Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Drug Metab Pharmacokinet ; 52: 100515, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37481830

ABSTRACT

Intravenously infused treosulfan was evaluated in adult and pediatric patients for conditioning regimen prior to allogeneic hematopoietic stem cell transplantation. A population pharmacokinetic (PK) model was initially developed on 116 adult and pediatric PK profiles from historical trials, to support treosulfan dose recommendations for children in 2 prospective trials. The aim was to assess and update the initial population PK model by inclusion of additional 83 pediatric PK profiles from these 2 trials. The final population PK model was 2-compartmental with dosing in the central compartment, linear elimination, and inter-compartmental clearance. Inter-individual variability was included on clearance (CL), central volume (V1), peripheral volume (V2), and inter-compartmental clearance (Q). The final model described an effect of the body surface area (BSA) on CL, V1, V2, and Q. The final model resulted in a modified dose recommendation for children and advises treosulfan doses of 10 g/m2, 12 g/m2, and 14 g/m2 for BSAs of <0.4 m2, ≥0.4 to <0.9 m2, and ≥0.9 m2, respectively. This simplified BSA-dependent dose recommendation was developed for children, ensuring a well comparable treosulfan exposure as a dose of 14 g/m2 in adults - irrespective of their age and without applying individual therapeutic drug monitoring.


Subject(s)
Hematopoietic Stem Cell Transplantation , Transplantation Conditioning , Adult , Humans , Child , Prospective Studies , Transplantation Conditioning/methods , Busulfan/pharmacokinetics , Busulfan/therapeutic use
2.
Cancers (Basel) ; 15(11)2023 Jun 03.
Article in English | MEDLINE | ID: mdl-37297005

ABSTRACT

T-cell lymphomas are heterogeneous and rare lymphatic malignancies with unfavorable prognosis. Consequently, new therapeutic strategies are needed. The enhancer of zeste homologue 2 (EZH2) is the catalytic subunit of the polycomb repressive complex 2 and responsible for lysine 27 trimethylation of histone 3. EZH2 is overexpressed in several tumor entities including T-cell neoplasms leading to epigenetic and consecutive oncogenic dysregulation. Thus, pharmacological EZH2 inhibition is a promising target and its clinical evaluation in T-cell lymphomas shows favorable results. We have investigated EZH2 expression in two cohorts of T-cell lymphomas by mRNA-profiling and immunohistochemistry, both revealing overexpression to have a negative impact on patients' prognosis. Furthermore, we have evaluated EZH2 inhibition in a panel of leukemia and lymphoma cell lines with a focus on T-cell lymphomas characterized for canonical EZH2 signaling components. The cell lines were treated with the inhibitors GSK126 or EPZ6438 that inhibit EZH2 specifically by competitive binding at the S-adenosylmethionine (SAM) binding site in combination with the common second-line chemotherapeutic oxaliplatin. The change in cytotoxic effects under pharmacological EZH2 inhibition was evaluated revealing a drastic increase in oxaliplatin resistance after 72 h and longer periods of combinational incubation. This outcome was independent of cell type but associated to reduced intracellular platinum. Pharmacological EZH2 inhibition revealed increased expression in SRE binding proteins, SREBP1/2 and ATP binding cassette subfamily G transporters ABCG1/2. The latter are associated with chemotherapy resistance due to increased platinum efflux. Knockdown experiments revealed that this was independent of the EZH2 functional state. The EZH2 inhibition effect on oxaliplatin resistance and efflux was reduced by additional inhibition of the regulated target proteins. In conclusion, pharmacological EZH2 inhibition is not suitable in combination with the common chemotherapeutic oxaliplatin in T-cell lymphomas revealing an EZH2-independent off-target effect.

3.
Br J Clin Pharmacol ; 88(4): 1722-1734, 2022 02.
Article in English | MEDLINE | ID: mdl-34519068

ABSTRACT

AIMS: The aim of this work is the development of a mechanistic physiologically-based pharmacokinetic (PBPK) model using in vitro to in vivo extrapolation to conduct a drug-drug interaction (DDI) assessment of treosulfan against two cytochrome p450 (CYP) isoenzymes and P-glycoprotein (P-gp) substrates. METHODS: A PBPK model for treosulfan was developed de novo based on literature and unpublished clinical data. The PBPK DDI analysis was conducted using the U.S. Food and Drug Administration (FDA) DDI index drugs (probe substrates) midazolam, omeprazole and digoxin for CYP3A4, CYP2C19 and P-gp, respectively. Qualified and documented PBPK models of the probe substrates have been adopted from an open-source online model database. RESULTS: The PBPK model for treosulfan, based on both in vitro and in vivo data, was able to predict the plasma concentration-time profiles and exposure levels of treosulfan applied for a standard conditioning treatment. Medium and low potentials for DDI on CYP3A4 (maximum area under the concentration-time curve ratio (AUCRmax = 2.23) and CYP2C19 (AUCRmax = 1.6) were predicted, respectively, using probe substrates midazolam and omeprazole. Treosulfan was not predicted to cause a DDI on P-gp. CONCLUSION: Medicinal products with a narrow therapeutic index (eg, digoxin) that are substrates for CYP3A4, CYP2C19 or P-gp should not be given during treatment with treosulfan. However, considering the comprehensive treosulfan-based conditioning treatment schedule and the respective pharmacokinetic properties of the concomitantly used drugs (eg, half-life), the potential for interaction on all evaluated mechanisms would be low (AUCR < 1.25), if concomitantly administered drugs are dosed either 2 hours before or 8 hours after the 2-hour intravenous infusion of treosulfan.


Subject(s)
Cytochrome P-450 CYP3A , Midazolam , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Busulfan/analogs & derivatives , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP3A/metabolism , Digoxin , Drug Interactions , Humans , Midazolam/pharmacokinetics , Models, Biological , Omeprazole , Pharmaceutical Preparations
4.
Gynecol Oncol ; 159(3): 850-859, 2020 12.
Article in English | MEDLINE | ID: mdl-32980128

ABSTRACT

OBJECTIVES: Extending the therapeutic spectrum of PARP-inhibitors (PARPi) beyond BRCA1-deficiency and/or overcoming PARPi-resistance is of high clinical interest. This is particularly true for the identification of innovative therapeutic strategies for ovarian cancer, given the recent advances in the use of PARPi in clinical practice. In this regard, the combination of PARPi with chemotherapy is a possible strategy for defining new therapeutic standards. In this study, we analyzed the therapeutic effect of novel triazene derivatives, including the drug CT913 and its metabolite CT913-M1 on ovarian cancer cells and describe their interaction with the PARPi olaparib. METHODS: In vitro assays for drug characterization including RNA-Seq were applied in a selected panel of ovarian cancer cell lines. RESULTS: CT913 treatment conferred a dose-dependent reduction of cell viability in a set of platinum-sensitive and platinum-resistant ovarian cancer cell lines with an IC50 in the higher micromolar range (553-1083 µM), whereas its metabolite CT913-M1 was up to 69-fold more potent, especially among long-term treatment (IC50 range: 8-138 µM). Neither of the drugs sensitized for cisplatin. CT913 conferred synthetic lethality in BRCA1-deficient ovarian cancer cells, indicating that its effect is augmented by a deficiency in homologous recombination repair (HR). Furthermore, CT913 showed a synergistic interaction with olaparib, independently of BRCA1 mutational status. CT913 strongly induced CDKN1A transcription, suggesting cell cycle arrest as an early response to this drug. It moreover downregulated a variety of transcripts involved in DNA-repair pathways. CONCLUSIONS: This is the first study, suggesting the novel triazene drug CT913 as enhancer drug for extending the therapeutic spectrum of PARPi.


Subject(s)
Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Triazenes/pharmacology , BRCA1 Protein/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mutation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phthalazines/pharmacology , Phthalazines/therapeutic use , Piperazines/pharmacology , Piperazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , RNA-Seq , Recombinational DNA Repair/drug effects , Synthetic Lethal Mutations/drug effects , Triazenes/therapeutic use
6.
Cell Death Discov ; 6: 12, 2020.
Article in English | MEDLINE | ID: mdl-32194992

ABSTRACT

Oncogenic KRAS mutations are encountered in more than 90% of pancreatic ductal adenocarcinomas. MEK inhibition has failed to procure any clinical benefits in mutant RAS-driven cancers including pancreatic ductal adenocarcinoma (PDAC). To identify potential resistance mechanisms underlying MEK inhibitor (MEKi) resistance in PDAC, we investigated lysosomal drug accumulation in PDAC models both in vitro and in vivo. Mouse PDAC models and human PDAC cell lines as well as human PDAC xenografts treated with the MEK inhibitor trametinib or refametinib led to an enhanced expression of lysosomal markers and enrichment of lysosomal gene sets. A time-dependent, increase in lysosomal content was observed upon MEK inhibition. Strikingly, there was a strong activation of lysosomal biogenesis in cell lines of the classical compared to the basal-like molecular subtype. Increase in lysosomal content was associated with nuclear translocation of the Transcription Factor EB (TFEB) and upregulation of TFEB target genes. siRNA-mediated depletion of TFEB led to a decreased lysosomal biogenesis upon MEK inhibition and potentiated sensitivity. Using LC-MS, we show accumulation of MEKi in the lysosomes of treated cells. Therefore, MEK inhibition triggers lysosomal biogenesis and subsequent drug sequestration. Combined targeting of MEK and lysosomal function may improve sensitivity to MEK inhibition in PDAC.

7.
Mol Oncol ; 14(4): 686-703, 2020 04.
Article in English | MEDLINE | ID: mdl-32037720

ABSTRACT

Platinum-based compounds remain a well-established chemotherapy for cancer treatment despite their adverse effects which substantially restrict the therapeutic windows of the drugs. Both the cell type-specific toxicity and the clinical responsiveness of tumors have been associated with mechanisms that alter drug entry and export. We sought to identify pharmacological agents that promote cisplatin (CP) efficacy by augmenting the levels of drug-induced DNA lesions in malignant cells and simultaneously protecting normal tissues from accumulating such damage and from functional loss. Formation and persistence of platination products in the DNA of individual nuclei were measured in drug-exposed cell lines, in primary human tumor cells and in tissue sections using an immunocytochemical method. Using a mouse model of CP-induced toxicity, the antihistaminic drug diphenhydramine (DIPH) and two methylated derivatives decreased DNA platination in normal tissues and also ameliorated nephrotoxicity, ototoxicity, and neurotoxicity. In addition, DIPH sensitized multiple cancer cell types, particularly ovarian cancer cells, to CP by increasing intracellular uptake, DNA platination, and/or apoptosis in cell lines and in patient-derived primary tumor cells. Mechanistically, DIPH diminished transport capacity of CP efflux pumps MRP2, MRP3, and MRP5 particularly in its C2+C6 bimethylated form. Overall, we demonstrate that DIPH reduces side effects of platinum-based chemotherapy and simultaneously inhibits key mechanisms of platinum resistance. We propose that measuring DNA platination after ex vivo exposure may predict the responsiveness of individual tumors to DIPH-like modulators.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Diphenhydramine/pharmacology , Histamine H1 Antagonists/pharmacology , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Line, Tumor , Cisplatin/toxicity , DNA Adducts/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Female , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology
8.
Biol Blood Marrow Transplant ; 24(8): 1651-1656, 2018 08.
Article in English | MEDLINE | ID: mdl-29753157

ABSTRACT

This multicenter study evaluated a treosulfan-based regimen in children and young adults with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS) undergoing allogeneic hematopoietic cell transplant (HCT). Forty patients with median age 11 years (range, 1 to 19) underwent allogeneic HCT for AML in first (n = 18), second (n = 11), and third or greater remission (n = 3) or MDS (n = 8) using bone marrow (n = 25), peripheral blood stem cells (n = 5), or cord blood (n = 9). The regimen consisted of body surface area (BSA)-based treosulfan 10 g/m2/day (BSA ≤ .5 m2), 12 g/m2/day (BSA > .5 to 1.0 m2), or 14 g/m2/day (BSA > 1.0 m2) on days -6 to -4; fludarabine 30 mg/m2/day on days -6 to -2; and a single fraction of 200 cGy total body irradiation on day -1. Graft-versus-host disease (GVHD) prophylaxis included tacrolimus and methotrexate for marrow and peripheral blood stem cell and cyclosporine/mycophenolate mofetil for cord blood. One-year overall survival, disease-free survival, and nonrelapse mortality were 80%, 73%, and 3%, respectively. One-year relapse was 38% for AML and 13% for MDS. No serious organ toxicities were observed. All 37 assessable patients engrafted. Cumulative incidences of grades II to IV acute GVHD and chronic GVHD were 22% and 40%, respectively. BSA-based treosulfan dosing resulted in predictable area under the curve and maximum concentration, which is required for dosing without measuring individual pharmacokinetic parameters. Observed differences in pharmacokinetics did not impact disease control or regimen toxicity. This BSA-based treosulfan regimen resulted in excellent engraftment and disease-free survival and minimal toxicity and transplant-related mortality (3%) in children and young adults with AML and MDS.


Subject(s)
Busulfan/analogs & derivatives , Hematopoietic Stem Cell Transplantation/methods , Leukemia, Myeloid, Acute/therapy , Myelodysplastic Syndromes/therapy , Whole-Body Irradiation , Adolescent , Busulfan/administration & dosage , Busulfan/therapeutic use , Child , Child, Preschool , Female , Graft Survival , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/mortality , Humans , Infant , Leukemia, Myeloid, Acute/mortality , Male , Myelodysplastic Syndromes/mortality , Survival Analysis , Transplantation, Homologous , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use , Young Adult
9.
Ther Drug Monit ; 39(6): 617-624, 2017 12.
Article in English | MEDLINE | ID: mdl-28937535

ABSTRACT

BACKGROUND: The value of therapeutic drug monitoring (TDM) for paclitaxel (PTX) was recently demonstrated in the largest TDM trial ever conducted in oncology. The trial demonstrated significant reduction in neuropathy when using TDM. Dose adjustment for PTX was based on time above a threshold concentration (Tc>0.05). Tc>0.05 must be calculated with a pharmacokinetic model and complex nonlinear mixed-effects software. The use of the software and chromatographic methods to measure PTX requires specialized expertise. User-friendly methods to quantitate PTX and calculate Tc>0.05 could simplify the introduction of TDM into routine clinical practice. METHODS: The immunoassay (MyPaclitaxel) was used to quantitate PTX in samples from the clinical trial; the results were used to calculate Tc>0.05 using a stand-alone computer program with a simple, friendly graphical user interface for nonlinear mixed-effects pharmacokinetic calculations (MyCare Drug Exposure Calculator). The resulting dose recommendations from the calculated Tc>0.05 were compared with those using liquid chromatography-ultraviolet detection and NONMEM to examine the efficacy of the simpler tools for TDM. RESULTS: There was a good agreement between the immunoassay and liquid chromatography-ultraviolet detection: Passing-Bablok regression slope was 1.045 and intercept was -6.00, R was 0.9757, and mean bias was -1.77 ng/mL (-2.07 nmol/L). Dosing recommendations were identical for 70% of the cycles and within 10% for 89% of the samples. All Tc>0.05 values were at the same or adjacent medical decision points. CONCLUSIONS: MyPaclitaxel assay and MyCare Drug Exposure Calculator are convenient, user-friendly tools that may be suitable for routine TDM of PTX in clinical care.


Subject(s)
Antineoplastic Agents, Phytogenic/blood , Antineoplastic Agents, Phytogenic/pharmacokinetics , Decision Support Techniques , Drug Monitoring/methods , Paclitaxel/blood , Paclitaxel/pharmacokinetics , Humans , Immunoassay/methods , Reproducibility of Results , Software
10.
J Biomed Nanotechnol ; 12(1): 56-68, 2016 Jan.
Article in English | MEDLINE | ID: mdl-27301172

ABSTRACT

The anti-cancer drug oxaliplatin (OxP) has rarely been used to treat breast carcinoma, as it cannot cross the BBB to treat the frequently subsequent brain metastases. Here, we encapsulated OxP in liposomes prepared to reduce side effects and to simultaneously treat primary tumor and brain metastasis. The angiopep LRP-receptor ligand was bound to the vesicular surface for targeting. Targeted and non-targeted OxP liposomes were tested in vitro (binding, uptake, and transcytosis) and in vivo. Liposomes contained 0.65 mg OxP/mL, their mean diameter was 165 nm, and they released 50% of OxP within 8 days at 4 degrees C and within 22 h at 36 degrees C. MDCK cells were used for uptake and transcytosis quantification. Compared to non-targeted liposomes, targeted liposomes showed 12-fold greater uptake, and 2.25-fold higher transcytosis. In vivo efficacy was tested using human MT-3 breast cancer cells transplanted subcutaneously and intracerebrally into female nude mice, and tumor growth inhibition was measured. OxP was injected (6 mg OxP/kg) four times. The best results were obtained with targeted liposomes (T/C: 21% for subcutaneous and 50% for intracerebral). OxP liposomes with a fluid membrane all inhibited MT-3 tumors significantly better than free OxP, with no significant difference between targeted and non-targeted liposomes. The therapeutic effect was accompanied with strong leukopenia and mild thrombocytopenia with all formulations. The newly developed OxP liposomes significantly improved the treatment of subcutaneously and intracerebrally growing breast cancer, but the targeted angiopep-equipped liposomes showed no superior effect in vivo.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Breast Neoplasms/drug therapy , Liposomes/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Organoplatinum Compounds/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Brain Neoplasms/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Diffusion , Female , Humans , Metallothionein 3 , Mice , Mice, Nude , Molecular Targeted Therapy/methods , Organoplatinum Compounds/chemistry , Oxaliplatin , Treatment Outcome
11.
J Inorg Biochem ; 153: 247-252, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26364260

ABSTRACT

Intracellular binding of cisplatin to non-DNA partners, such as proteins, has received increasing attention as an additional mode of action and as mechanism of resistance. We investigated two cisplatin-interacting isoforms of protein disulfide isomerase regarding their contribution to acquired cisplatin resistance using sensitive and resistant A2780/A2780cis ovarian cancer cells. Cisplatin cytotoxicity was assessed after knockdown of either protein disulfide isomerase family A member 1 (PDIA1) or protein disulfide isomerase family A member 3 (PDIA3). Whereas PDIA1 knockdown led to increased cytotoxicity in resistant A2780cis cells, PDIA3 knockdown showed no influence on cytotoxicity. Coincubation with propynoic acid carbamoyl methyl amide 31 (PACMA31), a PDIA1 inhibitor, resensitized A2780cis cells to cisplatin treatment. Determination of the combination index revealed that the combination of cisplatin and PACMA31 acts synergistically. Our results warrant further evaluation of PDIA1 as promising target for chemotherapy, and its inhibition by PACMA31 as a new therapeutic approach.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Procollagen-Proline Dioxygenase/metabolism , Protein Disulfide-Isomerases/metabolism , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cisplatin/chemistry , Enzyme Inhibitors/pharmacology , Humans , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Procollagen-Proline Dioxygenase/genetics , Protein Binding , Protein Disulfide-Isomerases/antagonists & inhibitors , Protein Disulfide-Isomerases/genetics
12.
Cell Cycle ; 13(10): 1560-70, 2014.
Article in English | MEDLINE | ID: mdl-24626197

ABSTRACT

Glioblastoma are the most frequent and malignant human brain tumors, having a very poor prognosis. The enhanced radio- and chemoresistance of glioblastoma and the glioblastoma stem cells might be the main reason why conventional therapies fail. The second messenger cyclic AMP (cAMP) controls cell proliferation, differentiation, and apoptosis. Downregulation of cAMP sensitizes tumor cells for anti-cancer treatment. Opioid receptor agonists triggering opioid receptors can activate inhibitory Gi proteins, which, in turn, block adenylyl cyclase activity reducing cAMP. In this study, we show that downregulation of cAMP by opioid receptor activation improves the effectiveness of anti-cancer drugs in treatment of glioblastoma. The µ-opioid receptor agonist D,L-methadone sensitizes glioblastoma as well as the untreatable glioblastoma stem cells for doxorubicin-induced apoptosis and activation of apoptosis pathways by reversing deficient caspase activation and deficient downregulation of XIAP and Bcl-xL, playing critical roles in glioblastomas' resistance. Blocking opioid receptors using the opioid receptor antagonist naloxone or increasing intracellular cAMP by 3-isobutyl-1-methylxanthine (IBMX) strongly reduced opioid receptor agonist-induced sensitization for doxorubicin. In addition, the opioid receptor agonist D,L-methadone increased doxorubicin uptake and decreased doxorubicin efflux, whereas doxorubicin increased opioid receptor expression in glioblastomas. Furthermore, opioid receptor activation using D,L-methadone inhibited tumor growth significantly in vivo. Our findings suggest that opioid receptor activation triggering downregulation of cAMP is a promising strategy to inhibit tumor growth and to improve the effectiveness of anti-cancer drugs in treatment of glioblastoma and in killing glioblastoma stem cells.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Cyclic AMP/metabolism , Glioblastoma/pathology , Receptors, Opioid, mu/genetics , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Synergism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Methadone/pharmacology , Methadone/therapeutic use , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism
13.
PLoS One ; 8(9): e74555, 2013.
Article in English | MEDLINE | ID: mdl-24040280

ABSTRACT

Pancreatic cancer is one of the leading cancer-related causes of death in the western world with an urgent need for new treatment strategies. Recently, hyperforin and nemorosone have been described as promising anti-cancer lead compounds. While hyperforin has been thoroughly investigated in vitro and in vivo, in vivo data for nemorosone are still missing. Thus, we investigated the growth-inhibitory potential of nemorosone on pancreatic cancer xenografts in NMRI nu/nu mice and determined basic pharmacokinetic parameters. Xenograft tumors were treated with nemorosone and gemcitabine, the current standard of care. Tumor sections were subjected to H&E as well as caspase 3 and Ki-67 staining. Nemorosone plasma kinetics were determined by HPLC and mass spectrometry. Induction of CYP3A4 and other metabolizing enzymes by nemorosone and hyperforin was tested on primary hepatocytes using qRT-PCR. At a dose of 50 mg/kg nemorosone per day, a significant growth-inhibitory effect was observed in pancreatic cancer xenografts. The compound was well tolerated and rapidly absorbed into the bloodstream with a half-life of approximately 30 min. Different metabolites were detected, possibly resembling CYP3A4-independent oxidation products. It is concluded that nemorosone is a potential anti-cancer lead compound with good bioavailability, little side-effects and promising growth-inhibitory effects, thus representing a valuable compound for a combination therapy approach.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Benzophenones/pharmacokinetics , Carcinoma/drug therapy , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Benzophenones/administration & dosage , Benzophenones/blood , Biological Availability , Biotransformation , Carcinoma/blood , Carcinoma/pathology , Caspase 3/genetics , Caspase 3/metabolism , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Administration Schedule , Female , Gene Expression , Half-Life , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Injections, Subcutaneous , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Mice , Mice, Nude , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/pathology , Phloroglucinol/administration & dosage , Phloroglucinol/analogs & derivatives , Primary Cell Culture , Terpenes/administration & dosage , Transplantation, Heterologous , Xenograft Model Antitumor Assays , Gemcitabine
14.
Oncotarget ; 4(5): 677-90, 2013 May.
Article in English | MEDLINE | ID: mdl-23633472

ABSTRACT

Cyclic AMP (cAMP) regulates a number of cellular processes and modulates cell death induction. cAMP levels are altered upon stimulation of specific G-protein-coupled receptors inhibiting or activating adenylyl cyclases. Opioid receptor stimulation can activate inhibitory Gi-proteins which in turn block adenylyl cyclase activity reducing cAMP. Opioids such as D,L-methadone induce cell death in leukemia cells. However, the mechanism how opioids trigger apoptosis and activate caspases in leukemia cells is not understood. In this study, we demonstrate that downregulation of cAMP induced by opioid receptor activation using the opioid D,L-methadone kills and sensitizes leukemia cells for doxorubicin treatment. Enhancing cAMP levels by blocking opioid-receptor signaling strongly reduced D,L-methadone-induced apoptosis, caspase activation and doxorubicin-sensitivity. Induction of cell death in leukemia cells by activation of opioid receptors using the opioid D,L-methadone depends on critical levels of opioid receptor expression on the cell surface. Doxorubicin increased opioid receptor expression in leukemia cells. In addition, the opioid D,L-methadone increased doxorubicin uptake and decreased doxorubicin efflux in leukemia cells, suggesting that the opioid D,L-methadone as well as doxorubicin mutually increase their cytotoxic potential. Furthermore, we found that opioid receptor activation using D,L-methadone alone or in addition to doxorubicin inhibits tumor growth significantly in vivo. These results demonstrate that opioid receptor activation via triggering the downregulation of cAMP induces apoptosis, activates caspases and sensitizes leukemia cells for doxorubicin treatment. Hence, opioid receptor activation seems to be a promising strategy to improve anticancer therapies.


Subject(s)
Adenylyl Cyclases/metabolism , Apoptosis , Cyclic AMP/metabolism , Leukemia, B-Cell/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptors, Opioid/metabolism , Adenylyl Cyclase Inhibitors , Analgesics, Opioid/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Caspases/metabolism , Cell Line, Tumor , Down-Regulation , Doxorubicin/blood , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Enzyme Activation/drug effects , Humans , Male , Methadone/blood , Methadone/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Receptors, Opioid/biosynthesis , Signal Transduction/drug effects
15.
PLoS One ; 8(1): e54193, 2013.
Article in English | MEDLINE | ID: mdl-23349823

ABSTRACT

INTRODUCTION: Inherent and acquired cisplatin resistance reduces the effectiveness of this agent in the management of non-small cell lung cancer (NSCLC). Understanding the molecular mechanisms underlying this process may result in the development of novel agents to enhance the sensitivity of cisplatin. METHODS: An isogenic model of cisplatin resistance was generated in a panel of NSCLC cell lines (A549, SKMES-1, MOR, H460). Over a period of twelve months, cisplatin resistant (CisR) cell lines were derived from original, age-matched parent cells (PT) and subsequently characterized. Proliferation (MTT) and clonogenic survival assays (crystal violet) were carried out between PT and CisR cells. Cellular response to cisplatin-induced apoptosis and cell cycle distribution were examined by FACS analysis. A panel of cancer stem cell and pluripotent markers was examined in addition to the EMT proteins, c-Met and ß-catenin. Cisplatin-DNA adduct formation, DNA damage (γH2AX) and cellular platinum uptake (ICP-MS) was also assessed. RESULTS: Characterisation studies demonstrated a decreased proliferative capacity of lung tumour cells in response to cisplatin, increased resistance to cisplatin-induced cell death, accumulation of resistant cells in the G0/G1 phase of the cell cycle and enhanced clonogenic survival ability. Moreover, resistant cells displayed a putative stem-like signature with increased expression of CD133+/CD44+cells and increased ALDH activity relative to their corresponding parental cells. The stem cell markers, Nanog, Oct-4 and SOX-2, were significantly upregulated as were the EMT markers, c-Met and ß-catenin. While resistant sublines demonstrated decreased uptake of cisplatin in response to treatment, reduced cisplatin-GpG DNA adduct formation and significantly decreased γH2AX foci were observed compared to parental cell lines. CONCLUSION: Our results identified cisplatin resistant subpopulations of NSCLC cells with a putative stem-like signature, providing a further understanding of the cellular events associated with the cisplatin resistance phenotype in lung cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Neoplastic Stem Cells/metabolism , AC133 Antigen , Aldehyde Dehydrogenase/metabolism , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Adducts/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Glycoproteins/metabolism , Homeodomain Proteins/metabolism , Humans , Hyaluronan Receptors/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Peptides/metabolism , Proto-Oncogene Proteins c-met/metabolism , SOXB1 Transcription Factors/metabolism , beta Catenin/metabolism
17.
Graefes Arch Clin Exp Ophthalmol ; 251(1): 279-84, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22782301

ABSTRACT

OBJECTIVE: In conjunctival melanoma, local chemotherapy has been based so far on clinical evidence and limited to the therapy of melanoma in situ. Our aim was to define substances that may have the potential to add to therapeutic options in extended local growth and metastatic disease. Two conjunctival cell lines (CRMM-1 and CRMM-2) have been established from recurrent conjunctival melanoma. In this study, we examined the chemosensitivity of these cell lines to different cytotoxic substances. MATERIALS AND METHODS: The cell lines CRMM-1 and CRMM-2 were exposed to chemotherapeutics for 24 h and the IC50 was generated. Sulforhodamin-B assays were used for quantification of in vitro efficacy. Time of exposure and escalating concentrations of the substances were adapted to the experimental setting. RESULTS: Bortezomib, clusianone 502 (nemorosone), ranpirnase, and sorafenib were efficient in inhibiting the growth of conjunctival melanoma cell lines. The IC50 achieved concentrations below or around 10 µM for these substances. CONCLUSIONS: Bortezomib, clusianone 502, ranpirnase, and sorafenib inhibited growth in conjunctival melanoma cell lines efficiently. The new substances may be a suitable alternative for local therapy. New therapeutic options with highly specific targeted agents for metastatic disease have to be evaluated in further experiments.


Subject(s)
Antineoplastic Agents/therapeutic use , Conjunctival Neoplasms/drug therapy , Melanoma/drug therapy , Benzophenones/therapeutic use , Boronic Acids/therapeutic use , Bortezomib , Cell Proliferation/drug effects , Conjunctival Neoplasms/pathology , Drug Screening Assays, Antitumor , Humans , Melanoma/pathology , Niacinamide/analogs & derivatives , Niacinamide/therapeutic use , Phenylurea Compounds/therapeutic use , Pyrazines/therapeutic use , Ribonucleases/therapeutic use , Sorafenib , Tumor Cells, Cultured
18.
Br J Ophthalmol ; 96(4): 591-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22275346

ABSTRACT

OBJECTIVE: Two conjunctival cell lines (CRMM-1 and CRMM-2) have been established from recurrent conjunctival melanoma. The authors examined the chemosensitivity of these cell lines to cytotoxic substances and combinations to identify substances that inhibit cell growth efficiently in vitro. MATERIAL AND METHODS: CRMM-1 and CRMM-2 were exposed to cisplatin, mitomycin C (MMC), all-trans-retinoic-acid (ATRA), fotemustine or imatinib for 24 h. Sulforhodamine-B assays were used to assess the IC(50). Isobolograms were performed to test possible synergism and antagonism with ATRA or imatinib. RESULTS: Cisplatin and MMC were efficient to inhibit the growth of CRMM-1 and CRMM-2. Combination of imatinib with MMC showed additive antitumoral effect on both cell lines. Combined treatment of imatinib with fotemustine or cisplatin resulted in antagonism. Strong antagonisms were also obtained with ATRA and fotemustine or cisplatin in both cell lines. A synergism was found for ATRA and mitomycin or imatinib in CRMM-2, in contrast to CRMM-1, where antagonism was obtained. CONCLUSIONS: Cisplatin and MMC inhibit cell growth in conjunctival melanoma cell lines. The potential of ATRA was evident only in combination with MMC or imatinib in CRMM-2 cells. Imatinib and mitomycin increased their efficiency under combination therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Conjunctival Neoplasms/pathology , Melanoma/pathology , Cell Line, Tumor , Cell Proliferation , Conjunctival Neoplasms/drug therapy , Drug Synergism , Drug Therapy, Combination , Humans , Melanoma/drug therapy
20.
Cancer Chemother Pharmacol ; 69(3): 753-61, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22020317

ABSTRACT

PURPOSE: S-1 is a novel oral fluoropyrimidine comprised of FT and two modulators, gimeracil (CDHP) and oteracil potassium (Oxo). This study investigated the food effects on the pharmacokinetics (PK) of Oxo, other components of S-1, and their metabolites at different gastric pH adjusted by proton pump inhibitor (PPI). METHODS: Patients with and without PPI were treated with S-1 at 30 mg/m(2) twice daily orally on days 1-7 under either fed or fasting condition, and then were crossed over to fasting/fed conditions on days 15-21 with washout on days 8-14 and 22-28. RESULTS: The study enrolled 55 patients including 27 PK-evaluable patients. For the single-dose and multiple-dose pharmacokinetics, the administration of S-1 under fed conditions resulted in decreased exposure to Oxo relative to fasting administration. There was a marginal decrease in exposure to CDHP and 5-FU under fed versus fasting conditions, although FT exposure was not altered by food, which demonstrated lack of food effect. PPI administration together with S-1 did not significantly change its bioavailability. CONCLUSIONS: Oxo exposure was reduced under fed compared to fasting condition. To increase the bioavailability of S-1, the administration of S-1 under fasting condition was more effective in the western countries.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Food-Drug Interactions , Neoplasms/drug therapy , Oxonic Acid/pharmacokinetics , Proton Pump Inhibitors/pharmacology , Tegafur/pharmacokinetics , Administration, Oral , Adult , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/therapeutic use , Biological Availability , Cross-Over Studies , Drug Administration Schedule , Drug Combinations , Fasting , Female , Humans , Male , Middle Aged , Oxonic Acid/administration & dosage , Oxonic Acid/adverse effects , Oxonic Acid/therapeutic use , Proton Pump Inhibitors/administration & dosage , Proton Pump Inhibitors/therapeutic use , Tegafur/administration & dosage , Tegafur/adverse effects , Tegafur/therapeutic use , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...