Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 343
Filter
Add more filters

Publication year range
1.
Drug Dev Res ; 79(6): 287-294, 2018 09.
Article in English | MEDLINE | ID: mdl-30295945

ABSTRACT

Preclinical Research & Development Pancreatic cancer is the third leading cause of death in the US with a poor 5-year survival rate of 8.5%. A novel anti-cancer drug, dimethylamino parthenolide (DMAPT), is the water-soluble analog of the natural sesquiterpene lactone, parthenolide. The putative modes of action of DMAPT are inhibition of the Nuclear chain factor kappa-light-chain enhancer of activated B cells (NFκB) pathway and depletion of glutathione levels; the latter causing cancer cells to be more susceptible to oxidative stress-induced cell death. Actinomycin-D (ActD) is a polypeptide antibiotic that binds to DNA, and inhibits RNA and protein synthesis by inhibiting RNA polymerase II. A phase 2 clinical trial indicated that ActD could be a potent drug against pancreatic cancer; however, it was not a favored drug due to toxicity issues. New drug entities and methods of drug delivery, used alone or in combination, are needed to treat pancreatic cancer more effectively. Thus, it was postulated that combining DMAPT and ActD would result in synergistic inhibition of Panc-1 pancreatic cancer cell growth because DMAPT's inhibition of NFκB would enhance induction of apoptosis by ActD, via phosphorylation of c-Jun, by minimizing NFκB inhibition of c-Jun phosphorylation. Combining these two drugs induced a higher level of cell death than each drug alone. A fixed drug ratio of DMAPT: ActD (1,200:1) was used. Data from metabolic (MTT) and colony formation assays were analyzed for synergism with CompuSyn software, which utilizes the Chou-Talalay equation. The analyses indicated synergism and moderate synergism at combination concentrations of DMAPT/ActD of 12/0.01 and 18/0.015 µM, respectively.


Subject(s)
Aminopyrine/administration & dosage , Antibiotics, Antineoplastic/administration & dosage , Dactinomycin/administration & dosage , Growth Inhibitors/administration & dosage , Pancreatic Neoplasms/drug therapy , Antibiotics, Antineoplastic/adverse effects , Apoptosis/drug effects , Dactinomycin/adverse effects , Drug Combinations , Drug Synergism , Growth Inhibitors/adverse effects , Humans , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured
2.
J Immunol ; 193(3): 1314-23, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24965778

ABSTRACT

In animal models of experimental cerebral malaria (ECM), neuropathology is associated with an overwhelming inflammatory response and sequestration of leukocytes and parasite-infected RBCs in the brain. In this study, we explored the effect of vitamin D (VD; cholecalciferol) treatment on host immunity and outcome of ECM in C57BL/6 mice during Plasmodium berghei ANKA (PbA) infection. We observed that oral administration of VD both before and after PbA infection completely protected mice from ECM. VD administration significantly dampened the inducible systemic inflammatory responses with reduced circulating cytokines IFN-γ and TNF and decreased expression of these cytokines by the spleen cells. Meanwhile, VD also resulted in decreased expression of the chemokines CXCL9 and CXCL10 and cytoadhesion molecules (ICAM-1, VCAM-1, and CD36) in the brain, leading to reduced accumulation of pathogenic T cells in the brain and ultimately substantial improvement of the blood-brain barriers of PbA-infected mice. In addition, VD inhibited the differentiation, activation, and maturation of splenic dendritic cells. Meanwhile, regulatory T cells and IL-10 expression levels were upregulated upon VD treatment. These data collectively demonstrated the suppressive function of VD on host inflammatory responses, which provides significant survival benefits in the murine ECM model.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Immunosuppressive Agents/administration & dosage , Malaria, Cerebral/immunology , Malaria, Cerebral/prevention & control , Plasmodium berghei/immunology , Vitamin D/administration & dosage , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Dendritic Cells/immunology , Dendritic Cells/pathology , Down-Regulation/immunology , Female , Growth Inhibitors/administration & dosage , Growth Inhibitors/therapeutic use , Host-Parasite Interactions/immunology , Immunosuppressive Agents/therapeutic use , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Primary Cell Culture , Random Allocation , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation/immunology , Vitamin D/therapeutic use
3.
Clin Exp Pharmacol Physiol ; 42(6): 711-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25903858

ABSTRACT

Metformin and vitamin D3 both exhibit a strong antiproliferative action in numerous cancer cell lines, including in human prostate cancer cells. Here we showed that the combination of the two drugs had a much stronger effect on DU145 human prostate cancer cell growth than either drug alone. In this research, cell proliferation was measured by methylthiazol tetrazolium (MTT) assay. Cell apoptosis was determined with Hoechst 33342 staining. Western blotting and cell cycle analyses were used to elucidate potential mechanisms of interaction between the drugs. It is shown that in cultured DU145 cells, vitamin D3 combined with metformin exhibits synergistic effects on cell proliferation and apoptosis. The underlying antitumor mechanisms may involve altered cycle distribution with a G1/S cell cycle arrest, activation of phospho-AMPK with subsequent inhibition of downstream mTOR signalling pathway, down-regulate c-Myc expression, and reducing the level of anti-apoptotic protein p-Bcl-2. In conclusion, metformin and vitamin D3 synergistically inhibit DU145 cell growth, indicating a promising clinical therapeutic strategy for the treatment of androgen-independent prostate cancer.


Subject(s)
AMP-Activated Protein Kinases/physiology , Cholecalciferol/administration & dosage , Growth Inhibitors/administration & dosage , Metformin/administration & dosage , Prostatic Neoplasms/metabolism , TOR Serine-Threonine Kinases/physiology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Humans , Male , Prostatic Neoplasms/drug therapy , Signal Transduction/drug effects , Signal Transduction/physiology
4.
Int J Mol Sci ; 16(8): 17637-54, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26263980

ABSTRACT

The increase in antibiotic resistant bacteria demands the development of new antibiotics against preferably new targets. The common approach is to test compounds for their ability to kill bacteria or to design molecules that inhibit essential protein activities in vitro. In the first case, the mode of action of the drug is unknown and in the second case, it is not known whether the compound will pass the impermeable barrier of the bacterial envelope. We developed an assay that detects the target of a compound, as well as its ability to pass the membrane(s) simultaneously. The Escherichia coli cytoskeletal protein MreB recruits protein complexes (elongasomes) that are essential for cell envelope growth. An in cell Förster Resonance Energy Transfer (FRET) assay was developed to detect the interaction between MreB molecules and between MreB and the elongasome proteins RodZ, RodA and PBP2. Inhibition of the polymerization of MreB by S-(3,4-dichlorobenzyl) isothiourea (A22) or of the activity of PBP2 by mecilinam resulted in loss or reduction of all measured interactions. This suggests that the interactions between the elongasome proteins are governed by a combination of weak affinities and substrate availability. This validated in cell FRET assay can be used to screen for cell envelope growth inhibitors.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/genetics , Fluorescence Resonance Energy Transfer , Gene Expression Regulation, Bacterial/drug effects , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/chemistry , Escherichia coli/drug effects , Escherichia coli/growth & development , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/chemistry , Growth Inhibitors/administration & dosage , Growth Inhibitors/chemistry , Membrane Proteins/biosynthesis , Membrane Proteins/chemistry , Penicillin-Binding Proteins/biosynthesis , Penicillin-Binding Proteins/chemistry , Substrate Specificity , Thiourea/administration & dosage , Thiourea/analogs & derivatives
5.
J Pharmacol Exp Ther ; 350(3): 646-56, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24993360

ABSTRACT

EPZ-5676 [(2R,3R,4S,5R)-2-(6-amino-9H-purin-9-yl)-5-((((1r,3S)-3-(2-(5-(tert-butyl)-1H-benzo[d]imidazol-2-yl)ethyl)cyclobutyl)(isopropyl)amino)methyl)tetrahydrofuran-3,4-diol], a small-molecule inhibitor of the protein methyltransferase DOT1L, is currently under clinical investigation for acute leukemias bearing MLL-rearrangements (MLL-r). In this study, we evaluated EPZ-5676 in combination with standard of care (SOC) agents for acute leukemias as well as other chromatin-modifying drugs in cellular assays with three human acute leukemia cell lines: MOLM-13 (MLL-AF9), MV4-11 (MLL-AF4), and SKM-1 (non-MLL-r). Studies were performed to evaluate the antiproliferative effects of EPZ-5676 combinations in a cotreatment model in which the second agent was added simultaneously with EPZ-5676 at the beginning of the assay, or in a pretreatment model in which cells were incubated for several days in the presence of EPZ-5676 prior to the addition of the second agent. EPZ-5676 was found to act synergistically with the acute myeloid leukemia (AML) SOC agents cytarabine or daunorubicin in MOLM-13 and MV4-11 MLL-r cell lines. EPZ-5676 is selective for MLL-r cell lines as demonstrated by its lack of effect either alone or in combination in the nonrearranged SKM-1 cell line. In MLL-r cells, the combination benefit was observed even when EPZ-5676 was washed out prior to the addition of the chemotherapeutic agents, suggesting that EPZ-5676 sets up a durable, altered chromatin state that enhances the chemotherapeutic effects. Our evaluation of EPZ-5676 in conjunction with other chromatin-modifying drugs also revealed a consistent combination benefit, including synergy with DNA hypomethylating agents. These results indicate that EPZ-5676 is highly efficacious as a single agent and synergistically acts with other chemotherapeutics, including AML SOC drugs and DNA hypomethylating agents in MLL-r cells.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzimidazoles/administration & dosage , Cell Proliferation/drug effects , Growth Inhibitors/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Methyltransferases/antagonists & inhibitors , Cell Line, Tumor , Drug Synergism , Histone-Lysine N-Methyltransferase , Humans , Leukemia, Myeloid, Acute/pathology , Methylation/drug effects , Methyltransferases/metabolism
6.
J Immunol ; 189(9): 4258-65, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23018462

ABSTRACT

Elevated levels of plasmacytoid dendritic cells (pDC) have been reported in breast cancer patients, but the significance remains undefined. Using three immunocompetent mouse models of breast cancer bone metastasis, we identified a key role for pDC in facilitating tumor growth through immunosuppression and aggressive osteolysis. Following infiltration of macrophages upon breast cancer dissemination, there was a steady increase in pDC within the bone, which resulted in a sustained Th2 response along with elevated levels of regulatory T cells and myeloid-derived suppressor cells. Subsequently, pDC and CD4(+) T cells, producing osteolytic cytokines, increased with tumor burden, causing severe bone damage. Microcomputed tomography and histology analyses of bone showed destruction of femur and tibia. The therapeutic significance of this finding was confirmed by depletion of pDC, which resulted in decreased tumor burden and bone loss by activating tumor-specific cytolytic CD8(+) T cells and decreasing suppressor cell populations. Thus, pDC depletion may offer a novel adjuvant strategy to therapeutically influence breast cancer bone metastasis.


Subject(s)
Bone Neoplasms/immunology , Bone Neoplasms/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Growth Inhibitors/administration & dosage , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Animals , Biomarkers, Tumor/metabolism , Bone Neoplasms/secondary , Cell Death/immunology , Cell Line, Tumor , Coculture Techniques , Dendritic Cells/cytology , Disease Progression , Female , Growth Inhibitors/therapeutic use , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Osteolysis/immunology , Survival Analysis
7.
Chemotherapy ; 60(5-6): 346-52, 2014.
Article in English | MEDLINE | ID: mdl-26314695

ABSTRACT

BACKGROUND: To design novel polychemotherapy regimens for gastric adenocarcinoma therapy with wider therapeutic windows using a novel duplex drug (D-D). METHODS: Two gastric adenocarcinoma (MKN-45 and 23132/87) and 2 non-malignant (NHDF and CCL-241) cell lines were treated with different drug regimens that included different doses of the standard triple-drug combination epirubicin (E) + cisplatin (C) + 5-fluorouracil (5-FU, F), i.e. ECF, and a new D-D that combined 2'-deoxy-5-fluorouridine (5FdU) and 3'ethinylcytidine. The cells were cultured for 14 days and the effect of the drug combinations was evaluated using CASY cell counting technology. RESULTS: Overall growth inhibition of the cell lines with ECF was not cancer cell line-specific. Replacing 5-FU in ECF with a D-D resulted in greater growth inhibition of cancer cells than of the non-malignant cell lines and the inversion of the chemosensitivity of MKN-45 and 23132/87 cells. The type and quantity of the combined drug regimen determined the cytotoxicity and chemosensitivity of the cell lines. CONCLUSION: The cytotoxicity and tumour-cell specificity of standard single drugs can be markedly changed and determined using multidrug combinations that include D-Ds.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Resistance, Neoplasm/drug effects , Growth Inhibitors/administration & dosage , Stomach Neoplasms/pathology , Adenocarcinoma/drug therapy , Cell Line , Cell Line, Tumor , Cisplatin/administration & dosage , Humans , Stomach Neoplasms/drug therapy , Uridine/administration & dosage , Uridine/analogs & derivatives
8.
ScientificWorldJournal ; 2014: 487969, 2014.
Article in English | MEDLINE | ID: mdl-25009830

ABSTRACT

Retroviral vectors are potent tools for gene delivery and various biomedical applications. To accomplish a gene transfer task successfully, retroviral vectors must effectively transduce diverse cell cultures at different phases of a cell cycle. However, very promising retroviral vectors based on the foamy viral (FV) backbone lack the capacity to efficiently transduce quiescent cells. It is hypothesized that this phenomenon might be explained as the inability of foamy viruses to form a pre-integration complex (PIC) with nuclear import activity in growth-arrested cells, which is the characteristic for lentiviruses (HIV-1). In this process, the HIV-1 central polypurine tract (cPPT) serves as a primer for plus-strand synthesis to produce a "flap" element and is believed to be crucial for the subsequent double-stranded cDNA formation of all retroviral RNA genomes. In this study, the effects of the lentiviral cPPT element on the FV transduction potential in dividing and growth-arrested (G1/S phase) adenocarcinomic human alveolar basal epithelial (A549) cells are investigated by experimental and theoretical methods. The results indicated that the HIV-1 cPPT element in a foamy viral vector background will lead to a significant reduction of the FV transduction and viral titre in growth-arrested cells due to the absence of PICs with nuclear import activity.


Subject(s)
Cell Division/physiology , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Growth Inhibitors/administration & dosage , HIV-1/genetics , Simian foamy virus/genetics , Cell Division/drug effects , Cell Line, Tumor , Forecasting , Humans , Lentivirus/genetics
9.
Int Wound J ; 11(4): 398-403, 2014 Aug.
Article in English | MEDLINE | ID: mdl-23116288

ABSTRACT

Oncostatin M (OSM) is a multifunctional cytokine found in a variety of pathologic conditions, which leads to excessive collagen deposition. Current studies demonstrate that OSM is also a mitogen for fibroblasts and has an anti-inflammatory action. It was therefore hypothesised that OSM may play an important role in healing of chronic wounds that usually involve decreased fibroblast function and persist in the inflammatory stage for a long time. In a previous in vitro study, the authors showed that OSM increased wound healing activities of diabetic dermal fibroblasts. However, wound healing in vivo is a complex process involving multiple factors. Thus, the purpose of this study was to evaluate the effect of OSM on diabetic wound healing in vivo. Five diabetic mice were used in this study. Four full-thickness round wounds were created on the back of each mouse (total 20 wounds). OSM was applied on the two left-side wounds (n = 10) and phosphate-buffered saline was applied on the two right-side wounds (n = 10). After 10 days, unhealed wound areas of the OSM and control groups were compared using the stereoimage optical topometer system. Also, epithelialisation, wound contraction and reduction in wound volume in each group were compared. The OSM-treated group showed superior results in all of the tested parameters. In particular, the unhealed wound area and the reduction in wound volume demonstrated statistically significant differences (P < 0·05). The results of this study indicate that topical application of OSM may have the potential to accelerate healing of diabetic wounds.


Subject(s)
Diabetes Mellitus, Experimental , Oncostatin M/administration & dosage , Wound Healing/drug effects , Wounds and Injuries/drug therapy , Administration, Topical , Animals , Bandages, Hydrocolloid , Growth Inhibitors/administration & dosage , Male , Mice , Mice, Transgenic , Treatment Outcome , Wounds and Injuries/pathology
10.
J Tradit Chin Med ; 34(3): 309-16, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24992758

ABSTRACT

OBJECTIVE: Few studies have investigated the effects produced by combinations of polysaccharides and chemotherapeutic drugs in cancer treatment. We hypothesized that a combination of polysaccharides (COP) from Lentinus edodes and Tricholoma matsutake would improve the efficacy of 5-fluorouracil (5-FU)-mediated inhibition of H22 cell growth. METHODS: Mice were injected H22 cells and then treated with either 5-FU, polysaccharides from Tricholoma matsutake (PTM), polysaccharides from Lentinus edodes (PL), PTM+PL, 5-FU+PTM, 5-FU+ PL, or 5-FU + COP. The tumor weight and volume, and splenic CD4 + and CD8 + T cell frequencies, were determined. Additionally, splenic natural killer (NK) cell and cytotoxic T lymphocyte (CTL) activities were assessed and the serum levels of tumor necrosis factor-alpha (TNF-alpha), Interleukin-2 (IL-2), and Interferon-gamma (IFN-gamma) were measured. RESULTS: Compared with mice from the control, 5-FU, PL, PTM, PTM + PL, 5-FU + PL, and 5-FU + PTM groups, mice treated with 5-FU + COP showed: (a) significantly reduced tumor weight and volume (P < 0.05); (b) significantly higher serum levels of TNF-alpha, IL-2, and IFN-gamma (P < 0.05); (c) significantly increased CD4+ and CD8+ T cell frequencies in the spleen (P < 0.05); and (d) significantly increased splenic NK cell and CTL activities (P < 0.05). The tumor weight and volume in mice treated with 5-FU+PL or 5-FU+PTM were significantly reduced compared with mice treated with 5-FU alone (P < 0.05). Serum levels of TNF-alpha, IL-2, and IFN-gamma, frequencies of CD4 + and CD8+ T cells in the spleen, and splenic NK and CTL activities were also significantly increased in mice treated with 5-FU+PL or 5-FU+PTM compared with mice treated with 5-FU alone (P < 0.05). CONCLUSION: Polysaccharides from Lentinus edodes and Tricholoma matsutake could enhance the efficacy of 5-FU-mediated H22 cell growth inhibition.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Fluorouracil/administration & dosage , Liver Neoplasms/drug therapy , Plant Extracts/administration & dosage , Polysaccharides/administration & dosage , Shiitake Mushrooms/chemistry , Tricholoma/chemistry , Animals , Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/physiopathology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Drug Synergism , Growth Inhibitors/administration & dosage , Humans , Immunologic Factors/administration & dosage , Immunomodulation/drug effects , Liver Neoplasms/immunology , Liver Neoplasms/physiopathology , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Vegetables/chemistry
11.
Prostate ; 73(11): 1135-46, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23657951

ABSTRACT

BACKGROUND: Resveratrol (Res) is recognized as a promising cancer chemoprevention dietary polyphenol with antioxidative, anti-inflammatory, and anticancer properties. However, the role of its analogues in prostate cancer (PCa) chemoprevention is unknown. METHODS: We synthesized several natural and synthetic analogues of Res and characterized their effects on PCa cells in vitro using a cell proliferation assay. A colony formation assay and in vitro validation of luciferase (Luc) activity was done for LNCaP-Luc cells that were consequently used for in vivo studies. The efficacy of Res, trimethoxy-resveratrol (3M-Res) and piceatannol (PIC) was studied in a subcutaneous (s.c.) model of PCa using oral gavage. Tumor progression was monitored by traditional caliper and bioluminescent imaging. The levels of cytokines in serum were examined by ELISA, and the levels of compounds in serum and tumor tissues were determined by gas chromatography-mass spectrometry. RESULTS: We examined the anti-proliferative activities of Res/analogues in three PCa cell lines. We further compared the chemopreventive effects of oral Res, 3M-Res, and PIC in LNCaP-Luc-xenografts. We found that 2 weeks pretreatment with the compounds diminished cell colonization, reduced tumor volume, and decreased tumor growth in the xenografts. Both 3M-Res and PIC demonstrated higher potency in inhibiting tumor progression compared to Res. Notably, 3M-Res was the most active in inhibiting cell proliferation and suppressing colony formation, and its accumulation in both serum and tumor tissues was the highest. CONCLUSIONS: Our findings offer strong pre-clinical evidence for the utilization of dietary stilbenes, particularly 3M-Res, as novel, potent, effective chemopreventive agents in PCa.


Subject(s)
Antineoplastic Agents/administration & dosage , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Stilbenes/administration & dosage , Administration, Oral , Animals , Cell Line, Tumor , Growth Inhibitors/administration & dosage , Male , Mice , Mice, Nude , Prostatic Neoplasms/prevention & control , Resveratrol , Xenograft Model Antitumor Assays/methods
12.
Biochem Biophys Res Commun ; 435(3): 385-90, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23665331

ABSTRACT

Epithelial ovarian cancer (EOC) is one of the leading causes of gynecological cancer death. Approximately 70% of the patients experience recurrence accompanied by the development of drug resistance 2-3 years after chemotherapy. Picropodophyllin (PPP) is a newly identified insulin-like growth factor-1 receptor (IGF-1R) inhibitor that has been shown to have anticancer properties. In this study, we investigated the effect of PPP on EOC growth in vitro and in vivo. The EOC cell line SKOV-3 was treated with increasing concentrations of PPP or cisplatin, and cell viability and apoptosis were evaluated. To study the effects of PPP on EOC growth, apoptosis, and toxicity in vivo, a BALB/c nude mouse xenograft model was established. Mice were treated with normal saline (controls), PPP, cisplatin, or PPP in combination with cisplatin. In addition, the expression of phosphorylated IGF-1R (pIGF-1R) was examined in vitro and in vivo. PPP induced a dose-dependent decrease in SKOV-3 cell viability in vitro and reduced tumor volume and weight in the in vivo xenograft model. Furthermore, PPP in combination with cisplatin was more effective in inhibiting the growth of SKOV-3 cells and xenografts than either drug alone. PPP-mediated growth inhibition was associated with apoptosis induction in vitro and in vivo. PPP was well tolerated in vivo and exerted its effects with minimal hepatotoxicity and renal toxicity. PPP downregulated the expression of pIGF-1R in vitro and in vivo, an effect that appeared to be associated with its growth inhibitory properties. Our results indicate that PPP may have therapeutic application in the treatment of EOC.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Growth Inhibitors/pharmacology , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Podophyllotoxin/analogs & derivatives , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/toxicity , Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Growth Inhibitors/administration & dosage , Growth Inhibitors/toxicity , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Podophyllotoxin/administration & dosage , Podophyllotoxin/pharmacology , Podophyllotoxin/toxicity , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
13.
Toxicol Appl Pharmacol ; 272(3): 780-6, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23948739

ABSTRACT

Mono-hydroxy methoxychlor (mono-OH MXC) is a metabolite of the pesticide, methoxychlor (MXC). Although MXC is known to decrease antral follicle numbers, and increase follicle death in rodents, not much is known about the ovarian effects of mono-OH MXC. Previous studies indicate that mono-OH MXC inhibits mouse antral follicle growth, increases follicle death, and inhibits steroidogenesis in vitro. Further, previous studies indicate that CYP11A1 expression and production of progesterone (P4) may be the early targets of mono-OH MXC in the steroidogenic pathway. Thus, this study tested whether supplementing pregnenolone, the precursor of progesterone and the substrate for HSD3B, would prevent decreased steroidogenesis, inhibited follicle growth, and increased follicle atresia in mono-OH MXC-treated follicles. Mouse antral follicles were exposed to vehicle (dimethylsulfoxide), mono-OH MXC (10 µg/mL), pregnenolone (1 µg/mL), or mono-OH MXC and pregnenolone together for 96 h. Levels of P4, androstenedione (A), testosterone (T), estrone (E1), and 17ß-estradiol (E2) in media were determined, and follicles were processed for histological evaluation of atresia. Pregnenolone treatment alone stimulated production of all steroid hormones except E2. Mono-OH MXC-treated follicles had decreased sex steroids, but when given pregnenolone, produced levels of P4, A, T, and E1 that were comparable to those in vehicle-treated follicles. Pregnenolone treatment did not prevent growth inhibition and increased atresia in mono-OH MXC-treated follicles. Collectively, these data support the idea that the most upstream effect of mono-OH MXC on steroidogenesis is by reducing the availability of pregnenolone, and that adding pregnenolone may not be sufficient to prevent inhibited follicle growth and survival.


Subject(s)
Follicular Atresia/drug effects , Growth Inhibitors/toxicity , Insecticides/toxicity , Methoxychlor/analogs & derivatives , Pregnenolone/administration & dosage , Animals , Cells, Cultured , Female , Follicular Atresia/metabolism , Gonadal Steroid Hormones/metabolism , Growth Inhibitors/administration & dosage , Growth Inhibitors/antagonists & inhibitors , Humans , Insecticides/administration & dosage , Methoxychlor/administration & dosage , Methoxychlor/toxicity , Mice , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Treatment Outcome
14.
Haematologica ; 98(9): 1450-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23539538

ABSTRACT

Patients with advanced systemic mastocytosis, including mast cell leukemia, have a poor prognosis. In these patients, neoplastic mast cells usually harbor the KIT mutant D816V that confers resistance against tyrosine kinase inhibitors. We examined the effects of the multi-kinase blocker ponatinib on neoplastic mast cells and investigated whether ponatinib acts synergistically with other antineoplastic drugs. Ponatinib was found to inhibit the kinase activity of KIT G560V and KIT D816V in the human mast cell leukemia cell line HMC-1. In addition, ponatinib was found to block Lyn- and STAT5 activity in neoplastic mast cells. Ponatinib induced growth inhibition and apoptosis in HMC-1.1 cells (KIT G560V(+)) and HMC-1.2 cells (KIT G560V(+)/KIT D816V(+)) as well as in primary neoplastic mast cells. The effects of ponatinib were dose-dependent, but higher IC50-values were obtained in HMC-1 cells harboring KIT D816V than in those lacking KIT D816V. In drug combination experiments, ponatinib was found to synergize with midostaurin in producing growth inhibition and apoptosis in HMC-1 cells and primary neoplastic mast cells. The ponatinib+midostaurin combination induced substantial inhibition of KIT-, Lyn-, and STAT5 activity, but did not suppress Btk. We then applied a Btk short interfering RNA and found that Btk knockdown sensitizes HMC-1 cells against ponatinib. Finally, we were able to show that ponatinib synergizes with the Btk-targeting drug dasatinib to produce growth inhibition in HMC-1 cells. In conclusion, ponatinib exerts major growth-inhibitory effects on neoplastic mast cells in advanced systemic mastocytosis and synergizes with midostaurin and dasatinib in inducing growth arrest in neoplastic mast cells.


Subject(s)
Growth Inhibitors/administration & dosage , Imidazoles/administration & dosage , Mastocytosis, Systemic/drug therapy , Mastocytosis, Systemic/genetics , Proto-Oncogene Proteins c-kit/genetics , Pyridazines/administration & dosage , Staurosporine/analogs & derivatives , Adult , Aged , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Drug Synergism , Female , Humans , Male , Mastocytosis, Systemic/diagnosis , Middle Aged , Staurosporine/administration & dosage
15.
J Immunol ; 186(1): 264-74, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21135171

ABSTRACT

The interaction between cancer vaccine adjuvants and myeloid-derived suppressor cells (MDSCs) is currently poorly understood. Very small size proteoliposomes (VSSP) are a nanoparticulated adjuvant under investigation in clinical trials in patients with renal carcinoma, breast cancer, prostate cancer, and cervical intraepithelial neoplasia grade III. We found that VSSP adjuvant induced a significant splenomegaly due to accumulation of CD11b(+)Gr-1(+) cells. However, VSSP-derived MDSCs showed a reduced capacity to suppress both allogeneic and Ag-specific CTL response compared with that of tumor-induced MDSCs. Moreover, splenic MDSCs isolated from tumor-bearing mice treated with VSSP were phenotypically more similar to those isolated from VSSP-treated tumor-free mice and much less suppressive than tumor-induced MDSCs, both in vitro and in vivo. Furthermore, different from dendritic cell vaccination, inoculation of VSSP-based vaccine in EG.7-OVA tumor-bearing mice was sufficient to avoid tumor-induced tolerance and stimulate an immune response against OVA Ag, similar to that observed in tumor-free mice. This effect correlated with an accelerated differentiation of MDSCs into mature APCs that was promoted by VSSP. VSSP used as a cancer vaccine adjuvant might thus improve antitumor efficacy not only by stimulating a potent immune response against tumor Ags but also by reducing tumor-induced immunosuppression.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antineoplastic Agents/administration & dosage , Cancer Vaccines/administration & dosage , Lymphoma/prevention & control , Myeloid Cells/immunology , Nanoparticles/administration & dosage , Proteolipids/administration & dosage , Sarcoma, Experimental/prevention & control , Animals , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Outer Membrane Proteins/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Female , G(M3) Ganglioside/administration & dosage , G(M3) Ganglioside/immunology , Growth Inhibitors/administration & dosage , Lymphoma/immunology , Lymphoma/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/pathology , Neisseria meningitidis/immunology , Proteolipids/immunology , Sarcoma, Experimental/immunology , Sarcoma, Experimental/pathology
16.
J Immunol ; 187(2): 1031-8, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21670311

ABSTRACT

One fourth of women with HER-2(+) metastatic breast carcinoma are treated with a combination regimen with trastuzumab, but the frequent resistance to this Ab requires definition of new means to improve its bioactivity. The mechanisms of action of trastuzumab involve several pathways including Ab-dependent cellular cytotoxicity. Because human γδ T lymphocytes mediate Ab-dependent cellular cytotoxicity and can be activated further by phosphoantigens, these cells are prone to improve the efficacy of Abs, as recently demonstrated for CD20(+) B cell lymphomas. Whether this concept applies as well with carcinomas remained to be demonstrated in vivo, however. In this study, we asked whether a combination of trastuzumab and phosphoantigen-stimulated γδ lymphocytes increases the efficacy of trastuzumab against HER-2(+) breast carcinoma cell lines in vivo. We report that repeated infusions of this combination had a better efficacy than that of trastuzumab alone against HER-2(+) mammary carcinoma xenografts in mice. In these models, reduction of tumor growth was observed together with trastuzumab opsonization of HER-2(+) cells and tumor infiltration by γδ lymphocytes. In addition in humans, the mammary carcinomas of 27 of 30 patients showed significant γδ T cell infiltrates. Altogether, these findings indicate that combination of trastuzumab and stimulated γδ cells represents a new strategy to improve the efficacy of Herceptin (trastuzumab) in HER-2(+) breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Lymphocyte Activation/immunology , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/therapy , Receptor, ErbB-2/biosynthesis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/transplantation , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Female , Growth Inhibitors/administration & dosage , Growth Inhibitors/therapeutic use , Humans , Immunotherapy, Adoptive/methods , Mammary Neoplasms, Animal/pathology , Mice , Mice, SCID , Phosphoproteins/administration & dosage , Phosphoproteins/therapeutic use , Receptors, Antigen, T-Cell, gamma-delta/administration & dosage , Receptors, Antigen, T-Cell, gamma-delta/therapeutic use , T-Lymphocyte Subsets/metabolism , Transplantation, Heterologous/immunology , Transplantation, Heterologous/methods , Transplantation, Heterologous/pathology , Trastuzumab
17.
J Immunol ; 186(4): 2254-61, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21242516

ABSTRACT

Thymic stromal lymphopoietin (TSLP) is an essential cytokine for the initiation and development of allergic inflammation. In this study, we have investigated the role of TSLP in the breakdown of immune tolerance and generation of inducible regulatory T cells (iTregs). Our results demonstrated that TSLP diverted airway tolerance against OVA to Th2 sensitization and inhibited the generation of OVA-specific iTregs. TSLP exerted a direct inhibitory effect on both human and mouse iTreg development in vitro. Low doses of TSLP were capable of inhibiting iTreg induction without significantly promoting Th2 development, indicating that these two functions of TSLP are separable. Moreover, the TSLP-mediated inhibition of iTreg generation was only partially dependent on IL-4 and Stat6, and was effective when TSLP was present for the first 24 h of T cell activation. These results define a novel role for TSLP in regulating the balance of airway tolerance and allergic inflammation.


Subject(s)
Asthma/immunology , Cytokines/physiology , Epitopes, T-Lymphocyte/immunology , Growth Inhibitors/physiology , Immune Tolerance , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Allergens/administration & dosage , Allergens/immunology , Animals , Asthma/drug therapy , Asthma/pathology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/pathology , Cytokines/administration & dosage , Cytokines/blood , Epitopes, T-Lymphocyte/biosynthesis , Gene Knock-In Techniques , Growth Inhibitors/administration & dosage , Growth Inhibitors/blood , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Stromal Cells/immunology , Stromal Cells/metabolism , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Th2 Cells/immunology , Th2 Cells/pathology , Thymus Gland/cytology , Thymic Stromal Lymphopoietin
18.
J Immunol ; 187(1): 212-21, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21622866

ABSTRACT

A fundamental problem in immunoregulation is how CD4(+) T cells react to immunogenic peptides derived from the V region of the BCR that are created by somatic mechanisms, presented in MHC II, and amplified to abundance by B cell clonal expansion during immunity. BCR neo Ags open a potentially dangerous avenue of T cell help in violation of the principle of linked Ag recognition. To analyze this issue, we developed a murine adoptive transfer model using paired donor B cells and CD4 T cells specific for a BCR-derived peptide. BCR peptide-specific T cells aborted ongoing germinal center reactions and impeded the secondary immune response. Instead, they induced the B cells to differentiate into short-lived extrafollicular plasmablasts that secreted modest quantities of Ig. These results uncover an immunoregulatory process that restricts the memory pathway to B cells that communicate with CD4 T cells via exogenous foreign Ag.


Subject(s)
B-Lymphocyte Subsets/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes, B-Lymphocyte/immunology , Germinal Center/immunology , Immunoglobulin Variable Region/immunology , Immunologic Memory , Peptides/immunology , Receptors, Antigen, B-Cell/immunology , Adoptive Transfer , Amino Acid Sequence , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , B-Lymphocyte Subsets/pathology , B-Lymphocyte Subsets/transplantation , CD4-Positive T-Lymphocytes/classification , CD4-Positive T-Lymphocytes/pathology , Cell Communication/genetics , Cell Communication/immunology , Epitopes, B-Lymphocyte/administration & dosage , Female , Germinal Center/pathology , Growth Inhibitors/administration & dosage , Growth Inhibitors/genetics , Growth Inhibitors/immunology , Immunoglobulin Variable Region/administration & dosage , Immunoglobulin kappa-Chains/genetics , Immunologic Memory/genetics , Mice , Mice, Inbred A , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Peptides/administration & dosage , Peptides/genetics , Plasma Cells/immunology , Plasma Cells/pathology , Receptors, Antigen, B-Cell/administration & dosage
19.
J Immunol ; 187(9): 4611-20, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21957150

ABSTRACT

Atopic dermatitis (AD) is a common pruritic inflammatory disease triggered by a defective skin barrier and immunodysregulation. AD has been considered a typical example of a Th2 response associated with allergic disease. In the early phases of the disease, symptoms include IgE hyperproduction, eosinophil accumulation, and mast cell activation; in the chronic phase, a Th1-dominant immune response is also observed at the sites of AD skin lesions. The role of IL-17-producing Th (Th17) cells in AD has not been established. In the current study, we found that pyridone 6 (P6), a pan-JAK inhibitor, delayed the onset and reduced the magnitude of skin disease in an AD-like skin-disease model of NC/Nga mice. P6 reduced IFN-γ and IL-13, whereas it enhanced IL-17 and IL-22 expression. In vitro, P6 also inhibited both Th1 and Th2 development, whereas it promoted Th17 differentiation from naive T cells when present within a certain range of concentrations. This was probably because P6 strongly inhibited STAT1, STAT5, and STAT6 phosphorylation, whereas STAT3 phosphorylation was less efficiently suppressed by P6 at the same concentration. Furthermore, IL-22 protects keratinocytes from apoptosis induced by IFN-γ, and administration of IL-17 and IL-22 partially ameliorated skin diseases in NC/Nga mice. These results suggested that the JAK inhibitor P6 is therapeutic for AD by modulating the balance of Th2 and Th17.


Subject(s)
Benzimidazoles/therapeutic use , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/pathology , Down-Regulation/immunology , Janus Kinases/antagonists & inhibitors , Pyridones/therapeutic use , Th17 Cells/pathology , Th2 Cells/pathology , Up-Regulation/immunology , Animals , Benzimidazoles/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Dermatitis, Atopic/immunology , Dermatophagoides farinae/immunology , Disease Models, Animal , Down-Regulation/drug effects , Growth Inhibitors/administration & dosage , Growth Inhibitors/pharmacology , Inflammation Mediators/administration & dosage , Inflammation Mediators/pharmacology , Interleukin-17/biosynthesis , Interleukins/biosynthesis , Mice , Pyridones/pharmacology , Th17 Cells/drug effects , Th17 Cells/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Up-Regulation/drug effects , Interleukin-22
20.
Acta Pharmacol Sin ; 34(7): 930-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23708557

ABSTRACT

AIM: Noscapine (NOS) is a non-narcotic opium alkaloid with anti-tumor activity. The aim of this study was to investigate the effects of the combination of NOS with conventional chemotherapeutics temozolamide (TMZ), bis-chloroethylnitrosourea (BCNU), or cisplatin (CIS)on human glioblastoma cells. METHODS: U87MG human glioblastoma cells were examined. Cell proliferation was quantified using MTT assay. Western blotting and flow cytometry were used to examine apoptosis and the expression of active caspase-3 and cleaved PARP. Mouse tumor xenograft model bearing U87MG cells was treated with TMZ (2 mg·kg(-1)·d(-1), ip) or CIS (2 mg/kg, ip 3 times a week) alone or in combination with NOS (200 mg·kg(-1)·d(-1), ig) for 3 weeks. Immunohistochemistry was used to investigate the expression of active caspase-3 and Ki67 following treatment in vivo. The safety of the combined treatments was evaluated based on the body weight and histological studies of the animal's organs. RESULTS: NOS (10 or 20 mol/L) markedly increased the anti-proliferation effects of TMZ, BCNU, and CIS on U87MG cells in vitro. The calculated combination index (CI) values of NOS-CIS, NOS-TMZ, and NOS-BCNU (20 µmol/L) were 0.45, 0.51, and 0.57, respectively, demonstrating synergistic inhibition of the drug combinations. In tumor xenograft models, combined treatment with NOS robustly augmented the anti-cancer actions of TMZ and CIS, and showed no detectable toxicity. The combined treatments significantly enhanced the apoptosis, the activated caspase-3 and PARP levels in U87MG cells in vitro, and reduced Ki67 staining and increased the activated caspase-3 level in the shrinking xenografts in vivo. CONCLUSION: NOS synergistically potentiated the efficacy of FDA-approved anti-cancer drugs against human glioblastoma cells, thereby allowing them to be used at lower doses and hence minimizing their toxic side effects.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell Proliferation/drug effects , Glioblastoma/pathology , Growth Inhibitors/administration & dosage , Noscapine/administration & dosage , Animals , Cell Line, Tumor , Drug Synergism , Female , Glioblastoma/drug therapy , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Xenograft Model Antitumor Assays/methods
SELECTION OF CITATIONS
SEARCH DETAIL