Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Control Release ; 286: 1-9, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30016731

ABSTRACT

CX-5461 is currently in Phase I/II clinical trials for advanced hematologic malignancies and triple negative or BRCA-deficient breast cancer. The compound is currently administered to patients intravenously (i.v.) at low pH (3.5) due to solubility challenges. Reliance of low pH to enhance solubility of CX-5461 can adversely impact pharmacokinetics, biodistribution and therapeutic potential. We have addressed this solubility issue through a formulation method that relies on the interactions between CX-5461 and copper. Copper binds CX-5461 through the nitrogens of the pyrazine ring. Here, we describe synthesizing this copper-complexed CX-5461 (Cu(CX-5461)) within liposomes. CX-5461 was added to copper-containing liposomes and incubated at 60 °C for 30 min. The pharmacokinetics of CX-5461 was assessed in mice following a single i.v. injection at 30 mg/kg. Efficacy studies were completed in multiple subcutaneous mouse xenografts as well as in a bone marrow engraftment model of acute myeloid leukemia (AML). The novel Cu(CX-5461) formulation was stable at pH 7.4 and exhibited increased plasma circulation longevity, increasing the total exposure to CX5461 by an order of magnitude. Cu(CX-5461) was more active than CX-5461 in AML models in vivo. In HCT116-B46 and Capan-1 solid tumour models that are BRCA-deficient, the Cu(CX-5461) formulation engendered activity that was comparable to that of the low pH CX-5461 formulation. We have generated the first Cu(CX-5461) formulation suitable for i.v. administration that is more efficacious than the existing low-pH formulation in pre-clinical models of AML. The Cu(CX-5461) formulation may serve as an alternative formulation for CX-5461 in BRCA-deficient cancers.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzothiazoles/administration & dosage , Copper/administration & dosage , Naphthyridines/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Benzothiazoles/chemistry , Benzothiazoles/pharmacokinetics , Benzothiazoles/therapeutic use , Cell Line, Tumor , Coordination Complexes/administration & dosage , Coordination Complexes/chemistry , Coordination Complexes/pharmacokinetics , Coordination Complexes/therapeutic use , Copper/chemistry , Copper/pharmacokinetics , Copper/therapeutic use , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Liposomes/chemistry , Mice , Naphthyridines/chemistry , Naphthyridines/pharmacokinetics , Naphthyridines/therapeutic use , RNA, Ribosomal/antagonists & inhibitors , RNA, Ribosomal/metabolism , Tissue Distribution
2.
Drug Deliv Transl Res ; 8(1): 239-251, 2018 02.
Article in English | MEDLINE | ID: mdl-29247315

ABSTRACT

Clioquinol (CQ) is an FDA-approved topical antifungal agent known to kill cancer cells. This facilitated the initiation of clinical trials in patients with refractory hematologic malignancies. These repurposing efforts were not successful; this was likely due to low intracellular levels of the drug owing to poor absorption and rapid metabolism upon oral administration. CQ forms a sparingly soluble copper complex (Cu(CQ)2) that exhibits enhanced anticancer activity in some cell lines. We have utilized a novel method to synthesize Cu(CQ)2 inside liposomes, an approach that maintains the complex suspended in solution and in a format suitable for intravenous administration. The complex was prepared inside 100-nm liposomes composed of 1,2-distearoyl-sn-glycero-3-phosphocholine/cholesterol (55:45). The therapeutic activity of the resultant formulation was evaluated in two subcutaneous tumor models (glioblastoma and ovarian cancers) but was not active. We also assessed the ability of the Cu(CQ)2 formulation to increase copper delivery to cancer cells in vitro and its potential to be used in combination with disulfiram (DSF). The results suggested that addition of Cu(CQ)2 enhanced cellular copper levels and the activity of DSF in vitro; however, this combination did not result in a statistically significant reduction in tumor growth in vivo. These studies demonstrate that a Cu(CQ)2 formulation suitable for intravenous use can be prepared, but this formulation used alone or in combination with DSF was not efficacious. The methods described are suitable for development formulations of other analogues of 8-hydroxyquinoline which could prove to be more potent.


Subject(s)
Antifungal Agents/administration & dosage , Antineoplastic Agents/administration & dosage , Clioquinol/administration & dosage , Copper/administration & dosage , Administration, Intravenous , Animals , Antifungal Agents/chemistry , Antifungal Agents/pharmacokinetics , Antifungal Agents/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Cholesterol/chemistry , Clioquinol/chemistry , Clioquinol/pharmacokinetics , Clioquinol/therapeutic use , Copper/chemistry , Copper/pharmacokinetics , Copper/therapeutic use , Disulfiram/administration & dosage , Disulfiram/chemistry , Disulfiram/therapeutic use , Drug Therapy, Combination , Humans , Liposomes , Mice , Neoplasms/drug therapy , Neoplasms/pathology , Phosphatidylcholines/chemistry , Tumor Burden/drug effects
3.
Invest New Drugs ; 35(6): 682-690, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28733701

ABSTRACT

Insensitivity to platinum, either through inherent or acquired resistance, is a major clinical problem in the treatment of many solid tumors. Here, we explored the therapeutic potential of diethyldithiocarbamate (DDC), pyrithione (Pyr), plumbagin (Plum), 8-hydroxyquinoline (8-HQ), clioquinol (CQ) copper complexes in a panel of cancer cell lines that differ in their sensitivity to platins (cisplatin/carboplatin) using a high-content imaging system. Our data suggest that the copper complexes were effective against both platinum sensitive (IC50 ~ 1 µM platinum) and insensitive (IC50 > 5 µM platinum) cell lines. Furthermore, copper complexes of DDC, Pyr and 8-HQ had greater therapeutic activity compared to the copper-free ligands in all cell lines; whereas the copper-dependent activities of Plum and CQ were cell-line specific. Four of the copper complexes (Cu(DDC)2, Cu(Pyr)2, Cu(Plum)2 and Cu(8-HQ)2) showed IC50 values less than that of cisplatin in all tested cell lines. The complex copper DDC (Cu(DDC)2) was selected for in vivo evaluation due to its low nano-molar range activity in vitro and the availability of an injectable liposomal formulation. Liposomal (Cu(DDC)2) was tested in a fast-growing platinum-resistant A2780-CP ovarian xenograft model and was found to achieve a statistically significant reduction (50%; p < 0.05) in tumour size. This work supports the potential use of copper-based therapeutics to treat cancers that are insensitive to platinum drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Coordination Complexes/metabolism , Copper/metabolism , Drug Resistance, Neoplasm/drug effects , Organoplatinum Compounds/pharmacology , Ovarian Neoplasms/pathology , Animals , Cell Proliferation/drug effects , Coordination Complexes/chemistry , Copper/chemistry , Female , Humans , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Int J Nanomedicine ; 12: 4129-4146, 2017.
Article in English | MEDLINE | ID: mdl-28615941

ABSTRACT

Copper diethyldithiocarbamate (Cu(DDC)2) is the active anticancer agent generated when disulfiram (DSF) is provided in the presence of copper. To date, research directed toward repurposing DSF as an anticancer drug has focused on administration of DSF and copper in combination, efforts that have proven unsuccessful in clinical trials. This is likely due to the inability to form Cu(DDC)2 at relevant concentrations in regions of tumor growth. Little effort has been directed toward the development of Cu(DDC)2 because of the inherent aqueous insolubility of the complex. Here, we describe an injectable Cu(DDC)2 formulation prepared through a method that involves synthesis of Cu(DDC)2 inside the aqueous core of liposomes. Convection-enhanced delivery of a Cu(DDC)2 formulation prepared using 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC)/cholesterol liposomes into a rat model of F98 glioma engendered a 25% increase in median survival time relative to vehicle-treated animals. In a murine subcutaneous MV-4-11 model, treatment resulted in a 45% reduction in tumor burden when compared to controls. Pharmacokinetic studies indicated that the Cu(DDC)2 was rapidly eliminated after intravenous administration while the liposomes remained in circulation. To test whether liposomal lipid composition could increase Cu(DDC)2 circulation lifetime, a number of different formulations were evaluated. Studies demonstrated that liposomes composed of DSPC and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-n-(carboxy[polyethylene glycol]-2000) (95:5) enhanced Cu(DDC)2 concentrations in the circulation as reflected by a 4.2-fold increase in plasma AUC(0-∞) relative to the DSPC/cholesterol formulation. The anticancer activity of this Cu(DDC)2 formulation was subsequently evaluated in the MV-4-11 model. At its maximum tolerated dose, this formulation exhibited comparable activity to the DSPC/cholesterol formulation. This is the first report demonstrating the therapeutic effects of an injectable Cu(DDC)2 formulation in vivo.


Subject(s)
Antineoplastic Agents/administration & dosage , Copper/administration & dosage , Ditiocarb/administration & dosage , Liposomes/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cholesterol/chemistry , Copper/chemistry , Copper/pharmacokinetics , Ditiocarb/chemistry , Ditiocarb/pharmacokinetics , Drug Compounding , Injections, Intravenous , Male , Maximum Tolerated Dose , Mice , Neoplasms/drug therapy , Phosphatidylcholines/chemistry , Polyethylene Glycols/chemistry , Rats, Inbred F344 , Xenograft Model Antitumor Assays
5.
Drug Deliv Transl Res ; 7(4): 544-557, 2017 08.
Article in English | MEDLINE | ID: mdl-28432657

ABSTRACT

Topotecan is a drug that is under investigation for the treatment of neuroblastoma and has been encapsulated into liposomes to improve its therapeutic efficacy. However, liposomal formulations still need to be optimized for drug retention and new techniques to measure drug release are required to better understand this process. Here, a novel in vitro method based on fluorescence de-quenching and an automated microscopy imaging platform were developed for monitoring, in real time, the release of topotecan from a liposomal formulation. Drug release from liposomes was monitored for up to 15 h under different conditions including topotecan concentrations, fetal bovine serum amounts (0-20%), and temperatures (25 and 37 °C). A cell-based assay was used to assess liposome association with cells in culture and to quantify amounts of topotecan internalized into cells after release from liposomes. Our results show that the liposomal topotecan concentration had an influence on drug release kinetics: there was a reduction in release rate as a function of increasing concentration. Our data also show that topotecan release from the liposomal formulation was dependent on serum concentration where faster release was observed at higher serum concentrations, and on temperature where faster release was found at 37 °C. This real-time liposomal drug release assay allows for better understanding of the factors important in governing release of topotecan. The assay will be essential towards designing liposomal formulations of topotecan (and potentially of other camptothecin derivatives such as irinotecan) with optimized retention times and better therapeutic efficacy for testing in the clinic.


Subject(s)
Topoisomerase I Inhibitors/administration & dosage , Topotecan/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Drug Liberation , Humans , Kinetics , Liposomes , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/pharmacology , Topotecan/chemistry , Topotecan/pharmacology , Transcytosis
6.
Oncotarget ; 7(41): 66970-66988, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27556700

ABSTRACT

Autophagy, a lysosome-mediated degradation and recycling process, functions in advanced malignancies to promote cancer cell survival and contribute to cancer progression and drug resistance. While various autophagy inhibition strategies are under investigation for cancer treatment, corresponding patient selection criteria for these autophagy inhibitors need to be developed. Due to its central roles in the autophagy process, the cysteine protease ATG4B is one of the autophagy proteins being pursued as a potential therapeutic target. In this study, we investigated the expression of ATG4B in breast cancer, a heterogeneous disease comprised of several molecular subtypes. We examined a panel of breast cancer cell lines, xenograft tumors, and breast cancer patient specimens for the protein expression of ATG4B, and found a positive association between HER2 and ATG4B protein expression. We showed that HER2-positive cells, but not HER2-negative breast cancer cells, require ATG4B to survive under stress. In HER2-positive cells, cytoprotective autophagy was dependent on ATG4B under both starvation and HER2 inhibition conditions. Combined knockdown of ATG4B and HER2 by siRNA resulted in a significant decrease in cell viability, and the combination of ATG4B knockdown with trastuzumab resulted in a greater reduction in cell viability compared to trastuzumab treatment alone, in both trastuzumab-sensitive and -resistant HER2 overexpressing breast cancer cells. Together these results demonstrate a novel association of ATG4B positive expression with HER2 positive breast cancers and indicate that this subtype is suitable for emerging ATG4B inhibition strategies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagy-Related Proteins/biosynthesis , Autophagy/physiology , Breast Neoplasms/metabolism , Cysteine Endopeptidases/biosynthesis , Drug Resistance, Neoplasm/drug effects , Adult , Aged , Animals , Autophagy/drug effects , Drug Resistance, Neoplasm/physiology , Female , Heterografts , Humans , Mice , Middle Aged , Receptor, ErbB-2/biosynthesis , Trastuzumab/pharmacology , Xenograft Model Antitumor Assays
7.
Sci Rep ; 6: 18537, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26727049

ABSTRACT

Overexpresssion of HER-2 in the MDA-MB-435/LCC6 (LCC6(HER-2)) tumour model is associated with significantly increased hypoxia and reduced necrosis compared to isogenic control tumours (LCC6(Vector)); this difference was not related to tumour size or changes in vascular architecture. To further evaluate factors responsible for HER-2-associated changes in the tumour microenvironment, small animal magnetic resonance imaging (MRI) and positron emission tomography (PET) were used to measure tumour tissue perfusion and metabolism, respectively. The imaging data was further corroborated by analysis of molecular markers pertaining to energy homeostasis, and measurements of hypoxia and glucose consumption. The results showed a strong trend towards higher perfusion rates (~58% greater, p = 0.14), and significantly higher glucose uptake in LCC6(HER-2) (~2-fold greater; p = 0.025), relative to control tumours. The expression of proteins related to energy stress (P-AMPK, P-ACC) and glucose transporters (GLUT1) were lower in LCC6(HER-2) tumours (~2- and ~4-fold, respectively). The in vitro analysis showed that LCC6(HER-2) cells become more hypoxic in 1% oxygen and utilise significantly more glucose in normoxia compared to LCC6(Vector)cells (p < 0.005). Amalgamation of all the data points suggests a novel metabolic adaptation driven by HER-2 overexpression where higher oxygen and glucose metabolic rates produce rich energy supply but also a more hypoxic tumour mass.


Subject(s)
Energy Metabolism , Gene Expression , Neoplasms/genetics , Neoplasms/metabolism , Receptor, ErbB-2/genetics , Stress, Physiological , Animals , Cell Line, Tumor , Cell Survival/genetics , Disease Models, Animal , Female , Glucose/metabolism , Humans , Hypoxia , Magnetic Resonance Imaging , Neoplasms/diagnosis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Oxygen Consumption , Positron-Emission Tomography
8.
Oncotarget ; 6(19): 17161-77, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-26220590

ABSTRACT

Standard treatment for advanced non-small cell lung cancer (NSCLC) with no known driver mutation is platinum-based chemotherapy, which has a response rate of only 30-33%. Through an siRNA screen, 3'-phosphoadenosine 5'-phosphosulfate (PAPS) synthase 1 (PAPSS1), an enzyme that synthesizes the biologically active form of sulfate PAPS, was identified as a novel platinum-sensitizing target in NSCLC cells. PAPSS1 knockdown in combination with low-dose (IC10) cisplatin reduces clonogenicity of NSCLC cells by 98.7% (p < 0.001), increases DNA damage, and induces G1/S phase cell cycle arrest and apoptosis. PAPSS1 silencing also sensitized NSCLC cells to other DNA crosslinking agents, radiation, and topoisomerase I inhibitors, but not topoisomerase II inhibitors. Chemo-sensitization was not observed in normal epithelial cells. Knocking out the PAPSS1 homolog did not sensitize yeast to cisplatin, suggesting that sulfate bioavailability for amino acid synthesis is not the cause of sensitization to DNA damaging agents. Rather, sensitization may be due to sulfation reactions involved in blocking the action of DNA damaging agents, facilitating DNA repair, promoting cancer cell survival under therapeutic stress or reducing the bioavailability of DNA damaging agents. Our study demonstrates for the first time that PAPSS1 could be targeted to improve the activity of multiple anticancer agents used to treat NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/genetics , Multienzyme Complexes/genetics , Sulfate Adenylyltransferase/genetics , Antineoplastic Agents/pharmacology , Blotting, Western , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Line, Tumor , Cisplatin/pharmacology , DNA Damage/drug effects , Flow Cytometry , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Lung Neoplasms/enzymology , Multienzyme Complexes/metabolism , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Sulfate Adenylyltransferase/metabolism , Transfection
9.
Clin Cancer Res ; 20(12): 3159-73, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24721646

ABSTRACT

PURPOSE: Triple-negative breast cancers (TNBC) are defined by a lack of expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (ERBB2/HER2). Although initially responsive to chemotherapy, most recurrent TNBCs develop resistance, resulting in disease progression. Autophagy is a lysosome-mediated degradation and recycling process that can function as an adaptive survival response during chemotherapy and contribute to chemoresistance. Our goal was to determine whether autophagy inhibition improves treatment efficacy in TNBC cells in tumors either sensitive or refractory to anthracyclines. EXPERIMENTAL DESIGN: We used in vitro and in vivo models of TNBC using cell lines sensitive to epirubicin and other anthracyclines, as well as derivative lines, resistant to the same drugs. We assessed basal autophagy levels and the effects of chemotherapy on autophagy in parental and resistant cells. Applying various approaches to inhibit autophagy alone and in combination with chemotherapy, we assessed the effects on cell viability in vitro and tumor growth rates in vivo. RESULTS: We demonstrated that epirubicin induced autophagic flux in TNBC cells. Epirubicin-resistant lines exhibited at least 1.5-fold increased basal autophagy levels and, when treated with autophagy inhibitors, showed a significant loss in viability, indicating dependence of resistant cells on autophagy for survival. Combination of epirubicin with the autophagy inhibitor hydroxychloroquine resulted in a significant reduction in tumor growth compared with monotherapy with epirubicin. CONCLUSION: Autophagy inhibition enhances therapeutic response in both anthracycline-sensitive and -resistant TNBC and may be an effective new treatment strategy for this disease.


Subject(s)
Anthracyclines/pharmacology , Antibiotics, Antineoplastic/pharmacology , Autophagy/drug effects , Drug Resistance, Neoplasm/drug effects , Epirubicin/pharmacology , Triple Negative Breast Neoplasms/prevention & control , Animals , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Female , Humans , Mice , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
PLoS One ; 8(10): e76503, 2013.
Article in English | MEDLINE | ID: mdl-24146879

ABSTRACT

Gefitinib (Iressa(®), ZD1839) is a small molecule inhibitor of the epidermal growth factor receptor (EGFR) tyrosine kinase. We report on an early cellular response to gefitinib that involves induction of functional autophagic flux in phenotypically diverse breast cancer cells that were sensitive (BT474 and SKBR3) or insensitive (MCF7-GFPLC3 and JIMT-1) to gefitinib. Our data show that elevation of autophagy in gefitinib-treated breast cancer cells correlated with downregulation of AKT and ERK1/2 signaling early in the course of treatment. Inhibition of autophagosome formation by BECLIN-1 or ATG7 siRNA in combination with gefitinib reduced the abundance of autophagic organelles and sensitized SKBR3 but not MCF7-GFPLC3 cells to cell death. However, inhibition of the late stage of gefitinib-induced autophagy with hydroxychloroquine (HCQ) or bafilomycin A1 significantly increased (p<0.05) cell death in gefitinib-sensitive SKBR3 and BT474 cells, as well as in gefitinib-insensitive JIMT-1 and MCF7-GFPLC3 cells, relative to the effects observed with the respective single agents. Treatment with the combination of gefitinib and HCQ was more effective (p<0.05) in delaying tumor growth than either monotherapy (p>0.05), when compared to vehicle-treated controls. Our results also show that elevated autophagosome content following short-term treatment with gefitinib is a reversible response that ceases upon removal of the drug. In aggregate, these data demonstrate that elevated autophagic flux is an early response to gefitinib and that targeting EGFR and autophagy should be considered when developing new therapeutic strategies for EGFR expressing breast cancers.


Subject(s)
Autophagy/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Quinazolines/pharmacology , Quinazolines/therapeutic use , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 7 , Beclin-1 , Breast Neoplasms/ultrastructure , Cadaverine/analogs & derivatives , Cadaverine/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cytoplasmic Vesicles/drug effects , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/ultrastructure , ErbB Receptors/metabolism , Female , Gefitinib , Gene Knockdown Techniques , Gene Silencing/drug effects , Humans , Hydroxychloroquine/pharmacology , Hydroxychloroquine/therapeutic use , Membrane Proteins/metabolism , Mice , Phagosomes/drug effects , Phagosomes/metabolism , Phagosomes/ultrastructure , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Staining and Labeling , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Treatment Outcome , Ubiquitin-Activating Enzymes/metabolism , Xenograft Model Antitumor Assays
11.
BMC Cancer ; 11: 420, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21961653

ABSTRACT

BACKGROUND: HER2-positive breast cancers exhibit high rates of innate and acquired resistance to trastuzumab (TZ), a HER2-directed antibody used as a first line treatment for this disease. TZ resistance may in part be mediated by frequent co-expression of EGFR and by sustained activation of the mammalian target of rapamycin (mTOR) pathway. Here, we assessed feasibility of combining the EGFR inhibitor gefitinib and the mTOR inhibitor everolimus (RAD001) for treating HER2 overexpressing breast cancers with different sensitivity to TZ. METHODS: The gefitinib and RAD001 combination was broadly evaluated in TZ sensitive (SKBR3 and MCF7-HER2) and TZ resistant (JIMT-1) breast cancer models. The effects on cell growth were measured in cell based assays using the fixed molar ratio design and the median effect principle. In vivo studies were performed in Rag2M mice bearing established tumors. Analysis of cell cycle, changes in targeted signaling pathways and tumor characteristics were conducted to assess gefitinib and RAD001 interactions. RESULTS: The gefitinib and RAD001 combination inhibited cell growth in vitro in a synergistic fashion as defined by the Chou and Talalay median effect principle and increased tumor xenograft growth delay. The improvement in therapeutic efficacy by the combination was associated in vitro with cell line dependent increases in cytotoxicity and cytostasis while treatment in vivo promoted cytostasis. The most striking and consistent therapeutic effect of the combination was increased inhibition of the mTOR pathway (in vitro and in vivo) and EGFR signaling in vivo relative to the single drugs. CONCLUSIONS: The gefitinib and RAD001 combination provides effective control over growth of HER2 overexpressing cells and tumors irrespective of the TZ sensitivity status.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Quinazolines/therapeutic use , Receptor, ErbB-2/genetics , Sirolimus/analogs & derivatives , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Everolimus , Female , Gefitinib , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Phosphorylation/drug effects , Quinazolines/administration & dosage , Receptor, ErbB-2/antagonists & inhibitors , Signal Transduction/drug effects , Sirolimus/administration & dosage , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/metabolism , Trastuzumab , Xenograft Model Antitumor Assays
12.
Breast Cancer Res ; 11(3): R25, 2009.
Article in English | MEDLINE | ID: mdl-19409087

ABSTRACT

INTRODUCTION: Substantial preclinical evidence has indicated that inhibition of integrin linked-kinase (ILK) correlates with cytotoxic/cytostatic cellular effects, delayed tumor growth in animal models of cancer, and inhibition of angiogenesis. Widely anticipated to represent a very promising therapeutic target in several cancer indications, it is increasingly evident that optimal therapeutic benefits obtained using ILK targeting strategies will only be achieved in combination settings. The purpose of this study was to investigate the therapeutic potential of the ILK small molecule inhibitor, QLT0267 (267), alone or in combination with chemotherapies commonly used to treat breast cancer patients. METHODS: A single end-point metabolic assay was used as an initial screen for 267 interactions with selected chemotherapeutic agents. These in vitro assays were completed with seven breast cancer cell lines including several which over-expressed human epidermal growth factor receptor 2 (Her2). One agent, docetaxel (Dt), consistently produced synergistic interactions when combined with 267. Dt/267 interactions were further characterized by measuring therapeutic endpoints linked to phosphorylated protein kinase B (P-AKT) suppression, inhibition of vascular endothelial growth factor (VEGF) secretion and changes in cytoarchitecture. In vivo efficacy studies were completed in mice bearing orthotopic xenografts where tumor growth was assessed by bioluminescence and calliper methods. RESULTS: The combination of 267 and Dt resulted in increased cytotoxic activity, as determined using an assay of metabolic activity. Combinations of cisplatin, doxorubicin, vinorelbine, paclitaxel, and trastuzumab produced antagonistic interactions. Further endpoint analysis in cell lines with low Her2 levels revealed that the 267/Dt combinations resulted in: a three-fold decrease in concentration (dose) of 267 required to achieve 50% inhibition of P-AKT; and a dramatic disruption of normal filamentous-actin cellular architecture. In contrast to Her2-positive cell lines, three-fold higher concentrations of 267 were required to achieve 50% inhibition of P-AKT when the drug was used in combination with Dt. In vivo studies focusing on low Her2-expressing breast cancer cells (LCC6) implanted orthotopically demonstrated that treatment with 267/Dt engendered improved therapeutic effects when compared with mice treated with either agent alone. CONCLUSIONS: The findings indicate that the 267/Dt drug combination confers increased (synergistic) therapeutic efficacy towards human breast cancer cells that express low levels of Her2.


Subject(s)
Actins/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Taxoids/administration & dosage , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Docetaxel , Drug Synergism , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Nude , Neoplasm Transplantation
13.
Clin Cancer Res ; 14(22): 7260-71, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-19010842

ABSTRACT

PURPOSE: To examine the antitumor effects of Irinophore C, a nanopharmaceutical formulation of irinotecan, on the tissue morphology and function of tumor vasculature in HT-29 human colorectal tumors. EXPERIMENTAL DESIGN: Fluorescence microscopy was used to map and quantify changes in tissue density, tumor vasculature, hypoxia, and the distribution of Hoechst 33342, a perfusion marker, and the anticancer drug, doxorubicin. Noninvasive magnetic resonance imaging was used to quantify Ktrans, the volume transfer constant of a solute between the blood vessels and extracellular tissue compartment of the tumor, as a measure of vascular function. Following treatment with Irinophore C, 19F magnetic resonance spectroscopy was used to monitor the delivery of 5-fluorouracil (5-FU) to the tumor tissue, whereas scintigraphy was used to quantify the presence of bound [14C]5-FU. RESULTS: Irinophore C decreased cell density (P = 8.42 x 10(-5)), the overall number of endothelial cells in the entire section (P = 0.014), tumor hypoxia (P = 5.32 x 10(-9)), and K(trans) (P = 0.050). However, treatment increased the ratio of endothelial cells to cell density (P = 0.00024) and the accumulation of Hoechst 33342 (P = 0.022), doxorubicin (P = 0.243 x 10(-5)), and 5-FU (P = 0.0002) in the tumor. Vascular endothelial growth factor and interleukin-8, two proangiogenic factors, were down-regulated, whereas the antiangiogenic factor TIMP-1 was up-regulated in Irinophore C-treated tumors. CONCLUSIONS: Irinophore C treatment improves the vascular function of the tumor, thereby reducing tumor hypoxia and increasing the delivery and accumulation of a second drug. Reducing hypoxia would enhance radiotherapy, whereas improving delivery of a second drug to the tumor should result in higher cell kill.


Subject(s)
Antineoplastic Agents/administration & dosage , Camptothecin/analogs & derivatives , Doxorubicin/pharmacokinetics , Fluorouracil/pharmacokinetics , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Camptothecin/administration & dosage , Camptothecin/pharmacokinetics , Camptothecin/therapeutic use , Cell Hypoxia/drug effects , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Irinotecan , Liposomes , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Mice , Nanocapsules , Neoplasms, Experimental/blood supply , Tissue Distribution , Xenograft Model Antitumor Assays
14.
Mol Cancer Ther ; 7(1): 59-70, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18202010

ABSTRACT

Integrin-linked kinase (ILK) was assesed as a therapeutic target in glioblastoma xenograft models through multiple endpoints including treatment related changes in the tumor microenvironment. Glioblastoma cell lines were tested in vitro for sensitivity toward the small-molecule inhibitors QLT0254 and QLT0267. Cell viability, cell cycle, and apoptosis were evaluated using MTT assay, flow cytometry, caspase activation, and DAPI staining. Western blotting and ELISA were used for protein analysis (ILK, PKB/Akt, VEGF, and HIF-1alpha). In vivo assessment of growth rate, cell proliferation, BrdUrd, blood vessel mass (CD31 labeling), vessel perfusion (Hoechst 33342), and hypoxia (EF-5) was done using U87MG glioblastoma xenografts in RAG2-M mice treated orally with QLT0267 (200 mg/kg q.d.). ILK inhibition in vitro with QLT0254 and QLT0267 resulted in decreased levels of phospho-PKB/Akt (Ser473), secreted VEGF, G2-M block, and apoptosis induction. Mice treated with QLT0267 exhibited significant delays in tumor growth (treated 213 mm3 versus control 549 mm3). In situ analysis of U87MG tumor cell proliferation from QLT0267-treated mice was significantly lower relative to untreated mice. Importantly, VEGF and HIF-1alpha expression decreased in QLT0267-treated tumors as did the percentage of blood vessel mass and numbers of Hoechst 33342 perfused tumor vessels compared with control tumors (35% versus 83%). ILK inhibition with novel small-molecule inhibitors leads to treatment-associated delays in tumor growth, decreased tumor angiogenesis, and functionality of tumor vasculature. The therapeutic effects of a selected ILK inhibitor (QLT0267) should be determined in the clinic in cancers that exhibit dysregulated ILK, such as PTEN-null glioblastomas.


Subject(s)
Glioblastoma/enzymology , Glioblastoma/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factors/metabolism , Animals , Cell Cycle/drug effects , Cell Hypoxia/drug effects , Cell Line, Tumor , Glioblastoma/blood supply , Glioblastoma/drug therapy , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Molecular Structure , Neovascularization, Pathologic/drug therapy , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Xenograft Model Antitumor Assays
15.
Breast Cancer Res Treat ; 106(3): 319-31, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17347776

ABSTRACT

Developing novel synergistic and more effective combination treatments is necessary for better management of breast cancer in the clinic. It is established that HER-2 overexpressing breast cancers are sensitive to the HER-1 (epidermal growth factor receptor (EGFR)) inhibitor gefitinib, but that this targeted agent produces only moderate therapeutic effects in vivo. Here, we use a model of ER(+) HER-2 overexpressing MCF-7 breast cancer (MCF-7(HER-2)) to identify, as broadly as possible, the in vivo microenvironmental and molecular therapeutic responses to gefitinib to predict a therapeutically viable target for gefitinib-based combination treatment. Our data show a link between in vivo reductions in tumor hypoxia (3-fold decrease, P = 0.002) and elevated activity of the mTOR pathway (3.8-fold increase in phospho-p70-S6K protein, P = 0.006) in gefitinib treated MCF-7(HER-2) tumors. Despite decreased levels of phosphorylated EGFR, HER-2 and Erk1/2 (P = 0.081, 0.005 and 0.034, respectively) the expression of phospho-AKT was not reduced in MCF-7(HER-2) tumors after gefitinib treatment. Levels of ERalpha receptor were, however, 1.8-fold higher in gefitinib treated compared to control tumors (P = 0.008). Based on these results we predict that gefitinib activity against ER(+) HER-2 overexpressing EGFR co-expressing breast cancers should be enhanced if used with agents that target the mTOR pathway. In vitro studies using MCF-7(HER-2) and BT474 breast cancer cells exposed to gefitinib and rapamycin in combination show that this combination produced significantly greater growth inhibitory effects than either of the drugs alone. Chou and Talalay analysis of the data suggested that combination of gefitinib and rapamycin was synergistic (CI < 1) at a number of selected drug ratios and over a broad range of effective doses.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Protein Kinases/physiology , Quinazolines/administration & dosage , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Sirolimus/administration & dosage , Animals , Breast Neoplasms/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Hypoxia , Cell Line, Tumor , ErbB Receptors/analysis , Female , Gefitinib , Humans , Mice , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , Quinazolines/therapeutic use , TOR Serine-Threonine Kinases
16.
Mol Cancer Ther ; 6(3): 844-55, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17339368

ABSTRACT

Various methods have been explored to enhance antibody-based cancer therapy. The use of multivalent antibodies or fragments against tumor antigens has generated a great deal of interest, as various cellular signals, including induction of apoptosis, inhibition of cell growth/survival, or internalization of the surface molecules, can be triggered or enhanced on extensive cross-linking of the target/antibody complex by the multivalent form of the antibody. The goal of the studies reported here was to develop multivalent antibody constructs via grafting of antibody molecules onto liposome membranes to enhance antibody activity. Using trastuzumab and rituximab as examples, up to a 25-fold increase in the antibody potency in cell viability assay was observed when the antibodies were presented in the multivalent liposome formulation. Key cell survival signaling molecules, such as phosphorylated Akt and phosphorylated p65 nuclear factor-kappaB, were down-regulated on treatment with multivalent liposomal trastuzumab and liposomal rituximab, respectively. Potent in vivo antitumor activity was shown for liposomal trastuzumab. The data presented here showed the potential of liposome technology to enhance the therapeutic effect of antibodies via a mechanism that modulates cell survival through clustering of the target/antibody complex.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Breast Neoplasms/therapy , Animals , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived , Antibodies, Neoplasm , Antigens, CD20/immunology , Antigens, Neoplasm/immunology , Blotting, Western , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Survival , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Down-Regulation , Female , Flow Cytometry , Genes, erbB-2/genetics , Genes, erbB-2/immunology , Humans , Liposomes , Mice , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/immunology , Rituximab , Signal Transduction , Transcription Factor RelA/metabolism , Trastuzumab
17.
BJU Int ; 99(5): 1154-60, 2007 May.
Article in English | MEDLINE | ID: mdl-17309552

ABSTRACT

OBJECTIVE: To evaluate hypoxia non-invasively in androgen-dependent (AD), regressing (6-days after castration, RG) and androgen-independent (AI) Shionogi tumours, using the radiolabelled tracer for hypoxia, 18F-EF5, and positron emission tomography (PET). MATERIALS AND METHODS: Groups of mice bearing AD, RG and AI Shionogi tumours were co-injected with 18F-EF5 and unlabelled EF5. The mice were imaged non-invasively with PET to examine the accumulation of 18F-EF5 in hypoxic regions of the tumour. The tumours were subsequently placed in a gamma-counter, or disaggregated for flow cytometry, to determine the levels of 18F-EF5 and the percentage of hypoxic cells present in the tumour, respectively. RESULTS: The mean (sd) levels of hypoxia in AD Shionogi tumours decreased significantly 6 days after androgen ablation as measured by flow cytometry, from 17.1 (4.77) to 1.74 (0.46)% (P=0.003). There were no significant differences in the levels of 18F-EF5 in the tissue between AD and RG tumours using region-of-interest analysis of PET images or gamma-counting, although the differences were significant when measured by flow cytometry. However, mean (sd) levels of hypoxia in AI Shionogi tumours were significantly higher than in AD tumours regardless of the analysis method; PET, 10.5 (4.93)x10(-5)) Bq/cm2 (P=0.017), flow cytometry, 42.98 (3.35)% (P<0.001), well count, 6.81 (1.17)x10(4) and 13.1 (1.99)x10(4) cpm/g, for AD and AI tumours, respectively (P<0.001). CONCLUSIONS: Differences in hypoxia between AD and AI, but not RG, Shionogi tumours can be detected non-invasively with 18F-EF5 and PET. As prostate tumours are hypoxic and the oxygen levels can change with androgen ablation, noninvasive imaging of hypoxia with PET and 18F-EF5 might ultimately have a prognostic and/or diagnostic role in the clinical management of the disease.


Subject(s)
Hypoxia/pathology , Positron-Emission Tomography/methods , Prostatic Neoplasms/pathology , Androgen Antagonists/administration & dosage , Androgens/metabolism , Animals , Flow Cytometry , Fluorodeoxyglucose F18 , Humans , Hypoxia/diagnostic imaging , Male , Mice , Oxygen/metabolism , Prostatic Neoplasms/diagnostic imaging , Radiopharmaceuticals , Sensitivity and Specificity
18.
Eur J Pharm Biopharm ; 65(3): 289-99, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17123800

ABSTRACT

The purpose of these studies was to design an intravenous drug formulation consisting of two active agents having synergistic in vitro activity. Specifically, we describe a novel drug combination consisting of a cytotoxic agent (vinorelbine) with an apoptosis-inducing lipid (phosphatidylserine, PS). In vitro cytotoxicity screening of PS and vinorelbine, alone and in combination, against human MDA435/LCC6 breast cancer and H460 lung cancer cells was used to identify the molar ratio of these two agents required for synergistic activity. PS and vinorelbine were co-formulated in a lipid-based system at the synergistic molar ratio and the pharmacokinetic and antitumor characteristics of the combination assessed in mice bearing H460 tumors. The cytotoxicity of the lipid, and the synergy between the lipid and vinorelbine, were specific to PS; these effects were not observed using control lipids. A novel formulation of PS, incorporated as a membrane component in liposomes, and encapsulating vinorelbine using a pH gradient based loading method was developed. The PS to vinorelbine ratio in this formulation was 1/1, a ratio that produced synergistic in vitro cytotoxicity over a broad concentration range. The vinorelbine and PS dual-agent treatment significantly delayed the growth of subcutaneous human H460 xenograft tumors in Rag2M mice compared to the same dose of free vinorelbine given alone or given as a cocktail of the free vinorelbine simultaneously with empty PS-containing liposomes. These studies demonstrate the potential to develop clinically relevant drug combinations identified using in vitro drug-drug interactions combined with lipid-based delivery systems to co-formulate drugs at their synergistic ratios.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lung Neoplasms/drug therapy , Phosphatidylserines/administration & dosage , Vinblastine/analogs & derivatives , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Drug Compounding , Drug Design , Drug Synergism , Female , Humans , Injections, Intravenous , Liposomes , Lung Neoplasms/pathology , Mice , Phosphatidylserines/chemistry , Phosphatidylserines/pharmacokinetics , Technology, Pharmaceutical , Vinblastine/administration & dosage , Vinblastine/chemistry , Vinblastine/pharmacokinetics , Vinorelbine , Xenograft Model Antitumor Assays
19.
Mol Cancer Ther ; 5(3): 645-54, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16546979

ABSTRACT

The present study uses cell-based screening assays to assess the anticancer effects of targeting phosphatidylinositol 3-kinase-regulated integrin-linked kinase (ILK) in combination with small-molecule inhibitors of Raf-1 or mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase kinase (MEK). The objective was to determine if synergistic interactions are achievable through the use of agents targeting two key cell signaling pathways involved in regulating glioblastoma cancer. The phosphatidylinositol 3-kinase/protein kinase B (PKB)/Akt and the Ras/MAPK pathway were targeted for their involvement in cell survival and cell proliferation, respectively. The glioblastoma cell lines U87MG, SF-188, and U251MG were transiently transfected with an antisense oligonucleotide targeting ILK (ILKAS) alone or in combination with the Raf-1 inhibitor GW5074 or with the MEK inhibitor U0126. Dose and combination effects were analyzed by the Chou and Talalay median-effect method and indicated that combinations targeting ILK with either Raf-1 or MEK resulted in a synergistic interaction. Glioblastoma cells transfected with ILKAS exhibited reduced levels of ILK and phosphorylated PKB/Akt on Ser473 but not PKB/Akt on Thr308 as shown by immunoblot analysis. These results were confirmed using glioblastoma cells transfected with ILK small interfering RNA, which also suggested enhanced gene silencing when used in combination with U0126. U87MG glioblastoma cells showed a 90% (P < 0.05) reduction in colony formation in soft agar with exposure to ILKAS in combination with GW5074 compared with control colonies. A substantial increase in Annexin V-positive cells as determined by using fluorescence-activated cell sorting methods were seen in combinations that included ILKAS. Combinations targeting ILK and components of the Ras/MAPK pathway result in synergy and could potentially be more effective against glioblastoma cancer than monotherapy.


Subject(s)
Central Nervous System Neoplasms/drug therapy , Glioblastoma/drug therapy , Indoles/therapeutic use , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Oligonucleotides, Antisense/therapeutic use , Phenols/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Butadienes/therapeutic use , Cell Proliferation/drug effects , Central Nervous System Neoplasms/enzymology , Glioblastoma/enzymology , Humans , Nitriles/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Transfection , ras Proteins/metabolism
20.
Curr Drug Deliv ; 2(4): 341-51, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16305437

ABSTRACT

The introduction of combination chemotherapeutic regimens for the treatment of childhood leukaemia in the 1960s provided the proof-of-principle that cytotoxic drugs were capable of curing cancer. However, in the four decades since this discovery, the majority of cancers still cannot be cured by chemotherapy. Clinical evidence supports the hypothesis of Goldie and Coldman that treating cancers with all the available effective agents simultaneously provides the greatest chance of eliciting a cure. Unfortunately, for traditional cytotoxic agents with narrow therapeutic indices, life-threatening toxicity precludes combination chemotherapy regimens employing multiple agents. This review discusses the concept of fixed dose combination chemotherapy with emphasis on capturing therapeutic efficacy described as synergistic as a basis for improving the effectiveness of combination chemotherapy. The use of lipid-based nanotechnologies, focusing on liposomes, as an enabling technology to facilitate the delivery of cytotoxic agents to the tumour site at concentrations and/or drug ratios judged to be synergistic will be discussed. It is envisaged that the development of this model system will be supported by cell-based screening technologies, pharmacokinetic and pharmacodynamic parameters and mathematical models describing therapeutic drug:drug interactions (the Median Effect Principle of Chou and Talalay). Experiments using preclinical models are presented to support the benefits of drug delivery systems as a foundation for fixed dose anticancer drug combinations. The ultimate goal of this research is to prepare a 'single vial' fixed dose combination product that encompasses both traditional cytotoxic agents and new molecularly targeted modalities with optimum therapeutic effects and acceptable toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Delivery Systems , Nanotechnology , Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Humans , Liposomes
SELECTION OF CITATIONS
SEARCH DETAIL
...