Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Clin Invest ; 133(21)2023 11 01.
Article in English | MEDLINE | ID: mdl-37651197

ABSTRACT

The triggering receptor expressed on myeloid cell 1 (TREM1) plays a critical role in development of chronic inflammatory disorders and the inflamed tumor microenvironment (TME) associated with most solid tumors. We examined whether loss of TREM1 signaling can abrogate the immunosuppressive TME and enhance cancer immunity. To investigate the therapeutic potential of TREM1 in cancer, we used mice deficient in Trem1 and developed a novel small molecule TREM1 inhibitor, VJDT. We demonstrated that genetic or pharmacological TREM1 silencing significantly delayed tumor growth in murine melanoma (B16F10) and fibrosarcoma (MCA205) models. Single-cell RNA-Seq combined with functional assays during TREM1 deficiency revealed decreased immunosuppressive capacity of myeloid-derived suppressor cells (MDSCs) accompanied by expansion in cytotoxic CD8+ T cells and increased PD-1 expression. Furthermore, TREM1 inhibition enhanced the antitumorigenic effect of anti-PD-1 treatment, in part, by limiting MDSC frequency and abrogating T cell exhaustion. In patient-derived melanoma xenograft tumors, treatment with VJDT downregulated key oncogenic signaling pathways involved in cell proliferation, migration, and survival. Our work highlights the role of TREM1 in cancer progression, both intrinsically expressed in cancer cells and extrinsically in the TME. Thus, targeting TREM1 to modify an immunosuppressive TME and improve efficacy of immune checkpoint therapy represents what we believe to be a promising therapeutic approach to cancer.


Subject(s)
Melanoma , Myeloid-Derived Suppressor Cells , Humans , Animals , Mice , Triggering Receptor Expressed on Myeloid Cells-1/genetics , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Myeloid Cells/pathology , Cell Line, Tumor , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , T-Lymphocytes, Cytotoxic/pathology , Disease Models, Animal , Tumor Microenvironment
3.
Hum Immunol ; 84(8): 366-373, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36934068

ABSTRACT

Despite the growing interest in the role of regulatory B cells (Bregs) in autoimmunity, their distinct role and function in kidney transplant outcomes remain elusive. Here, we retrospectively analyzed the proportion of Bregs, transitional Bregs (tBregs) and memory Bregs (mBregs) and their capacity to produce IL-10 in non-rejected (NR) versus rejected (RJ) kidney transplant recipients. In the NR group, we observed a significant increase in the proportion of mBregs (CD19+CD24hiCD27+) but no difference in tBregs (CD19+CD24hiCD38+), as compared to the RJ group. We also observed a significant increase in IL-10-producing mBregs (CD19+CD24hiCD27+IL-10+) in the NR group. As our group and others have previously reported a potential role of the human leukocyte antigen G (HLA-G) in human renal allograft survival, notably through IL-10, we then investigated possible crosstalk between HLA-G and IL-10+ mBregs. Our ex vivo data suggest a role of HLA-G in enhancing IL-10+ mBreg expansion upon stimulation, which further decreased CD3+ T cell proliferation capability. Using RNA-sequencing (RNA-seq), we identified potential key signaling pathways involved in HLA-G-driven IL-10+ mBreg expansion, such as the MAPK, TNF and chemokine signaling pathways. Together, our study highlights a novel HLA-G-mediated IL-10-producing mBreg pathway that may serve as a therapeutic target to improve kidney allograft survival.


Subject(s)
B-Lymphocytes, Regulatory , Kidney Transplantation , Humans , HLA-G Antigens/metabolism , Interleukin-10/metabolism , Retrospective Studies , Kidney , Allografts
4.
Neoplasia ; 23(12): 1261-1274, 2021 12.
Article in English | MEDLINE | ID: mdl-34781084

ABSTRACT

A major challenge to the treatment of advanced prostate cancer (PCa) is the development of resistance to androgen-deprivation therapy (ADT) and chemotherapy. It is imperative to discover effective therapies to overcome drug resistance and improve clinical outcomes. We have developed a novel class of silicon-containing compounds and evaluated the anticancer activities and mechanism of action using cellular and animal models of drug-resistant PCa. Five organosilicon compounds were evaluated for their anticancer activities in the NCI-60 panel and established drug-resistant PCa cell lines. GH1504 exhibited potent in vitro cytotoxicity in a broad spectrum of human cancer cells, including PCa cells refractory to ADT and chemotherapy. Molecular studies identified several potential targets of GH1504, most notably androgen receptor (AR), AR variant 7 (AR-v7) and survivin. Mechanistically, GH1504 may promote the protein turnover of AR, AR-v7 and survivin, thereby inducing apoptosis in ADT-resistant and chemoresistant PCa cells. Animal studies demonstrated that GH1504 effectively inhibited the in vivo growth of ADT-resistant CWR22Rv1 and chemoresistant C4-2B-TaxR xenografts in subcutaneous and intraosseous models. These preclinical results indicated that GH1504 is a promising lead that can be further developed as a novel therapy for drug-resistant PCa.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Organosilicon Compounds/pharmacology , Prostatic Neoplasms, Castration-Resistant , Animals , Cell Line , Drug Screening Assays, Antitumor , Humans , Male , Mice , Xenograft Model Antitumor Assays
5.
Theranostics ; 11(14): 6873-6890, 2021.
Article in English | MEDLINE | ID: mdl-34093859

ABSTRACT

Rationale: Chemoresistance is a major obstacle in prostate cancer (PCa) treatment. We sought to understand the underlying mechanism of PCa chemoresistance and discover new treatments to overcome docetaxel resistance. Methods: We developed a novel phenotypic screening platform for the discovery of specific inhibitors of chemoresistant PCa cells. The mechanism of action of the lead compound was investigated using computational, molecular and cellular approaches. The in vivo toxicity and efficacy of the lead compound were evaluated in clinically-relevant animal models. Results: We identified LG1980 as a lead compound that demonstrates high selectivity and potency against chemoresistant PCa cells. Mechanistically, LG1980 binds embryonic ectoderm development (EED), disrupts the interaction between EED and enhancer of zeste homolog 2 (EZH2), thereby inducing the protein degradation of EZH2 and inhibiting the phosphorylation and activity of EZH2. Consequently, LG1980 targets a survival signaling cascade consisting of signal transducer and activator of transcription 3 (Stat3), S-phase kinase-associated protein 2 (SKP2), ATP binding cassette B 1 (ABCB1) and survivin. As a lead compound, LG1980 is well tolerated in mice and effectively suppresses the in vivo growth of chemoresistant PCa and synergistically enhances the efficacy of docetaxel in xenograft models. Conclusions: These results indicate that pharmacological inhibition of EED-EZH2 interaction is a novel strategy for the treatment of chemoresistant PCa. LG1980 and its analogues have the potential to be integrated into standard of care to improve clinical outcomes in PCa patients.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Drug Discovery/methods , Drug Resistance, Neoplasm , Enhancer of Zeste Homolog 2 Protein/metabolism , Polycomb Repressive Complex 2/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Line, Tumor , Cell Survival/drug effects , Docetaxel/pharmacology , Docetaxel/therapeutic use , Drug Synergism , Humans , Inhibitory Concentration 50 , Male , Mice , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , S-Phase Kinase-Associated Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Survivin/metabolism , Xenograft Model Antitumor Assays
6.
Cancers (Basel) ; 13(8)2021 Apr 10.
Article in English | MEDLINE | ID: mdl-33920080

ABSTRACT

ß-arrestin 1 (ARRB1) is a scaffold protein that regulates signaling downstream of G protein-coupled receptors (GPCRs). In the current work, we investigated the role of ARRB1 in regulating the metabolic preference of cancer stem cell (CSC)-like cells in bladder cancer (BC). We show that ARRB1 is crucial for spheroid formation and tumorigenic potential. Furthermore, we measured mitochondrial respiration, glucose uptake, glycolytic rate, mitochondrial/glycolytic ATP production and fuel oxidation in previously established ARRB1 knock out (KO) cells and corresponding controls. Our results demonstrate that depletion of ARRB1 decreased glycolytic rate and induced metabolic reprogramming towards oxidative phosphorylation. Mechanistically, the depletion of ARRB1 dramatically increased the mitochondrial pyruvate carrier MPC1 protein levels and reduced the glucose transporter GLUT1 protein levels along with glucose uptake. Overexpression of ARRB1 in ARRB1 KO cells reversed the phenotype and resulted in the upregulation of glycolysis. In conclusion, we show that ARRB1 regulates the metabolic preference of BC CSC-like cells and functions as a molecular switch that promotes reprogramming towards glycolysis by negatively regulating MPC1 and positively regulating GLUT1/ glucose uptake. These observations open new therapeutic avenues for targeting the metabolic preferences of cancer stem cell (CSC)-like BC cells.

7.
Int J Mol Sci ; 22(5)2021 Feb 28.
Article in English | MEDLINE | ID: mdl-33671107

ABSTRACT

Metabolic reprogramming is a hallmark of malignancy. It implements profound metabolic changes to sustain cancer cell survival and proliferation. Although the Warburg effect is a common feature of metabolic reprogramming, recent studies have revealed that tumor cells also depend on mitochondrial metabolism. Due to the essential role of mitochondria in metabolism and cell survival, targeting mitochondria in cancer cells is an attractive therapeutic strategy. However, the metabolic flexibility of cancer cells may enable the upregulation of compensatory pathways, such as glycolysis, to support cancer cell survival when mitochondrial metabolism is inhibited. Thus, compounds capable of targeting both mitochondrial metabolism and glycolysis may help overcome such resistance mechanisms. Normal prostate epithelial cells have a distinct metabolism as they use glucose to sustain physiological citrate secretion. During the transformation process, prostate cancer cells consume citrate to mainly power oxidative phosphorylation and fuel lipogenesis. A growing number of studies have assessed the impact of triterpenoids on prostate cancer metabolism, underlining their ability to hit different metabolic targets. In this review, we critically assess the metabolic transformations occurring in prostate cancer cells. We will then address the opportunities and challenges in using triterpenoids as modulators of prostate cancer cell metabolism.


Subject(s)
Glycolysis , Mitochondria/drug effects , Oxidative Phosphorylation , Oxidative Stress , Prostatic Neoplasms/drug therapy , Triterpenes/pharmacology , Animals , Humans , Male , Mitochondria/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
8.
Int J Mol Sci ; 21(23)2020 Dec 07.
Article in English | MEDLINE | ID: mdl-33297302

ABSTRACT

ß-Arrestins (ARRBs) are ubiquitously expressed scaffold proteins that mediate inactivation of G-protein-coupled receptor signaling, and in certain circumstances, G-protein independent pathways. Intriguingly, the two known ARRBs, ß-arrestin1 (ARRB1) and ß-Arrestin2 (ARRB2), seem to have opposing functions in regulating signaling cascades in several models in health and disease. Recent evidence suggests that ARRBs are implicated in regulating stem cell maintenance; however, their role, although crucial, is complex, and there is no universal model for ARRB-mediated regulation of stem cell characteristics. For the first time, this review compiles information on the function of ARRBs in stem cell biology and will discuss the role of ARRBs in regulating cell signaling pathways implicated in stem cell maintenance in normal and malignant stem cell populations. Although promising targets for cancer therapy, the ubiquitous nature of ARRBs and the plethora of functions in normal cell biology brings challenges for treatment selectivity. However, recent studies show promising evidence for specifically targeting ARRBs in myeloproliferative neoplasms.


Subject(s)
Carcinogenesis/metabolism , Neoplastic Stem Cells/metabolism , beta-Arrestins/metabolism , Animals , Carcinogenesis/genetics , Cell Self Renewal , Humans , Neoplastic Stem Cells/physiology , Phenotype , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/physiology , beta-Arrestins/genetics
9.
Prostate ; 80(12): 993-1005, 2020 09.
Article in English | MEDLINE | ID: mdl-32559345

ABSTRACT

BACKGROUND: Androgen deprivation therapy (ADT) is the mainstay of treatment for castration-resistant prostate cancer (CRPC). Unfortunately, although ADT initially prolongs survival, most patients relapse and develop resistance. Clinical failure of these treatments in CRPC highlights the urgent need to develop novel strategies to more effectively block androgen receptor (AR) signaling and target other oncogenic factors responsible for ADT resistance. METHODS: We developed a small-molecule compound LG1836 and investigated the in vitro and in vivo activity of LG1836 against CRPC in cellular and animal models. RESULTS: LG1836 exhibits potent in vitro cytotoxicity in CRPC cells. Mechanistic studies demonstrated that LG1836 inhibits the expression of AR and AR variant 7, partially mediated via proteasome-dependent protein degradation. LG1836 also suppresses survivin expression and effectively induces apoptosis in CRPC cells. Significantly, as a single agent, LG1836 is therapeutically efficacious in suppressing the in vivo growth of CRPC in the subcutaneous and intraosseous models and extends the survival of tumor-bearing mice. CONCLUSIONS: These preclinical studies indicate that LG1836 is a promising lead compound for the treatment of CRPC.


Subject(s)
Piperidines/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Small Molecule Libraries/pharmacology , Androgen Receptor Antagonists/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Growth Processes/drug effects , HSP90 Heat-Shock Proteins/metabolism , Humans , Male , Mice , Mice, Nude , Mice, SCID , Prostatic Neoplasms, Castration-Resistant/pathology , Random Allocation , Receptors, Androgen/biosynthesis , Receptors, Androgen/metabolism , Survivin/antagonists & inhibitors , Survivin/biosynthesis , Ubiquitination , Xenograft Model Antitumor Assays
10.
Cancer Lett ; 446: 62-72, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30660650

ABSTRACT

Bone metastasis is a major cause of prostate cancer (PCa) mortality. Although docetaxel chemotherapy initially extends patients' survival, in most cases PCa becomes chemoresistant and eventually progresses without a cure. In this study, we developed a novel small-molecule compound BKM1972, which exhibited potent in vitro cytotoxicity in PCa and other cancer cells regardless of their differences in chemo-responsiveness. Mechanistic studies demonstrated that BKM1972 effectively inhibited the expression of anti-apoptotic protein survivin and membrane-bound efflux pump ATP binding cassette B 1 (ABCB1, p-glycoprotein), presumably via signal transducer and activator of transcription 3 (Stat3). BKM1972 was well tolerated in mice and as a monotherapy, significantly inhibited the intraosseous growth of chemosensitive and chemoresistant PCa cells. These results indicate that BKM1972 is a promising small-molecule lead to treat PCa bone metastasis and overcome docetaxel resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Bradykinin Receptor Antagonists/pharmacology , Cell Proliferation/drug effects , Docetaxel/pharmacology , Drug Resistance, Neoplasm , Organophosphonates/pharmacology , Prostatic Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Line, Tumor , Humans , Male , Mice, Nude , Molecular Targeted Therapy , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Survivin/genetics , Survivin/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Cell Death Dis ; 9(9): 931, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30209297

ABSTRACT

RHO GTPases regulate essential functions such as the organization of the actin cytoskeleton. The classic members cycle between an active GTP-bound and an inactive GDP-bound conformation whereas atypical members are predominantly GTP-bound. Besides their well-established role, the classic RHO GTPases RHOB and RAC1, are rapidly induced and/or activated by genotoxic stress and contribute to the DNA damage response. Here we used camptothecin, a selective topoisomerase I (TOP1) inhibitor that stabilizes TOP1 cleavage complexes (TOP1cc), to search for other potential early DNA damage-inducible RHO GTPase genes. We identified that an atypical RHO GTPase, RND1, is rapidly induced by camptothecin. RND1 induction is closely associated with the presence of TOP1cc induced by camptothecin or by DNA lesions that elevate TOP1cc levels such as UV and hydrogen peroxide. We further demonstrated that camptothecin increases RND1 gene transcription and mRNA stability. Camptothecin also increases poly(ADP-ribose) polymerase 1 (PARP-1) activity, whose inhibition reduces RND1 transcription. In addition, overexpression of RND1 increases PARP-1, suggesting a cross-talk between PARP-1 and RND1. Finally, RND1 protects cells against camptothecin-induced apoptosis, and hence favors cellular resistance to camptothecin. Together, these findings highlight RND1 as an atypical RHO GTPase early induced by TOP1cc, and show that the TOP1cc-PARP-1-RND1 pathway protects cells against apoptosis induced by camptothecin.


Subject(s)
Camptothecin/pharmacology , DNA Topoisomerases, Type I/genetics , DNA/genetics , Drug Resistance, Neoplasm/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Transcription, Genetic/genetics , rho GTP-Binding Proteins/genetics , Animals , Cell Line , Cell Line, Tumor , DNA Damage/drug effects , DNA Damage/genetics , HCT116 Cells , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Mice , NIH 3T3 Cells , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Topoisomerase I Inhibitors/pharmacology
12.
Neoplasia ; 20(8): 789-799, 2018 08.
Article in English | MEDLINE | ID: mdl-29981500

ABSTRACT

The high prevalence and long latency period of prostate cancer (PCa) provide a unique opportunity to control disease progression with dietary and nutraceutical approaches. We developed ProFine, a standardized composition of luteolin, quercetin, and kaempferol, and investigated its potential as a nutraceutical for PCa in preclinical models. The three ingredients of ProFine demonstrated synergistic in vitro cytotoxicity and effectively induced apoptosis in PCa cells. ProFine markedly affected the transcriptome of PCa cells, suppressed the expression of androgen receptor, and inhibited androgen-regulated genes. Oral administration of ProFine did not exhibit obvious toxicities in mice, and the three ingredients retained their individual pharmacokinetic and bioavailability profiles. Importantly, ProFine significantly retarded the growth of PCa xenografts in athymic nude mice and extended the survival of animals. This study provides preclinical evidence supporting the promise of ProFine as a safe, efficacious, and affordable intervention to control PCa progression and improve clinical outcomes.


Subject(s)
Cell Proliferation/drug effects , Flavonoids/pharmacology , Prostatic Neoplasms/drug therapy , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Androgens/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude , Transcriptome/drug effects , Xenograft Model Antitumor Assays/methods
13.
Mol Cancer Ther ; 17(9): 1859-1870, 2018 09.
Article in English | MEDLINE | ID: mdl-29907594

ABSTRACT

Docetaxel resistance remains a major obstacle in the treatment of prostate cancer bone metastasis. In this study, we demonstrate that the dopamine D2 receptor (DRD2) agonist bromocriptine effectively enhances docetaxel efficacy and suppresses skeletal growth of prostate cancer in preclinical models. DRD2 is ubiquitously expressed in prostate cancer cell lines and significantly reduced in prostate cancer tissues with high Gleason score. Bromocriptine has weak to moderate cytotoxicity in prostate cancer cells, but effectively induces cell-cycle arrest. At the molecular level, bromocriptine inhibits the expression of c-Myc, E2F-1, and survivin and increases the expression of p53, p21, and p27. Intriguingly, bromocriptine markedly reduces androgen receptor levels, partially through Hsp90-mediated protein degradation. The combination of bromocriptine and docetaxel demonstrates enhanced in vitro cytotoxicity in prostate cancer cells and significantly retards the skeletal growth of C4-2-Luc tumors in mice. Collectively, these results provide the first experimental evidence for repurposing bromocriptine as an effective adjunct therapy to enhance docetaxel efficacy in prostate cancer. Mol Cancer Ther; 17(9); 1859-70. ©2018 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/drug therapy , Drug Repositioning , Prostatic Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Bromocriptine/administration & dosage , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Docetaxel/administration & dosage , Dopamine Agonists/administration & dosage , Drug Synergism , Humans , Male , Mice, Nude , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/metabolism
14.
Biomed Chromatogr ; 32(2)2018 Feb.
Article in English | MEDLINE | ID: mdl-28801923

ABSTRACT

A simple and sensitive liquid chromatography/electrospray ionization tandem mass spectrometry (LC-ESI-MS/MS) method was developed and validated for determination of two highly lipophilic anticancer drug candidates, LG1980 and GH501, in rat plasma and tissues (liver, kidney and femur bones). LG1980 and GH501 were extracted from rat plasma and tissue homogenates using liquid-liquid extraction. The method provided a linear range of 1.0-200.0 ng/mL for GH501 in plasma and LG1980 in plasma and liver. For both analytes in other tissue homogenates the linear range was 2.0-400.0 ng/mL. The method was validated with precision within 15% relative standard deviation, accuracy within 15% relative error and a consistent recovery. This method has been successfully applied in two preclinical studies for LG1980 and GH501 to determine their concentrations in rat plasma, liver, kidney and bone over 24 h after intravenous injection of compounds.


Subject(s)
Antineoplastic Agents/analysis , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Hydrophobic and Hydrophilic Interactions , Kidney/chemistry , Limit of Detection , Linear Models , Liver/chemistry , Male , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Tissue Distribution
15.
Anticancer Res ; 37(11): 6235-6243, 2017 11.
Article in English | MEDLINE | ID: mdl-29061806

ABSTRACT

BACKGROUND: Mifepristone has gained great interest in its potential as a novel agent against human cancers, including prostate cancer (PCa). However, recent clinical trials using mifepristone in PCa and other cancers have been disappointing. We evaluated the in vitro and in vivo activities of mifepristone, in combination with docetaxel and enzalutamide, against bone metastatic castration-resistant PCa. MATERIALS AND METHODS: The effects of mifepristone, alone or in combination with docetaxel or enzalutamide, on PCa cell viability, in vitro, were determined by the colorimetric assay. Intratibial model of C4-2-Luc tumors in athymic nude mice was used to evaluate the in vivo efficacy of mifepristone alone or in combination with docetaxel or enzalutamide. Tumor growth in mouse bone was assessed by serum prostate-specific antigen (PSA) levels and radiography. RESULTS: Although mifepristone exhibits a certain degree of synergism with docetaxel or enzalutamide in cell culture, statistical analyses showed that combination regimens fail to demonstrate effectiveness in suppressing the skeletal growth of PCa and enhancing the in vivo efficacy of docetaxel or enzalutamide in athymic nude mice (p>0.05). CONCLUSION: These results provide the first pre-clinical evidence suggesting that mifepristone may not effectively inhibit bone metastatic PCa, either as a single agent or combined with standard chemotherapy and androgen-deprivation therapy. This report may raise concerns over the clinical use of mifepristone in the management of advanced PCa.


Subject(s)
Bone Neoplasms/drug therapy , Mifepristone/pharmacology , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant/drug therapy , Taxoids/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzamides , Bone Neoplasms/secondary , Cell Proliferation/drug effects , Docetaxel , Hormone Antagonists/pharmacology , Humans , Male , Mice , Nitriles , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms, Castration-Resistant/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Oncotarget ; 7(19): 27489-98, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27050371

ABSTRACT

Bone metastasis is a major cause of prostate cancer (PCa) morbidity and mortality. Despite some success in transiently controlling clinical symptoms with docetaxel-based therapy, PCa patients become docetaxel-resistant and inevitably progress with no cure. We synthesized an acyl-tyrosine bisphosphonate amide derivative, BKM1644, with the intent of targeting bone metastatic PCa and enhancing docetaxel's efficacy. BKM1644 exhibits potent anti-cancer activity in the NCI-60 panel and effectively inhibits the proliferation of metastatic, castration-resistant PCa (mCRPC) cells, with IC50 ranging between 2.1 µM and 6.3 µM. Significantly, BKM1644 sensitizes mCRPC cells to docetaxel treatment. Mice with pre-established C4-2 tumors in the tibia show a marked decrease in serum prostate-specific antigen (control: 173.72 ± 37.52 ng/ml, combined treatment: 64.45 ± 22.19 ng/ml; p < 0.0001) and much improved bone architecture after treatment with the combined regimen. Mechanistic studies found that docetaxel temporarily but significantly increases survivin, an anti-apoptotic protein whose overexpression has been correlated with PCa bone metastasis and therapeutic resistance. Intriguingly, BKM1644 effectively inhibits survivin expression, which may antagonize docetaxel-induced survivin in bone metastatic PCa cells. Signal transducer and activator of transcription 3 (Stat3) may be involved in the suppression of survivin transcription by BKM1644, as confirmed by a survivin reporter assay. Collectively, these data indicate that BKM1644 could be a promising small-molecule agent to improve docetaxel efficacy and retard the bone metastatic growth of PCa.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Diphosphonates/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Taxoids/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Docetaxel , Drug Interactions , Humans , Male , Mice , Mice, Nude , Neoplasm Metastasis , Random Allocation , Xenograft Model Antitumor Assays
17.
Nanoscale ; 8(25): 12668-82, 2016 Jul 07.
Article in English | MEDLINE | ID: mdl-26961235

ABSTRACT

Prostate-specific membrane antigen (PSMA) is one of the most specific cell surface markers for prostate cancer diagnosis and targeted treatment. However, achieving molecular imaging using non-invasive MRI with high resolution has yet to be achieved due to the lack of contrast agents with significantly improved relaxivity for sensitivity, targeting capabilities and metal selectivity. We have previously reported our creation of a novel class of protein Gd(3+) contrast agents, ProCA32, which displayed significantly improved relaxivity while exhibiting strong Gd(3+) binding selectivity over physiological metal ions. In this study, we report our effort in further developing biomarker-targeted protein MRI contrast agents for molecular imaging of PSMA. Among three PSMA targeted contrast agents engineered with addition of different molecular recognition sequences, ProCA32.PSMA exhibits a binding affinity of 1.1 ± 0.1 µM for PSMA while the metal binding affinity is maintained at 0.9 ± 0.1 × 10(-22) M. In addition, ProCA32.PSMA exhibits r1 of 27.6 mM(-1) s(-1) and r2 of 37.9 mM(-1) s(-1) per Gd (55.2 and 75.8 mM(-1) s(-1) per molecule r1 and r2, respectively) at 1.4 T. At 7 T, ProCA32.PSMA also has r2 of 94.0 mM(-1) s(-1) per Gd (188.0 mM(-1) s(-1) per molecule) and r1 of 18.6 mM(-1) s(-1) per Gd (37.2 mM(-1) s(-1) per molecule). This contrast capability enables the first MRI enhancement dependent on PSMA expression levels in tumor bearing mice using both T1 and T2-weighted MRI at 7 T. Further development of these PSMA-targeted contrast agents are expected to be used for the precision imaging of prostate cancer at an early stage and to monitor disease progression and staging, as well as determine the effect of therapeutic treatment by non-invasive evaluation of the PSMA level using MRI.


Subject(s)
Antigens, Surface/analysis , Contrast Media , Glutamate Carboxypeptidase II/analysis , Magnetic Resonance Imaging , Molecular Imaging , Neoplasms, Experimental/diagnostic imaging , Prostatic Neoplasms/diagnostic imaging , Animals , Cell Line, Tumor , Gadolinium , Humans , Male , Mice , Mice, Nude
18.
Mol Cell Biol ; 34(16): 3144-55, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24912678

ABSTRACT

Unlike other Rho GTPases, RhoB is rapidly induced by DNA damage, and its expression level decreases during cancer progression. Because inefficient repair of DNA double-strand breaks (DSBs) can lead to cancer, we investigated whether camptothecin, an anticancer drug that produces DSBs, induces RhoB expression and examined its role in the camptothecin-induced DNA damage response. We show that in camptothecin-treated cells, DSBs induce RhoB expression by a mechanism that depends notably on Chk2 and its substrate HuR, which binds to RhoB mRNA and protects it against degradation. RhoB-deficient cells fail to dephosphorylate γH2AX following camptothecin removal and show reduced efficiency of DSB repair by homologous recombination. These cells also show decreased activity of protein phosphatase 2A (PP2A), a phosphatase for γH2AX and other DNA damage and repair proteins. Thus, we propose that DSBs activate a Chk2-HuR-RhoB pathway that promotes PP2A-mediated dephosphorylation of γH2AX and DSB repair. Finally, we show that RhoB-deficient cells accumulate endogenous γH2AX and chromosomal abnormalities, suggesting that RhoB loss increases DSB-mediated genomic instability and tumor progression.


Subject(s)
DNA Breaks, Double-Stranded , Histones/metabolism , Protein Phosphatase 2/metabolism , rhoB GTP-Binding Protein/genetics , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Cell Line, Tumor , Checkpoint Kinase 2/metabolism , Chromosome Aberrations , DNA Repair/genetics , ELAV Proteins/metabolism , Genomic Instability/genetics , HCT116 Cells , Humans , Mice , Mice, Knockout , Phosphorylation , Protein Binding/genetics , Protein Phosphatase 2/genetics , RNA Interference , RNA, Small Interfering , RNA-Binding Proteins/metabolism , Topoisomerase I Inhibitors/pharmacology , rhoB GTP-Binding Protein/biosynthesis
19.
J Invest Dermatol ; 134(1): 203-212, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23792460

ABSTRACT

The role of UVB-induced apoptosis in the formation of squamous cell carcinoma (SCC) is recognized. We previously identified the small RhoB (Ras homolog gene family, member B) GTPase, an early response gene to cellular stress, as a critical protein controlling apoptosis of human keratinocytes after UVB exposure. Here we generated SKH1 (hairless immunocompetent mouse) mice invalidated for RhoB to evaluate its role in UVB-induced skin carcinogenesis in vivo. We show that rhob-/- mice have a lower risk of developing UVB-induced keratotic tumors and actinic keratosis that is associated with a higher sensitivity of UVB-exposed keratinocytes to apoptosis. We extend this observation to primary cultures of normal human keratinocytes in which RhoB was downregulated with small interfering RNA (siRNA) and further show that the hypersensitivity to apoptosis depends on B-cell lymphoma 2 (Bcl-2) downregulation. In rhob-/- mice, the UVB-induced tumors were preferentially undifferentiated and highly proliferative. Finally, we show in humans an almost constant loss of RhoB expression in undifferentiated SCCs. These undifferentiated and RhoB-deficient tumors have elevated phosphorylated histone H2AX (γH2AX) and 53BP1, two markers of DNA double-strand breaks. Together, our results indicate that UVB-induced RhoB expression participates in in vivo SCC initiation by increasing keratinocyte survival. Conversely, RhoB may limit tumor aggressiveness as loss of RhoB expression in tumor cells is associated with tumor progression.


Subject(s)
Apoptosis/physiology , Carcinoma, Squamous Cell/pathology , Keratinocytes/pathology , Skin Neoplasms/pathology , rhoB GTP-Binding Protein/physiology , Animals , Apoptosis/genetics , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Female , Humans , Keratinocytes/radiation effects , Male , Mice , Mice, 129 Strain , Mice, Hairless , Mice, Knockout , RNA, Small Interfering/genetics , Ultraviolet Rays/adverse effects , rhoB GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL