proliferation is strongly reduced by the epidermal growth factor receptor tyrosine kinase inhibitor ZD1839 in vitro on human cell lines and primary cultures.
- Author
-
Vicentini C, Festuccia C, Gravina GL, Angelucci A, Marronaro A, and Bologna M
- Subjects
- Apoptosis drug effects, Cell Cycle drug effects, Cell Division drug effects, Enzyme Inhibitors pharmacology, ErbB Receptors metabolism, Gefitinib, Humans, In Vitro Techniques, Male, Phosphorylation, Prostatic Neoplasms metabolism, Tumor Cells, Cultured, Tyrosine, Antineoplastic Agents pharmacology, Epidermal Growth Factor antagonists & inhibitors, Prostatic Neoplasms pathology, Protein-Tyrosine Kinases antagonists & inhibitors, Quinazolines pharmacology
- Abstract
Purpose: To investigate the effects of the epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) ZD1839 ('Iressa') on the cellular proliferation of androgen-sensitive and androgen-independent human prostatic cancer cell lines and primary cultures in vitro., Experimental Design: In this study, we investigated the effects of the quinazoline ZD1839, a potent, selective EGFR-TKI, on the EGFR autophosphorylation and cellular proliferation of androgen-sensitive (ND1, LNCaP, and ALVA-31) and androgen-independent (PC3, DU145, and TSU-Pr1) human prostatic cancer cell lines and 20 primary cultures derived from human prostatic cancer tissue., Results: EGFR was present and phosphorylated in all cell lines tested. ZD1839 reduced EGFR autophosphorylation in intact cell lines with IC(50)s of 0.46-0.97 microM, and inhibited cellular proliferation with IC(50)s of 0.37-1.03 microM. Constitutive EGFR autophosphorylation was low in primary cell cultures, but addition of EGF (50 ng/ml) caused marked EGFR autophosphorylation; cellular proliferation in the presence of EGF was inhibited by ZD1839 with a mean IC(50) of 0.45 microM. At doses >1 microM, ZD1839 induced apoptosis in both androgen-dependent and androgen-independent PCa cell lines. CONCLUSION. Our experiments suggest that EGFR-TKIs such as ZD1839 may have potential in blocking the growth and progression of human prostatic cancers even in early phases of the disease.
- Published
- 2003
- Full Text
- View/download PDF
288. Bombesin-dependent pro-MMP-9 activation in prostatic cancer cells requires beta1 integrin engagement.
- Author
-
Festuccia C, Angelucci A, Gravina G, Eleuterio E, Vicentini C, and Bologna M
- Subjects
- Cell Adhesion drug effects, Cell Line, Chemotaxis drug effects, Collagen Type I metabolism, Cytochalasin D pharmacology, Cytoskeleton metabolism, Enzyme Activation, Enzyme Inhibitors pharmacology, Extracellular Matrix metabolism, Fibrinolytic Agents pharmacology, Gelatinases metabolism, Humans, Male, Matrix Metalloproteinase 9, Nucleic Acid Synthesis Inhibitors pharmacology, Plasminogen pharmacology, Prostatic Neoplasms pathology, Signal Transduction, Tumor Cells, Cultured, Urokinase-Type Plasminogen Activator analysis, Urokinase-Type Plasminogen Activator metabolism, src-Family Kinases antagonists & inhibitors, Bombesin pharmacology, Collagenases metabolism, Enzyme Precursors metabolism, Integrin beta1 metabolism, Prostatic Neoplasms enzymology, Prostatic Neoplasms metabolism
- Abstract
Bombesin-like peptides, including the mammalian homologue gastrin-releasing peptide, are highly expressed and secreted by neuroendocrine cells in prostate carcinoma tissues and are likely to be related to the progression of this neoplastic disease. Previously, we demonstrated that bombesin increased migration and protease expression in androgen-independent cells. In this work we show that bombesin is able to activate pro-MMP-9 through a mechanism involving the beta1 integrin subunit. In fact, MMP-9 processing was evident only when beta1 integrin was engaged with specific adhesive substrates, such as type I collagen, or when cells were seeded on dishes coated with antibodies against beta1 integrin, resulting in activation of the surface ligand. When exogenous pro-MMP-9 was added to PC3 cells, MMP-9 active forms were produced within 30 min by bombesin-treated cultures while control cultures expressed activated forms only after a longer time and at lower levels. MMP-9 activation required cytoskeleton integrity since this effect was abolished by cytochalasin D. Engagement of beta1 integrin caused an increased membrane-linked uPA activity which was required for MMP-9 activation. The cross talk between bombesin- and beta1-integrin-engaged signals seems to be crucial for the modulation of both membrane-linked uPA activity and MMP-9 activation and triggers complex intracellular signaling pathways requiring activation of tyrosine kinase activity, including that of src and PI3K. The beta1 integrin may be considered an important mechanism by which bombesin induces MMP-9 activation. This finding supports the idea that cellular responses to growth factors may be driven by cell-matrix interactions and stresses the role of neuroendocrine factors in prostate carcinoma progression.
- Published
- 2002
- Full Text
- View/download PDF
289. Bicalutamide dose-dependently inhibits proliferation in human prostatic carcinoma cell lines and primary cultures.
- Author
-
Vicentini C, Festuccia C, Angelucci A, Gravina GL, Muzi P, Eleuterio E, Miano R, Marronaro A, Tubaro A, and Bologna M
- Subjects
- Acridine Orange, Cell Division drug effects, Dose-Response Relationship, Drug, Ethidium, Fluorescent Dyes, Humans, Inhibitory Concentration 50, Male, Nitriles, Prostatic Neoplasms pathology, Staining and Labeling methods, Tosyl Compounds, Tumor Cells, Cultured, Androgen Antagonists pharmacology, Anilides pharmacology, Antineoplastic Agents pharmacology, Prostatic Neoplasms drug therapy
- Abstract
Objective: Androgen antagonists inhibit prostatic cell proliferation in normal and pathological conditions and are useful antitumor agents in prostatic carcinoma (PCa). Bicalutamide (BCLT) is a well-known non-steroidal antiandrogenic agent able to interfere with androgen receptor (AR). We tested the efficacy of BCLT in inhibiting proliferation of human PCa cell lines and of primary cultures from biopsies of PCa patients., Materials and Methods: Human prostatic carcinoma cell lines (PC3, DU145, LNCaP ALVA 31 and ND1) and short-term primary tissue cultures from PCa patients were treated with BCLT. Cell proliferation and orange acridine and ethidium bromide fluorescence staining studies were performed., Results: BCLT was able to inhibit, significantly and dose-dependently, cell proliferation in AR-positive human PCa cell lines and in 10 cases of primary cultures with Gleason grades 4 to 8. Its action appears to be mainly apoptotic in AR-positive cells and cytotoxic in AR-negative cells., Conclusion: BCLT, which inhibits growth in both human PCa cell lines and PCa primary cultures from patients with medium and low-grade tumors, deserves attention as a potential widely effective antiandrogenic monotherapy in prostatic carcinoma. However, its efficacy in AR-negative cells requires further research.
- Published
- 2002
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.