26 results on '"Beraldi E"'
Search Results
2. Toxoplasmic Infection-induced Injury in the Ileal Myenteric Plexus in Rats Depends on the Dose of Toxoplasma gondii Oocysts
- Author
-
Tironi, L., additional, Beraldi, E., additional, Borges, S., additional, Massocato, C., additional, Vieira, S., additional, Sant'ana, D., additional, Araújo, E., additional, and Buttow, N., additional
- Published
- 2018
- Full Text
- View/download PDF
3. Toxoplasma gondiipromotes changes in VIPergic submucosal neurons, mucosal intraepithelial lymphocytes, and goblet cells during acute infection in the ileum of rats
- Author
-
Schneider, L. C. L., primary, do Nascimento, J. C. P., additional, Trevizan, A. R., additional, Góis, M. B., additional, Borges, S. C., additional, Beraldi, E. J., additional, Garcia, J. L., additional, Sant'Ana, D. M. G., additional, and Buttow, N. C., additional
- Published
- 2017
- Full Text
- View/download PDF
4. Targeting enzalutamide-resistant prostate cancer using the novel androgen receptor inhibitor ODM-201
- Author
-
Borgmann, H., primary, Ozistanbullu, D., additional, Beraldi, E., additional, Dalal, K., additional, Fazli, L., additional, and Gleave, M., additional
- Published
- 2017
- Full Text
- View/download PDF
5. 209 A phase 1 clinical trial assessing an intravesical administered second-generation antisense oligonucleotide targeting heat shock protein 27 in bladder cancer
- Author
-
Frees, S., primary, Beraldi, E., additional, Chi, K., additional, Fazli, L., additional, Black, P., additional, Gleave, M., additional, and So, A., additional
- Published
- 2016
- Full Text
- View/download PDF
6. 40 Efficacy of prostate cancer compound with novel mechanism of action targeting the DNA binding domain of the androgen receptor
- Author
-
Borgmann, H., primary, Dalal, K., additional, Beraldi, E., additional, Cherkasov, A., additional, Rennie, P., additional, and Gleave, M., additional
- Published
- 2016
- Full Text
- View/download PDF
7. 355 - Head-to-head comparison of efficacy of darolutamide (ODM-201) vs. enzalutamide on mutated forms of the androgen receptor
- Author
-
Borgmann, H., Lallous, N., Ozistanbullu, D., Beraldi, E., Paul, N., Dalal, K., Fazli, L., Haferkamp, A., Lejeune, P., Cherkasov, A., and Gleave, M.
- Published
- 2018
- Full Text
- View/download PDF
8. <italic>Toxoplasma gondii</italic> promotes changes in VIPergic submucosal neurons, mucosal intraepithelial lymphocytes, and goblet cells during acute infection in the ileum of rats.
- Author
-
Schneider, L. C. L., do Nascimento, J. C. P., Trevizan, A. R., Góis, M. B., Borges, S. C., Beraldi, E. J., Garcia, J. L., Sant'Ana, D. M. G., and Buttow, N. C.
- Subjects
SUBMUCOUS plexus ,TOXOPLASMA gondii ,LYMPHOCYTES ,EXFOLIATIVE cytology ,LABORATORY rats ,ILEUM diseases - Abstract
Abstract: Background: The intestinal mucosa plays an important role in the mechanical barrier against pathogens. During
Toxoplasma gondii infection, however, the parasites invade the epithelial cells of the small intestine and initiate a local immune response. In the submucosal plexus, this response promotes an imbalance of neurotransmitters and induces neuroplasticity, which can change the integrity of the epithelium and its secretory function. This study evaluated the submucosal neurons throughout acuteT. gondii infection and the relationship between possible alterations and the epithelial and immune defense cells of the mucosa. Methods: Forty Wistar rats were randomly assigned to 8 groups (n = 5): 1 control group, uninfected, and 7 groups infected with an inoculation of 5000 sporulatedT. gondii oocysts (ME‐49 strain, genotype II). Segments of the ileum were collected for standard histological processing, histochemical techniques, and immunofluorescence. Key Results: The infection caused progressive neuronal loss in the submucosal general population and changed the proportion of VIPergic neurons throughout the infection periods. These changes may be related to the observed reduction in goblet cells that secret sialomucins and increase in intraepithelial lymphocytes after 24 hours, and the increase in immune cells in the lamina propria after 10 days of infection. The submucosa also presented fibrogenesis, characterizing injury and tissue repair. Conclusions and Inferences: The acuteT. gondii infection in the ileum of rats changes the proportion of VIPergic neurons and the epithelial cells, which can compromise the mucosal defense during infection. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
9. 747 - Targeting enzalutamide-resistant prostate cancer using the novel androgen receptor inhibitor ODM-201
- Author
-
Borgmann, H., Ozistanbullu, D., Beraldi, E., Dalal, K., Fazli, L., and Gleave, M.
- Published
- 2017
- Full Text
- View/download PDF
10. Chaperone-mediated autophagy promotes PCa survival during ARPI through selective proteome remodeling.
- Author
-
Nikesitch N, Beraldi E, Zhang F, Adomat H, Bell R, Suzuki K, Fazli L, Hy Kung S, Wells C, Pinette N, Saxena N, Wang Y, and Gleave M
- Subjects
- Male, Humans, Receptors, Androgen genetics, Androgens metabolism, Proteome, Proteomics, Autophagy, Cell Line, Tumor, Prostatic Neoplasms, Castration-Resistant pathology, Chaperone-Mediated Autophagy
- Abstract
The androgen receptor (AR) plays an important role in PCa metabolism, with androgen receptor pathway inhibition (ARPI) subjecting PCa cells to acute metabolic stress caused by reduced biosynthesis and energy production. Defining acute stress response mechanisms that alleviate ARPI stress and therefore mediate prostate cancer (PCa) treatment resistance will help improve therapeutic outcomes of patients treated with ARPI. We identified the up-regulation of chaperone-mediated autophagy (CMA) in response to acute ARPI stress, which persisted in castration-resistant PCa (CRPC); previously undefined in PCa. CMA is a selective protein degradation pathway and a key stress response mechanism up-regulated under several stress stimuli, including metabolic stress. Through selective protein degradation, CMA orchestrates the cellular stress response by regulating cellular pathways through selective proteome remodeling. Through broad-spectrum proteomic analysis, CMA coordinates metabolic reprogramming of PCa cells to sustain PCa growth and survival during ARPI; through the upregulation of mTORC1 signaling and pathways associated with PCa biosynthesis and energetics. This not only promoted PCa growth during ARPI, but also promoted the emergence of CRPC in-vivo. During CMA inhibition, PCa metabolism is compromised, leading to ATP depletion, resulting in a profound anti-proliferative effect on PCa cells, and is enhanced when combined with ARPI. Furthermore, CMA inhibition prevented in-vivo tumour formation, and also re-sensitized enzalutamide-resistant cell lines in-vitro. The profound anti-proliferative effect of CMA inhibition was attributed to cell cycle arrest mediated through p53 transcriptional repression of E2F target genes. In summary, CMA is an acute ARPI stress response mechanism, essential in alleviating ARPI induced metabolic stress, essential for ensuring PCa growth and survival. CMA plays a critical role in the development of ARPI resistance in PCa., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
11. Regulation of AR mRNA translation in response to acute AR pathway inhibition.
- Author
-
Somasekharan SP, Saxena N, Zhang F, Beraldi E, Huang JN, Gentle C, Fazli L, Thi M, Sorensen PH, and Gleave M
- Subjects
- Cell Line, Tumor, Humans, Male, Protein Biosynthesis, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, RNA, Messenger metabolism, Receptors, Androgen genetics
- Abstract
We report a new mechanism of androgen receptor (AR) mRNA regulation and cytoprotection in response to AR pathway inhibition (ARPI) stress in prostate cancer (PCA). AR mRNA translation is coordinately regulated by RNA binding proteins, YTHDF3 and G3BP1. Under ambient conditions m6A-modified AR mRNA is bound by YTHDF3 and translationally stimulated, while m6A-unmodified AR mRNA is bound by G3BP1 and translationally repressed. When AR-regulated PCA cell lines are subjected to ARPI stress, m6A-modified AR mRNA is recruited from actively translating polysomes (PSs) to RNA-protein stress granules (SGs), leading to reduced AR mRNA translation. After ARPI stress, m6A-modified AR mRNA liquid-liquid phase separated with YTHDF3, while m6A-unmodified AR mRNA phase separated with G3BP1. Accordingly, these AR mRNA messages form two distinct YTHDF3-enriched or G3BP1-enriched clusters in SGs. ARPI-induced SG formation is cell-protective, which when blocked by YTHDF3 or G3BP1 silencing increases PCA cell death in response to ARPI stress. Interestingly, AR mRNA silencing also delays ARPI stress-induced SG formation, highlighting its supportive role in triggering this stress response. Our results define a new mechanism for stress adaptive cell survival after ARPI stress involving SG-regulated translation of AR mRNA, mediated by m6A RNA modification and their respective regulatory proteins., (© The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2022
- Full Text
- View/download PDF
12. Androgen receptor (AR) antagonism triggers acute succinate-mediated adaptive responses to reactivate AR signaling.
- Author
-
Saxena N, Beraldi E, Fazli L, Somasekharan SP, Adomat H, Zhang F, Molokwu C, Gleave A, Nappi L, Nguyen K, Brar P, Nikesitch N, Wang Y, Collins C, Sorensen PH, and Gleave M
- Subjects
- Cell Line, Tumor, Humans, Male, Receptors, Androgen genetics, Succinic Acid, Androgen Receptor Antagonists pharmacology, Prostatic Neoplasms
- Abstract
Treatment-induced adaptive pathways converge to support androgen receptor (AR) reactivation and emergence of castration-resistant prostate cancer (PCa) after AR pathway inhibition (ARPI). We set out to explore poorly defined acute adaptive responses that orchestrate shifts in energy metabolism after ARPI and identified rapid changes in succinate dehydrogenase (SDH), a TCA cycle enzyme with well-known tumor suppressor activity. We show that AR directly regulates transcription of its catalytic subunits (SDHA, SDHB) via androgen response elements (AREs). ARPI acutely suppresses SDH activity, leading to accumulation of the oncometabolite, succinate. Succinate triggers calcium ions release from intracellular stores, which in turn phospho-activates the AR-cochaperone, Hsp27 via p-CaMKK2/p-AMPK/p-p38 axis to enhance AR protein stabilization and activity. Activation of this pathway was seen in tissue microarray analysis on prostatectomy tissues and patient-derived xenografts. This adaptive response is blocked by co-targeting AR with Hsp27 under both in vitro and in vivo studies, sensitizing PCa cells to ARPI treatments., (© 2021 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2021
- Full Text
- View/download PDF
13. A polymeric paste-drug formulation for local treatment of upper tract urothelial carcinoma.
- Author
-
Kesch C, Schmitt V, Bidnur S, Thi M, Beraldi E, Moskalev I, Yago V, Bowden M, Adomat H, Fazli L, Jackson JK, and Gleave ME
- Subjects
- Administration, Topical, Animals, Deoxycytidine administration & dosage, Female, Humans, Polymers, Swine, Gemcitabine, Antimetabolites, Antineoplastic administration & dosage, Carcinoma, Transitional Cell drug therapy, Deoxycytidine analogs & derivatives, Drug Compounding, Kidney Neoplasms drug therapy, Kidney Pelvis, Ureteral Neoplasms drug therapy
- Abstract
Background: Intravesical instillation of chemo- or immunotherapy is commonly used in bladder cancer. Upper tract urothelial carcinoma (UTUC) shares similar pathological features, but current formulations are not suitable for direct instillation to the upper urinary tract., Objective: To evaluate in vivo applicability, characteristics and toxicity of ST-UC, a mucoadhesive polymeric paste formulation of gemcitabine, for upper urinary tract instillation., Material and Methods: Three pigs received 10 ml of ST-UC (100 mg/ml gemcitabine) retrogradely into 1 renal pelvis for pharmacokinetic studies. Four days later, a second injection into the contralateral renal pelvis was followed by serial euthanasia of the pigs and nephroureterectomy after 1, 3, and 6 hours. Adverse effects were monitored. Urine, serum, and tissue gemcitabine concentrations were measured, along with histologic examination of the upper urinary tract., Results: Retrograde instillation of ST-UC was well tolerated with mild, completely receding hydronephrosis. Urine gemcitabine concentrations were highest in the first 3-hour collection interval. Hundred percent of gemcitabine was recovered in the urine within 24 hours. Serum peak concentrations (c
max ) of gemcitabine were low at 5.5 µg/ml compared to the 10 to 30 µg/ml levels observed after a single intravenous dose of 1,000 mg/m2 gemcitabine. The formulation was still traceable after one hour and gemcitabine tissue concentrations are supportive of this extended drug exposure. No major histopathological changes were observed. The main limitation of this study is the lack of antitumor activity data., Conclusion: This preclinical evaluation of ST-UC demonstrated feasible instillation in the renal pelvis, no significant safety concerns, and sustained release of gemcitabine., (Copyright © 2020. Published by Elsevier Inc.)- Published
- 2021
- Full Text
- View/download PDF
14. Paternally Expressed Gene 10 (PEG10) Promotes Growth, Invasion, and Survival of Bladder Cancer.
- Author
-
Kawai Y, Imada K, Akamatsu S, Zhang F, Seiler R, Hayashi T, Leong J, Beraldi E, Saxena N, Kretschmer A, Oo HZ, Contreras-Sanz A, Matsuyama H, Lin D, Fazli L, Collins CC, Wyatt AW, Black PC, and Gleave ME
- Subjects
- Female, Humans, Male, Neoplasm Invasiveness, Survival Analysis, Urinary Bladder Neoplasms mortality, Urinary Bladder Neoplasms pathology, Apoptosis Regulatory Proteins metabolism, DNA-Binding Proteins metabolism, RNA-Binding Proteins metabolism, Urinary Bladder Neoplasms genetics
- Abstract
Paternally expressed gene 10 ( PEG10 ) has been associated with neuroendocrine muscle-invasive bladder cancer (MIBC), a subtype of the disease with the poorest survival. In this work, we further characterized the expression pattern of PEG10 in The Cancer Genome Atlas database of 412 patients with MIBC, and found that, compared with other subtypes, PEG10 mRNA level was enhanced in neuroendocrine-like MIBC and highly correlated with other neuroendocrine markers. PEG10 protein level also associated with neuroendocrine markers in a tissue microarray of 82 cases. In bladder cancer cell lines, PEG10 expression was induced in drug-resistant compared with parental cells, and knocking down of PEG10 resensitized cells to chemotherapy. Loss of PEG10 increased protein levels of cell-cycle regulators p21 and p27 and delayed G
1 -S-phase transition, while overexpression of PEG10 enhanced cancer cell proliferation. PEG10 silencing also lowered levels of SLUG and SNAIL, leading to reduced invasion and migration. In an orthotopic bladder cancer model, systemic treatment with PEG10 antisense oligonucleotide delayed progression of T24 xenografts. In summary, elevated expression of PEG10 in MIBC may contribute to the disease progression by promoting survival, proliferation, and metastasis. Targeting PEG10 is a novel potential therapeutic approach for a subset of bladder cancers., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
15. A polymeric paste-drug formulation for intratumoral treatment of prostate cancer.
- Author
-
Kesch C, Schmitt V, Bidnur S, Thi M, Beraldi E, Moskalev I, Yago V, Bowden M, Adomat H, Fazil L, Jackson JK, and Gleave ME
- Subjects
- Anilides administration & dosage, Animals, Antineoplastic Combined Chemotherapy Protocols chemistry, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Apoptosis, Cell Proliferation, Docetaxel administration & dosage, Humans, Male, Mice, Nitriles administration & dosage, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Rats, Tissue Distribution, Tosyl Compounds administration & dosage, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols pharmacology, Drug Compounding methods, Polymers chemistry, Prostatic Neoplasms drug therapy
- Abstract
Objective: Focal therapy has emerged as a treatment option for low- to intermediate-risk localized prostate cancer (PCa) patients, to balance the risks for urinary and sexual morbidity of radical treatment with the psychological burden of active surveillance. In this context, we developed ST-4PC, an injectable, polymeric paste formulation containing docetaxel (dtx) and bicalutamide (bic) for image-guided focal therapy of PCa. The objective of this work was to evaluate the in vitro characteristics and in vivo toxicity and efficacy of ST-4PC., Material and Methods: In vitro drug release was evaluated using high-performance liquid chromatography. In vivo toxicity of blank- and drug-loaded ST-4PC was assessed in mice and rats. Tumor growth inhibition was evaluated in LNCaP subcutaneous (s.c.) and LNCaP-luc orthotopic xenograft models. Using the s.c. model, mice were monitored weekly for weight loss, tumor volume (TV) and serum PSA. For the orthotopic model, mice were additionally monitored for bioluminescence as measure of tumor growth., Results: ST-4PC demonstrated a sustained and steady release of incorporated drugs with 50% dtx and 20% bic being released after 14 days. While no systemic toxicity was observed, dose-dependent local side effects from dtx developed in the s.c. but not in the orthotopic model, illustrating the limitations of s.c. models for evaluating local cytotoxic therapy. In the s.c. model, 0.1%/4% and 0.25%/4% dtx/bic ST-4PC paste significantly reduced PSA progression, but did not have a significant inhibitory effect on TV. ST-4PC loaded with 1%/4% dtx/bic significantly reduced TV, serum PSA, and bioluminescence in the orthotopic xenograft model. Compared with drugs dissolved in DMSO, ST-4PC significantly delayed tumor growth., Conclusion: Image-guided focal therapy using ST-4PC demonstrated promising inhibition of PSA progression and orthotopic tumor growth in vivo without significant toxicity, and warrants further clinical evaluation.
- Published
- 2020
- Full Text
- View/download PDF
16. Ivermectin inhibits HSP27 and potentiates efficacy of oncogene targeting in tumor models.
- Author
-
Nappi L, Aguda AH, Nakouzi NA, Lelj-Garolla B, Beraldi E, Lallous N, Thi M, Moore S, Fazli L, Battsogt D, Stief S, Ban F, Nguyen NT, Saxena N, Dueva E, Zhang F, Yamazaki T, Zoubeidi A, Cherkasov A, Brayer GD, and Gleave M
- Subjects
- A549 Cells, Animals, Humans, Intracellular Signaling Peptides and Proteins chemistry, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Mice, Protein Domains, Protein Multimerization, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Heat-Shock Proteins antagonists & inhibitors, Heat-Shock Proteins chemistry, Heat-Shock Proteins genetics, Heat-Shock Proteins metabolism, Ivermectin chemistry, Ivermectin pharmacology, Molecular Chaperones antagonists & inhibitors, Molecular Chaperones chemistry, Molecular Chaperones genetics, Molecular Chaperones metabolism, Neoplasms, Experimental drug therapy, Neoplasms, Experimental genetics, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Receptor, ErbB-2 genetics, Receptor, ErbB-2 metabolism
- Abstract
HSP27 is highly expressed in, and supports oncogene addiction of, many cancers. HSP27 phosphorylation is a limiting step for activation of this protein and a target for inhibition, but its highly disordered structure challenges rational structure-guided drug discovery. We performed multistep biochemical, structural, and computational experiments to define a spherical 24-monomer complex composed of 12 HSP27 dimers with a phosphorylation pocket flanked by serine residues between their N-terminal domains. Ivermectin directly binds this pocket to inhibit MAPKAP2-mediated HSP27 phosphorylation and depolymerization, thereby blocking HSP27-regulated survival signaling and client-oncoprotein interactions. Ivermectin potentiated activity of anti-androgen receptor and anti-EGFR drugs in prostate and EGFR/HER2-driven tumor models, respectively, identifying a repurposing approach for cotargeting stress-adaptive responses to overcome resistance to inhibitors of oncogenic pathway signaling.
- Published
- 2020
- Full Text
- View/download PDF
17. Design and Characterization of Injectable Poly(Lactic-Co-Glycolic Acid) Pastes for Sustained and Local Drug Release.
- Author
-
Schmitt V, Kesch C, Jackson JK, Bidnur S, Beraldi E, Yago V, Bowden M, and Gleave ME
- Subjects
- Anilides chemistry, Anilides pharmacology, Animals, Antineoplastic Agents chemistry, Antineoplastic Agents therapeutic use, Docetaxel chemistry, Docetaxel pharmacology, Drug Liberation, Drug Stability, Humans, Injections, Lidocaine chemistry, Lidocaine pharmacokinetics, Male, Mice, Mice, Nude, Neoplasms, Experimental, Nitriles chemistry, Nitriles pharmacology, Prostatic Neoplasms drug therapy, Rats, Tosyl Compounds chemistry, Tosyl Compounds pharmacology, Viscosity, Drug Carriers chemistry, Drug Compounding methods, Ointments chemistry, Polylactic Acid-Polyglycolic Acid Copolymer chemistry
- Abstract
Purpose: We describe the preparation of injectable polymeric paste (IPP) formulations for local and sustained release of drugs. Furthermore, we include the characterization and possible applications of such pastes. Particular attention is paid to characteristics relevant to the successful clinical formulation development, such as viscosity, injectability, degradation, drug release, sterilization, stability performance and pharmacokinetics., Methods: Paste injectability was characterized using measured viscosity and the Hagen-Poiseuille equation to determine injection forces. Drug degradation, release and formulation stability experiments were performed in vitro and drug levels were quantified using HPLC-UV methods. Pharmacokinetic evaluation of sustained-release lidocaine IPPs used five groups of six rats receiving increasing doses subcutaneously. An anti-cancer formulation was evaluated in a subcutaneous tumor xenograft mouse model., Results: The viscosity and injectability of IPPs could be controlled by changing the polymeric composition. IPPs demonstrated good long-term stability and tunable drug-release with low systemic exposure in vivo in rats. Preliminary data in a subcutaneous tumor model points to a sustained anticancer effect., Conclusions: These IPPs are tunable platforms for local and sustained delivery of drugs and have potential for further clinical development to treat a number of diseases.
- Published
- 2020
- Full Text
- View/download PDF
18. Toxoplasma gondii promotes changes in VIPergic submucosal neurons, mucosal intraepithelial lymphocytes, and goblet cells during acute infection in the ileum of rats.
- Author
-
Schneider LCL, do Nascimento JCP, Trevizan AR, Góis MB, Borges SC, Beraldi EJ, Garcia JL, Sant'Ana DMG, and Buttow NC
- Subjects
- Animals, Cell Count, Cell Death physiology, Goblet Cells microbiology, Goblet Cells pathology, Ileum microbiology, Ileum pathology, Intestinal Mucosa metabolism, Intestinal Mucosa microbiology, Intestinal Mucosa pathology, Intraepithelial Lymphocytes microbiology, Intraepithelial Lymphocytes pathology, Male, Myenteric Plexus metabolism, Myenteric Plexus microbiology, Myenteric Plexus pathology, Neurons microbiology, Neurons pathology, Rats, Rats, Wistar, Toxoplasma, Toxoplasmosis microbiology, Toxoplasmosis pathology, Goblet Cells metabolism, Ileum metabolism, Intraepithelial Lymphocytes metabolism, Neurons metabolism, Toxoplasmosis metabolism, Vasoactive Intestinal Peptide metabolism
- Abstract
Background: The intestinal mucosa plays an important role in the mechanical barrier against pathogens. During Toxoplasma gondii infection, however, the parasites invade the epithelial cells of the small intestine and initiate a local immune response. In the submucosal plexus, this response promotes an imbalance of neurotransmitters and induces neuroplasticity, which can change the integrity of the epithelium and its secretory function. This study evaluated the submucosal neurons throughout acute T. gondii infection and the relationship between possible alterations and the epithelial and immune defense cells of the mucosa., Methods: Forty Wistar rats were randomly assigned to 8 groups (n = 5): 1 control group, uninfected, and 7 groups infected with an inoculation of 5000 sporulated T. gondii oocysts (ME-49 strain, genotype II). Segments of the ileum were collected for standard histological processing, histochemical techniques, and immunofluorescence., Key Results: The infection caused progressive neuronal loss in the submucosal general population and changed the proportion of VIPergic neurons throughout the infection periods. These changes may be related to the observed reduction in goblet cells that secret sialomucins and increase in intraepithelial lymphocytes after 24 hours, and the increase in immune cells in the lamina propria after 10 days of infection. The submucosa also presented fibrogenesis, characterizing injury and tissue repair., Conclusions and Inferences: The acute T. gondii infection in the ileum of rats changes the proportion of VIPergic neurons and the epithelial cells, which can compromise the mucosal defense during infection., (© 2017 John Wiley & Sons Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
19. Moving Towards Precision Urologic Oncology: Targeting Enzalutamide-resistant Prostate Cancer and Mutated Forms of the Androgen Receptor Using the Novel Inhibitor Darolutamide (ODM-201).
- Author
-
Borgmann H, Lallous N, Ozistanbullu D, Beraldi E, Paul N, Dalal K, Fazli L, Haferkamp A, Lejeune P, Cherkasov A, and Gleave ME
- Subjects
- Androgen Receptor Antagonists chemistry, Animals, Benzamides, Cell Line, Tumor, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Humans, Male, Mice, Models, Molecular, Molecular Targeted Therapy, Mutation, Nitriles, Phenylthiohydantoin pharmacology, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Protein Conformation, Pyrazoles chemistry, Receptors, Androgen chemistry, Receptors, Androgen genetics, Receptors, Androgen metabolism, Signal Transduction drug effects, Structure-Activity Relationship, Time Factors, Transcription, Genetic drug effects, Tumor Burden drug effects, Xenograft Model Antitumor Assays, Androgen Receptor Antagonists pharmacology, Drug Resistance, Neoplasm genetics, Phenylthiohydantoin analogs & derivatives, Prostatic Neoplasms drug therapy, Pyrazoles pharmacology, Receptors, Androgen drug effects
- Abstract
Darolutamide (ODM-201) is a novel androgen receptor (AR) antagonist with a chemical structure distinctly different from currently approved AR antagonists that targets both wild-type and mutated ligand binding domain variants to inhibit AR nuclear translocation. Here, we evaluate the activity of darolutamide in enzalutamide-resistant castration resistant prostate cancer (CRPC) as well as in AR mutants detected in patients after treatment with enzalutamide, abiraterone, or bicalutamide. Darolutamide significantly inhibited cell growth and AR transcriptional activity in enzalutamide-resistant MR49F cells in vitro, and led to decreased tumor volume and serum prostate-specific antigen levels in vivo, prolonging survival in mice bearing enzalutamide-resistant MR49F xenografts. Moreover, darolutamide inhibited the transcriptional activity of AR mutants identified in the plasma of CRPC patients progressing on traditional therapies. In particular, darolutamide significantly inhibited the transcriptional activity of the F877L, H875Y/T878A, F877L/T878A, and the previously unreported T878G AR mutants, that transform enzalutamide into a partial agonist. In silico cheminformatics computer modeling provided atomic level insights confirming darolutamide antagonist effect in F877L and T878G AR mutants. In conclusion, our results provide a rationale for further clinical evaluation of darolutamide in enzalutamide-resistant CRPC, in particular in combination with circulating tumor DNA assays that detect AR mutants sensitive to darolutamide, in a precision oncology setting., Patient Summary: In this study we evaluated the novel drug darolutamide in preclinical models of prostate cancer. We found that darolutamide delays growth of enzalutamide-resistant prostate cancer, in particular in cells with mutated forms of the androgen receptor after previous treatment. Our data supports further evaluation of darolutamide in clinical trials., (Copyright © 2017. Published by Elsevier B.V.)
- Published
- 2018
- Full Text
- View/download PDF
20. Clusterin knockdown sensitizes prostate cancer cells to taxane by modulating mitosis.
- Author
-
Al Nakouzi N, Wang CK, Beraldi E, Jager W, Ettinger S, Fazli L, Nappi L, Bishop J, Zhang F, Chauchereau A, Loriot Y, and Gleave M
- Subjects
- Cell Line, Tumor, Clusterin genetics, Gene Knockdown Techniques, Humans, Male, Antineoplastic Agents pharmacology, Bridged-Ring Compounds pharmacology, Cell Proliferation drug effects, Clusterin metabolism, Mitosis drug effects, Prostatic Neoplasms pathology, Taxoids pharmacology
- Abstract
Clusterin (CLU) is a stress-activated molecular chaperone that confers treatment resistance to taxanes when highly expressed. While CLU inhibition potentiates activity of taxanes and other anti-cancer therapies in preclinical models, progression to treatment-resistant disease still occurs implicating additional compensatory survival mechanisms. Taxanes are believed to selectively target cells in mitosis, a complex mechanism controlled in part by balancing antagonistic roles of Cdc25C and Wee1 in mitosis progression. Our data indicate that CLU silencing induces a constitutive activation of Cdc25C, which delays mitotic exit and hence sensitizes cancer cells to mitotic-targeting agents such as taxanes. Unchecked Cdc25C activation leads to mitotic catastrophe and cell death unless cells up-regulate protective mechanisms mediated through the cell cycle regulators Wee1 and Cdk1. In this study, we show that CLU silencing induces a constitutive activation of Cdc25C via the phosphatase PP2A leading to relief of negative feedback inhibition and activation of Wee1-Cdk1 to promote survival and limit therapeutic efficacy. Simultaneous inhibition of CLU-regulated cell cycle effector Wee1 may improve synergistic responses of biologically rational combinatorial regimens using taxanes and CLU inhibitors., (© 2016 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2016
- Full Text
- View/download PDF
21. siRNA Lipid Nanoparticle Potently Silences Clusterin and Delays Progression When Combined with Androgen Receptor Cotargeting in Enzalutamide-Resistant Prostate Cancer.
- Author
-
Yamamoto Y, Lin PJ, Beraldi E, Zhang F, Kawai Y, Leong J, Katsumi H, Fazli L, Fraser R, Cullis PR, and Gleave M
- Subjects
- Animals, Antineoplastic Agents pharmacology, Apoptosis genetics, Benzamides, Cell Line, Tumor, Cell Proliferation, Disease Models, Animal, Disease Progression, Drug Resistance, Neoplasm, Gene Expression, Genes, Reporter, Humans, Male, Mice, Molecular Imaging methods, Neoplasm Metastasis, Nitriles, Oligonucleotides, Antisense genetics, Phenylthiohydantoin analogs & derivatives, Phenylthiohydantoin pharmacology, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant pathology, Xenograft Model Antitumor Assays, Clusterin genetics, Gene Silencing, Lipids, Nanoparticles, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, RNA, Small Interfering genetics, Receptors, Androgen genetics
- Abstract
Purpose: Lipid nanoparticle (LNP) formulations facilitate tumor uptake and intracellular processing through an enhanced permeation and retention effect (EPR), and currently multiple products are undergoing clinical evaluation. Clusterin (CLU) is a cytoprotective chaperone induced by androgen receptor (AR) pathway inhibition to facilitate adaptive survival pathway signaling and treatment resistance. In our study, we investigated the efficacy of siRNA tumor delivery using LNP systems in an enzalutamide-resistant (ENZ-R) castration-resistant prostate cancer (CRPC) model., Experimental Design: Gene silencing of a luciferase reporter gene in the PC-3M-luc stable cell line was first assessed in subcutaneous and metastatic PC-3 xenograft tumors. Upon validation, the effect of LNP siRNA targeting CLU in combination with AR antisense oligonucleotides (ASO) was assessed in ENZ-R CRPC LNCaP in vitro and in vivo models., Results: LNP LUC-siRNA silenced luciferase expression in PC-3M-luc subcutaneous xenograft and metastatic models. LNP CLU-siRNA potently suppressed CLU and AR ASO-induced CLU and AKT and ERK phosphorylation in ENZ-R LNCaP cells in vitro, more potently inhibiting ENZ-R cell growth rates and increased apoptosis when compared with AR-ASO monotherapy. In subcutaneous ENZ-R LNCaP xenografts, combinatory treatment of LNP CLU-siRNA plus AR-ASO significantly suppressed tumor growth and serum PSA levels compared with LNP LUC-siRNA (control) and AR-ASO., Conclusions: LNP siRNA can silence target genes in vivo and enable inhibition of traditionally non-druggable genes like CLU and other promising cotargeting approaches in ENZ-R CRPC therapeutics., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
22. Hsp27 Inhibition with OGX-427 Sensitizes Non-Small Cell Lung Cancer Cells to Erlotinib and Chemotherapy.
- Author
-
Lelj-Garolla B, Kumano M, Beraldi E, Nappi L, Rocchi P, Ionescu DN, Fazli L, Zoubeidi A, and Gleave ME
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antineoplastic Combined Chemotherapy Protocols, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, Cell Line, Tumor, Drug Resistance, Neoplasm drug effects, Drug Synergism, Erlotinib Hydrochloride pharmacology, Gene Expression Regulation, Neoplastic drug effects, HSP27 Heat-Shock Proteins antagonists & inhibitors, HSP27 Heat-Shock Proteins genetics, Heat-Shock Proteins, Humans, Lung Neoplasms genetics, Lung Neoplasms metabolism, Male, Mice, Molecular Chaperones, Oligonucleotides pharmacology, Xenograft Model Antitumor Assays, Antineoplastic Agents administration & dosage, Carcinoma, Non-Small-Cell Lung drug therapy, Erlotinib Hydrochloride administration & dosage, HSP27 Heat-Shock Proteins metabolism, Lung Neoplasms drug therapy, Oligonucleotides administration & dosage
- Abstract
Non-small cell lung cancer (NSCLC) is the most frequent cause of death from cancer worldwide. Despite the availability of active chemotherapy regimens and EGFR tyrosine kinase inhibitors, all advanced patients develop recurrent disease after first-line therapy. Although Hsp27 is a stress-induced chaperone that promotes acquired resistance in several cancers, its relationship to treatment resistance in NSCLC has not been defined. Understanding adaptive responses of acquired resistance will help guide new strategies to control NSCLC. Hsp27 levels were evaluated in an HCC827 erlotinib-resistant-derived cell line (HCC-827Resistant), and sensitivity to erlotinib was examined in Hsp27-overexpressing A549 cells. The role of Hsp27 in both erlotinib and cytotoxic treatment resistance was evaluated in HCC-827 and A549 NSCLC cells using the Hsp27 antisense drug OGX-427. The effect of OGX-427 in combination with erlotinib was also assessed in mice bearing A549 xenografts. Hsp27 is induced by erlotinib and protects NSCLC cells from treatment-induced apoptosis, whereas OGX-427 sensitizes NSCLC cells to erlotinib. Interestingly, increased resistance to erlotinib was observed when Hsp27 was increased either in HCC827 erlotinib-resistant or overexpressing A549 cells. Combining OGX-427 with erlotinib significantly enhanced antitumor effects in vitro and delayed A549 xenograft growth in vivo. OGX-427 also significantly enhanced the activity of cytotoxic drugs used for NSCLC. These data indicate that treatment-induced Hsp27 contributes to the development of resistance, and provides preclinical proof-of-principle that inhibition of stress adaptive pathways mediated by Hsp27 enhances the activity of erlotinib and chemotherapeutics., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
23. Inhibition of the HER2-YB1-AR axis with Lapatinib synergistically enhances Enzalutamide anti-tumor efficacy in castration resistant prostate cancer.
- Author
-
Shiota M, Bishop JL, Takeuchi A, Nip KM, Cordonnier T, Beraldi E, Kuruma H, Gleave ME, and Zoubeidi A
- Subjects
- Adenocarcinoma pathology, Animals, Apoptosis drug effects, Benzamides, Cell Division drug effects, Cell Line, Tumor, Drug Resistance, Neoplasm, Drug Synergism, ErbB Receptors antagonists & inhibitors, Humans, Lapatinib, Male, Mice, Mice, Nude, Nitriles, Orchiectomy, Phenylthiohydantoin pharmacology, Prostatic Neoplasms pathology, Protein Kinase Inhibitors pharmacology, Protein Processing, Post-Translational drug effects, Protein Transport drug effects, Proto-Oncogene Proteins c-akt metabolism, Receptors, Androgen drug effects, Signal Transduction drug effects, Adenocarcinoma drug therapy, Androgen Receptor Antagonists pharmacology, Neoplasm Proteins antagonists & inhibitors, Phenylthiohydantoin analogs & derivatives, Prostatic Neoplasms drug therapy, Quinazolines pharmacology, Receptor, ErbB-2 antagonists & inhibitors, Y-Box-Binding Protein 1 antagonists & inhibitors
- Abstract
Incurable castration-resistant prostate cancer (CRPC) is driven by androgen receptor (AR) activation. Potent therapies that prevent AR signaling, such as Enzalutamide (ENZ), are mainstay treatments for CRPC; however patients eventually progress with ENZ resistant (ENZR) disease. In this study, we investigated one mechanism of ENZ resistance, and tried to improve therapeutic efficiency of ENZ. We found HER2 expression is increased in ENZR tumors and cell lines, and is induced by ENZ treatment of LNCaP cells. ENZ-induced HER2 overexpression was dependent on AKT-YB1 activation and modulated AR activity. HER2 dependent AR activation in LNCaP and ENZR cells was effectively blocked by treatment with the EGFR/HER2 inhibitor Lapatinib, which reduced cell viability and increased apoptosis. Despite efficacy in vitro, in vivo monotherapy with Lapatinib did not prevent ENZR tumor growth. However, combination treatment of Lapatinib with ENZ most effectively induced cell death in LNCaP cells in vitro and was more effective than ENZ alone in preventing tumor growth in an in vivo model of CRPC. These results suggest that while HER2 overexpression and subsequent AR activation is a targetable mechanism of resistance to ENZ, therapy using Lapatinib is only a rational therapeutic approach when used in combination with ENZ in CRPC.
- Published
- 2015
- Full Text
- View/download PDF
24. Generation 2.5 antisense oligonucleotides targeting the androgen receptor and its splice variants suppress enzalutamide-resistant prostate cancer cell growth.
- Author
-
Yamamoto Y, Loriot Y, Beraldi E, Zhang F, Wyatt AW, Al Nakouzi N, Mo F, Zhou T, Kim Y, Monia BP, MacLeod AR, Fazli L, Wang Y, Collins CC, Zoubeidi A, and Gleave M
- Subjects
- Animals, Benzamides, Blotting, Western, Humans, Immunohistochemistry, Male, Mice, Nitriles, Phenylthiohydantoin pharmacology, Prostatic Neoplasms, Castration-Resistant drug therapy, Protein Isoforms, RNA, Small Interfering, Reverse Transcriptase Polymerase Chain Reaction, Transfection, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm genetics, Oligonucleotides, Antisense pharmacology, Phenylthiohydantoin analogs & derivatives, Prostatic Neoplasms, Castration-Resistant genetics, Receptors, Androgen genetics
- Abstract
Purpose: Enzalutamide (ENZ) is a potent androgen receptor (AR) antagonist with activity in castration-resistant prostate cancer (CRPC); however, progression to ENZ-resistant (ENZ-R) CRPC frequently occurs with rising serum PSA levels, implicating AR full-length (ARFL) or variants (AR-Vs) in disease progression., Experimental Design: To define functional roles of ARFL and AR-Vs in ENZ-R CRPC, we designed 3 antisense oligonucleotides (ASO) targeting exon-1, intron-1, and exon-8 in AR pre-mRNA to knockdown ARFL alone or with AR-Vs, and examined their effects in three CRPC cell lines and patient-derived xenografts., Results: ENZ-R-LNCaP cells express high levels of both ARFL and AR-V7 compared with CRPC-LNCaP; in particular, ARFL levels were approximately 12-fold higher than AR-V7. Both ARFL and AR-V7 are highly expressed in the nuclear fractions of ENZ-R-LNCaP cells even in the absence of exogenous androgens. In ENZ-R-LNCaP cells, knockdown of ARFL alone, or ARFL plus AR-Vs, similarly induced apoptosis, suppressed cell growth and AR-regulated gene expression, and delayed tumor growth in vivo. In 22Rv1 cells that are inherently ENZ-resistant, knockdown of both ARFL and AR-Vs more potently suppressed cell growth, AR transcriptional activity, and AR-regulated gene expression than knockdown of ARFL alone. Exon-1 AR-ASO also inhibited tumor growth of LTL-313BR patient-derived CRPC xenografts., Conclusions: These data identify the AR as an important driver of ENZ resistance, and while the contributions of ARFL and AR-Vs can vary across cell systems, ARFL is the key driver in the ENZ-R LNCaP model. AR targeting strategies against both ARFL and AR-Vs is a rational approach for AR-dependent CRPC., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
25. A Novel Triazole Nucleoside Suppresses Prostate Cancer Cell Growth by Inhibiting Heat Shock Factor 1 and Androgen Receptor.
- Author
-
Xia Y, Wang M, Beraldi E, Cong M, Zoubeidi A, Gleave M, and Peng L
- Subjects
- Cell Line, Tumor, Humans, Male, Prostatic Neoplasms metabolism, Heat-Shock Proteins metabolism, Prostatic Neoplasms pathology, Receptors, Androgen metabolism, Triazoles pharmacology
- Abstract
A novel triazole nucleoside analogue was discovered to exhibit potent anticancer activity in prostate cancer cells via down-regulating heat shock factor 1 (HSF1) and related heat shock proteins, along with the consequential inhibition of androgen receptor (AR) expression and transactivation, arresting the cell cycle in AR-governed phase. This triazole nucleoside therefore constitutes a novel structural paradigm and potential drug candidate for prostate cancer through inhibition of HSF1 and AR.
- Published
- 2015
- Full Text
- View/download PDF
26. Clusterin facilitates stress-induced lipidation of LC3 and autophagosome biogenesis to enhance cancer cell survival.
- Author
-
Zhang F, Kumano M, Beraldi E, Fazli L, Du C, Moore S, Sorensen P, Zoubeidi A, and Gleave ME
- Subjects
- Animals, Apoptosis drug effects, Apoptosis genetics, Autophagy drug effects, Autophagy genetics, Autophagy-Related Proteins, Cell Line, Tumor, Cell Survival drug effects, Clusterin antagonists & inhibitors, Clusterin deficiency, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Humans, Lipid Metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Nude, Microtubule-Associated Proteins antagonists & inhibitors, Microtubule-Associated Proteins metabolism, Phagosomes drug effects, Phagosomes pathology, Prostatic Neoplasms drug therapy, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Pyrimidines pharmacology, Pyrroles pharmacology, Signal Transduction, Thionucleotides genetics, Thionucleotides metabolism, Ubiquitin-Conjugating Enzymes genetics, Ubiquitin-Conjugating Enzymes metabolism, Xenograft Model Antitumor Assays, Clusterin genetics, Gene Expression Regulation, Neoplastic, Microtubule-Associated Proteins genetics, Phagosomes metabolism, Prostatic Neoplasms genetics
- Abstract
We define stress-induced adaptive survival pathways linking autophagy with the molecular chaperone clusterin (CLU) that function to promote anticancer treatment resistance. During treatment stress, CLU co-localizes with LC3 via an LIR-binding sequence within autophagosome membranes, functioning to facilitate LC3-Atg3 heterocomplex stability and LC3 lipidation, and thereby enhance autophagosome biogenesis and autophagy activation. Stress-induced autophagy is attenuated with CLU silencing in CLU(-/-) mice and human prostate cancer cells. CLU-enhanced cell survival occurs via autophagy-dependent pathways, and is reduced following autophagy inhibition. Combining CLU inhibition with anticancer treatments attenuates autophagy activation, increases apoptosis and reduces prostate cancer growth. This study defines a novel adaptor protein function for CLU under stress conditions, and highlights how co-targeting CLU and autophagy can amplify proteotoxic stress to delay cancer progression.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.