Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
3.
Phys Med Biol ; 61(24): 8736-8749, 2016 12 21.
Article in English | MEDLINE | ID: mdl-27893446

ABSTRACT

This study aimed to identify a set of stable radiomic parameters in CT perfusion (CTP) maps with respect to CTP calculation factors and image discretization, as an input for future prognostic models for local tumor response to chemo-radiotherapy. Pre-treatment CTP images of eleven patients with oropharyngeal carcinoma and eleven patients with non-small cell lung cancer (NSCLC) were analyzed. 315 radiomic parameters were studied per perfusion map (blood volume, blood flow and mean transit time). Radiomics robustness was investigated regarding the potentially standardizable (image discretization method, Hounsfield unit (HU) threshold, voxel size and temporal resolution) and non-standardizable (artery contouring and noise threshold) perfusion calculation factors using the intraclass correlation (ICC). To gain added value for our model radiomic parameters correlated with tumor volume, a well-known predictive factor for local tumor response to chemo-radiotherapy, were excluded from the analysis. The remaining stable radiomic parameters were grouped according to inter-parameter Spearman correlations and for each group the parameter with the highest ICC was included in the final set. The acceptance level was 0.9 and 0.7 for the ICC and correlation, respectively. The image discretization method using fixed number of bins or fixed intervals gave a similar number of stable radiomic parameters (around 40%). The potentially standardizable factors introduced more variability into radiomic parameters than the non-standardizable ones with 56-98% and 43-58% instability rates, respectively. The highest variability was observed for voxel size (instability rate >97% for both patient cohorts). Without standardization of CTP calculation factors none of the studied radiomic parameters were stable. After standardization with respect to non-standardizable factors ten radiomic parameters were stable for both patient cohorts after correction for inter-parameter correlations. Voxel size, image discretization, HU threshold and temporal resolution have to be standardized to build a reliable predictive model based on CTP radiomics analysis.


Subject(s)
Perfusion Imaging/methods , Tomography, X-Ray Computed/methods , Blood Volume , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/physiopathology , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , Lung Neoplasms/physiopathology , Oropharyngeal Neoplasms/diagnostic imaging , Oropharyngeal Neoplasms/pathology , Oropharyngeal Neoplasms/physiopathology , Prognosis , Tumor Burden
4.
Oncogene ; 30(25): 2823-35, 2011 Jun 23.
Article in English | MEDLINE | ID: mdl-21317922

ABSTRACT

Medulloblastoma (MB) is the most common malignant brain tumor in children. It is known that overexpression and/or amplification of the MYC oncogene is associated with poor clinical outcome, but the molecular mechanisms and the MYC downstream effectors in MB remain still elusive. Besides contributing to elucidate how progression of MB takes place, most importantly, the identification of novel MYC-target genes will suggest novel candidates for targeted therapy in MB. A group of 209 MYC-responsive genes was obtained from a complementary DNA microarray analysis of a MB-derived cell line, following MYC overexpression and silencing. Among the MYC-responsive genes, we identified the members of the bone morphogenetic protein (BMP) signaling pathway, which have a crucial role during the development of the cerebellum. In particular, the gene BMP7 was identified as a direct target of MYC. A positive correlation between MYC and BMP7 expression was documented by analyzing two distinct sets of primary MB samples. Functional studies in vitro using a small-molecule inhibitor of the BMP/SMAD signaling pathway reproduced the effect of the small interfering RNA-mediated silencing of BMP7. Both approaches led to a block of proliferation in a panel of MB cells and to inhibition of SMAD phosphorylation. Altogether, our findings indicate that high MYC levels drive BMP7 overexpression, promoting cell survival in MB cells. This observation suggests the potential relevance of targeting the BMP/SMAD pathway as a novel therapeutic approach for the treatment of childhood MB.


Subject(s)
Bone Morphogenetic Protein 7/genetics , Cell Survival/genetics , Cerebellar Neoplasms/genetics , Genes, myc , Medulloblastoma/genetics , Blotting, Western , Cerebellar Neoplasms/pathology , Child , Enzyme-Linked Immunosorbent Assay , Gene Silencing , Humans , Medulloblastoma/pathology , Phosphorylation , Smad Proteins/metabolism
5.
Eur J Cancer ; 43(12): 1833-41, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17627812

ABSTRACT

Loss of caspase-8 expression - which has been demonstrated in a subset of Medulloblastoma (MB) - might block important apoptotic signalling pathways and therefore contribute to treatment resistance. In this study, IFN-gamma mediated up-regulation of caspase-8 in human MB cells was found to result in chemosensitization to cisplatin, doxorubicin and etoposide, and sensitisation to radiation. These effects were more prominent in D425 and D341 MB cells (low basal caspase-8 expression) when compared to DAOY MB cells (high basal caspase-8 expression). IFN-gamma mediated chemosensitization and radiosensitization effects were reduced by treatment with the caspase-8 specific inhibitor z-IETD-fmk. Treatment of IFN-gamma resulted in activation of STAT1 in DAOY MB cells and to a lesser extent in D425, but not in D341, indicating that IFN-gamma acts in MB cells through STAT1-dependent and -independent signalling pathways. Taken together, our results demonstrate that IFN-gamma mediated restoration of caspase-8 in MB cells might enhance apoptotic pathways relevant to the response to chemo- and radiotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Caspase 8/metabolism , Cerebellar Neoplasms/metabolism , Interferon-gamma/pharmacology , Medulloblastoma/metabolism , Radiation-Sensitizing Agents/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/radiotherapy , Child , Female , Humans , Male , Medulloblastoma/drug therapy , Medulloblastoma/radiotherapy , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
6.
Gene Ther ; 14(15): 1143-51, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17495946

ABSTRACT

Standard treatment for glioblastoma multiforme and other brain tumors consists of surgical resection followed by combined radio-/chemotherapy. However, radiation resistance of tumor cells limits the success of this treatment, and the tumors invariably recur. Therefore, the selective inhibition of molecular mediators of radiation resistance may provide therapeutic benefit to the patient. One of these targets is the Rad51 protein, which is a key component of the homologous recombinational repair of DNA double-strand breaks. Here, we investigated whether post-transcriptional silencing of Rad51 by herpes simplex virus-type 1 (HSV-1) amplicon vector-mediated short interfering RNA expression can enhance the antitumor effect of radiation therapy. We demonstrate that these vectors specifically and efficiently inhibited the radiation-induced recruitment of Rad51 into nuclear foci in human glioma cells. The combination of vector-mediated silencing of Rad51 expression and treatment with ionizing radiation resulted in a pronounced reduction of the survival of human glioma cells in culture. In athymyc mice, a single intratumoral injection of Rad51-specific HSV-1 amplicon vector followed by a single radiation treatment resulted in a significant decrease in tumor size. In control animals, including mice that received an intratumoral injection of Rad51-specific amplicon vector but no radiation treatment, the tumor sizes increased.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Glioma/therapy , Herpesvirus 1, Human/genetics , Rad51 Recombinase/antagonists & inhibitors , Animals , Apoptosis , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Cell Line, Tumor , Combined Modality Therapy , Female , Gene Expression , Gene Silencing , Genetic Vectors/genetics , Glioblastoma/therapy , Glioma/pathology , Glioma/radiotherapy , Humans , Injections, Intralesional , Mice , Mice, Nude , RNA Interference , RNA, Small Interfering/genetics , Rad51 Recombinase/genetics , Radiation Tolerance/physiology , Treatment Outcome
8.
Ann Rheum Dis ; 62(12): 1139-44, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14644850

ABSTRACT

OBJECTIVE: To analyse the functional response of p53 in rheumatoid arthritis synovial fibroblasts (RASF) in vitro and in vivo and to investigate whether activation of p53 modulates the destructive process of RASF. METHODS: RASF and controls grown on chamber slides were either directly examined with DO7 anti-p53 antibodies by immunofluorescence or irradiated with 10 Gy x rays and analysed time dependently for the expression of p53. The percentage of positive cells was evaluated by a quantitative scoring system. RASF and normal (N) SF cultured in vitro were co-implanted with human cartilage in SCID mice for 60 days. Consecutively, the invasion score was evaluated, and the number of p53 positive cells was determined at the sites of invasion by immunohistochemistry. In addition, synovial tissues from RA, osteoarthritis, and normal synovia were stained with DO7 antibodies. RESULTS: In vitro the rate of expression of p53 in RASF was low (<5%), but transiently inducible by ionising irradiation (50%). In vitro low p53 expressing RASF disclosed, when invading articular cartilage, a nuclear p53 signal in 20% of the cells, indicating the induction of p53 in a distinct population of RASF during the invasive process. CONCLUSIONS: These data suggest an inductive p53 response at sites of cartilage invasion during the destructive process driven by activated RASF.


Subject(s)
Arthritis, Rheumatoid/metabolism , Fibroblasts/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Cells, Cultured , Fibroblasts/pathology , Fibroblasts/radiation effects , Gene Expression Regulation , Genes, p53 , Humans , Immunohistochemistry , Mice , Mice, SCID , Synovial Membrane/metabolism , Synovial Membrane/pathology
9.
Br J Cancer ; 88(11): 1785-92, 2003 Jun 02.
Article in English | MEDLINE | ID: mdl-12771996

ABSTRACT

Mistletoe extracts are used as alternative cancer treatment in addition to standard chemotherapy and radiation treatment and have an immunostimulatory and pain-relieving effect. A direct antitumour effect of mistletoe extracts against tumour cells of lymphoid origin has been linked to the D-galactoside-specific mistletoe lectin I. In this study, we investigated the cellular effect of bacterially expressed, recombinant mistletoe lectin alone or in combination with ionising radiation in a genetically defined p53-wild-type and p53-deficient E1A/ras-transformed murine tumour cells system. Downregulation of the proliferative activity and cell killing by recombinant mistletoe lectin occurred in a clear dose response (0.1-1 ng ml(-1)). Induction of apoptosis was p53-independent, but apoptosis-associated factor-1-dependent. Cellular treatment with lectin in combination with ionising radiation resulted in both p53-wild-type and p53-deficient tumour cells in an at least additive, antiproliferative effect and enhanced activation of caspase-3. Combined treatment with ionising radiation and lectin revealed a similar cytotoxic effect in human, p53-mutated adenocarcinoma cells. Thus, recombinant mistletoe lectin alone and in combination with ionising radiation bypasses often prevalent apoptotic deficiencies in treatment-resistant tumour cells.


Subject(s)
Apoptosis/drug effects , Plant Lectins/pharmacology , Plant Preparations/pharmacology , Plant Proteins , Toxins, Biological/pharmacology , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Annexin A5/metabolism , Apoptotic Protease-Activating Factor 1 , Blotting, Western , Caspase 3 , Caspases/metabolism , Cell Division/drug effects , Cell Division/radiation effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Fibroblasts/metabolism , Genes, ras , Humans , Membrane Potentials/drug effects , Membrane Potentials/radiation effects , Mice , Mistletoe/chemistry , Mitochondria/drug effects , Mitochondria/radiation effects , Mutation/genetics , Proteins/genetics , Proteins/metabolism , Radiation, Ionizing , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Ribosome Inactivating Proteins, Type 2 , Sensitivity and Specificity , Tumor Cells, Cultured
10.
Curr Med Chem Anticancer Agents ; 3(1): 35-46, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12678913

ABSTRACT

Cell cycle checkpoints are activated in response to DNA-damage to ensure that accurate copies of the cellular genome are passed on to the next generation and to avoid replication and segregation of damaged DNA. These cellular control systems can be overcome by combining conventional DNA-damaging agents with compounds that target the cell cycle regulatory pathways, to enhance cytotoxicity. Tumor cells often comprise a corrupted G(1) cell cycle checkpoint while the G(2) cell cycle checkpoint is still intact. This review describes the concept of G(2) checkpoint abrogation with recognized (methylxanthines, UCN-01) and novel G(2) checkpoint abrogators to potentiate the cytotoxicity of DNA-damaging drugs and ionizing radiation. It illustrates the potential of G(2) checkpoint abrogators to preferentially sensitize p53-mutated, treatment resistant tumor cells for genotoxic treatment. Identification of the targets of caffeine and UCN-01 to be key-players of the G(2) checkpoint (ATM/ATR and Chk1, respectively) promoted the search for novel inhibitors of this checkpoint. Even though a direct causal link between G(2) checkpoint abrogation and chemo-/radiosensitization is difficult to prove the multitude of these novel compounds validate that inhibition of critical elements of the G(2) checkpoint (ATM/ATR-Chk1/Chk2-CDC25C-cascade) potentiates the cytotoxicity of DNA-damaging agents.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , DNA Damage/drug effects , G2 Phase/drug effects , Alkaloids , Animals , Antineoplastic Agents/chemistry , Caffeine/chemistry , Caffeine/pharmacology , Cell Survival/drug effects , Combined Modality Therapy , DNA Damage/radiation effects , Drug Synergism , Humans , Staurosporine/analogs & derivatives , Xanthines/chemistry , Xanthines/pharmacology
11.
Curr Med Chem Anticancer Agents ; 2(6): 727-42, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12678723

ABSTRACT

DNA double strand breaks are the pivotal cellular damage induced by ionizing radiation. A plethora of molecular and cellular processes are activated as part of the cellular stress response that result in cell cycle arrest and induction of the DNA-repair machinery to restore the damage of DNA or to activate a cell death program. However ionizing radiation also initiates signal transduction cascades that are generated at cellular sites distant from and independent of DNA-damage. These signaling processes are similar to hormone activated growth factor receptor controlled signal transduction cascades and represent interesting targets for anticancer treatment modalities combining ionizing radiation with molecular defined pharmacological compounds. Activation of these signal transduction cascades upon irradiation or upregulation of growth factor mediated pathways due to oncogene-transformation often contribute to an acquired or inherent treatment resistance in malignant cells. Therefore pharmacological compounds inhibiting specific key-entities of these signal transduction cascades potentially sensitize for radiation induced cell death. Here we describe current preclinical concepts of combined treatment strategies with locoregional-applied ionizing radiation and molecular defined signal transduction inhibitors to overcome a high treatment threshold in tumor cells.


Subject(s)
Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Signal Transduction/drug effects , Animals , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Ligands , Neoplasms/drug therapy , Radiation-Sensitizing Agents/therapeutic use , Radiotherapy/adverse effects , Radiotherapy/methods
12.
Br J Cancer ; 85(12): 2010-6, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11747347

ABSTRACT

The vascular endothelial growth factor (VEGF) receptor is a major target for anti-angiogenesis-based cancer treatment. Here we report the treatment effect of ionizing radiation in combination with the novel orally bioavailable VEGF receptor tyrosine kinase inhibitor PTK787/ZK222584 on endothelial cell proliferation in vitro and with tumour xenografts in vivo. Combined treatment of human umbilical vein endothelial cells with increasing doses of PTK787/ZK222584 and ionizing radiation abrogated VEGF-dependent proliferation in a dose-dependent way, but inhibition of endothelial cell proliferation was not due to apoptosis induction. In vivo, a combined treatment regimen of PTK787/ZK222584 (4 x 100 mg/kg) during 4 consecutive days in combination with ionizing radiation (4 x 3 Gy) exerted a substantial tumour growth delay for radiation-resistant p53-dysfunctional tumour xenografts derived from SW480 colon adenocarcinoma cells while each treatment modality alone had only a minimal effect on tumour size and neovascularization. SW480 tumours from animals that received a combined treatment regimen, displayed not only an extended tumour growth delay but also a significant decrease in the number of microvessels in the tumour xenograft. These results support the model of a cooperative anti-tumoral effect of angiogenesis inhibitor and irradiation and show that the orally bioavailable VEGF receptor tyrosine kinase inhibitor PTK787/ZK222584 is suitable for combination therapy with irradiation.


Subject(s)
Adenocarcinoma/drug therapy , Angiogenesis Inhibitors/therapeutic use , Colonic Neoplasms/drug therapy , Endothelium, Vascular/drug effects , Enzyme Inhibitors/therapeutic use , Neoplasm Proteins/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Phthalazines/therapeutic use , Pyridines , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , Adenocarcinoma/blood supply , Adenocarcinoma/pathology , Adenocarcinoma/radiotherapy , Administration, Oral , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Cell Division/drug effects , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Colonic Neoplasms/radiotherapy , Combined Modality Therapy , Endothelium, Vascular/cytology , Endothelium, Vascular/radiation effects , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/radiotherapy , Phthalazines/administration & dosage , Phthalazines/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Radiotherapy, Adjuvant , Receptors, Vascular Endothelial Growth Factor , Xenograft Model Antitumor Assays
13.
Cancer Res ; 61(22): 8203-10, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11719451

ABSTRACT

Activation of the phosphatidylinositol 3'-kinase (PI3K)/Akt survival pathway protects against apoptotic stress stimuli. Therefore, compounds that down-regulate this pathway are of clinical interest for single and combined anticancer treatment modalities. Here we demonstrate that the cytotoxic effect of the protein kinase C (PKC)-inhibitor N-benzoylated staurosporine (PKC412) is mediated via the PI3K/Akt pathway. Dose-dependent down-regulation of the proliferative activity, activation of the apoptotic machinery, and cell killing by PKC412 (0-1 microM) in Rat1a-fibroblasts and H-ras-oncogene-transformed fibroblasts correlated with a decrease of Akt phosphorylation and a reduced phosphorylation of the endogenous Akt-substrate GSK3-alpha. Expression of the dominant-active myristoylated form of Akt abrogated this cytotoxic effect of PKC412. Experiments with Apaf-1-deficient cells revealed that PKC412-induced cytotoxicity depends on an intact apoptosome but that the decrease of Akt phosphorylation is not attributable to apoptosis execution. Comparative experiments indicate that PKC412 and the parent-compound staurosporine down-regulate this survival pathway upstream or at the level of Akt but by a different mechanism than the PI3K-inhibitor LY294002. Furthermore, inhibition of this pathway by PKC412 is relevant for sensitization to ionizing radiation. These results demonstrate the specific role of this signaling pathway for the PKC412-mediated down-regulation of an apoptotic threshold and its cytotoxicity.


Subject(s)
Enzyme Inhibitors/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Protein Kinase C/antagonists & inhibitors , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/physiology , Signal Transduction/drug effects , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Animals , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Chromones/pharmacology , Down-Regulation/drug effects , Genes, ras/physiology , Humans , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Signal Transduction/physiology , Tumor Cells, Cultured
14.
Diabetes ; 50(9): 2105-13, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11522678

ABSTRACT

Several studies support the concept of a diabetic cardiomyopathy in the absence of discernible coronary artery disease, although its mechanism remains poorly understood. We investigated the role of glucose and palmitic acid on cardiomyocyte apoptosis and on the organization of the contractile apparatus. Exposure of adult rat cardiomyocytes for 18 h to palmitic acid (0.25 and 0.5 mmol/l) resulted in a significant increase of apoptotic cells, whereas increasing glucose concentration to 33.3 mmol/l for up to 8 days had no influence on the apoptosis rate. However, both palmitic acid and elevated glucose concentration alone or in combination had a dramatic destructive effect on the myofibrillar apparatus. The membrane-permeable C2-ceramide but not the metabolically inactive C2-dihydroceramide enhanced apoptosis of cardiomyocytes by 50%, accompanied by detrimental effects on the myofibrils. The palmitic acid-induced effects were impaired by fumonisin B1, an inhibitor of ceramide synthase. Sphingomyelinase, which activates the catabolic pathway of ceramide by metabolizing sphingomyeline to ceramide, did not adversely affect cardiomyocytes. Palmitic acid-induced apoptosis was accompanied by release of cytochrome c from the mitochondria. Aminoguanidine did not prevent glucose-induced myofibrillar degeneration, suggesting that formation of nitric oxide and/or advanced glycation end products play no major role. Taken together, these results suggest that in adult rat cardiac cells, palmitic acid induces apoptosis via de novo ceramide formation and activation of the apoptotic mitochondrial pathway. Conversely, glucose has no influence on adult cardiomyocyte apoptosis. However, both cell nutrients promote degeneration of myofibrils. Thus, gluco- and lipotoxicity may play a central role in the development of diabetic cardiomyopathy.


Subject(s)
Apoptosis/drug effects , Glucose/pharmacology , Heart/physiology , Myofibrils/drug effects , Palmitic Acid/pharmacology , Animals , Cells, Cultured , Ceramides/physiology , Cytochrome c Group/metabolism , Female , Heart/drug effects , Myocardium/cytology , Myocardium/metabolism , Myofibrils/pathology , Myofibrils/physiology , Oleic Acids/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
16.
Int J Radiat Oncol Biol Phys ; 49(2): 561-7, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11173155

ABSTRACT

In many human hematologic and solid malignancies, intrinsic or acquired treatment resistance remains a major obstacle for successful cancer therapy. The molecular understanding of how tumor cells respond to chemotherapy and ionizing radiation is rapidly evolving. Induction of programmed cell death, apoptosis, is one important strategy for successful cancer therapy. This has been shown convincingly for oncogene-transformed normal cells as well as tumor cells of lymphoid origin. However, the relevance of apoptosis in solid human malignancies is less clear. Loss of apoptosis might be linked to specific mutations in the often tissue-specific apoptotic pathways due to aberrations in the stress-related signal transduction cascades. Restoration of a dysfunctional apoptotic program in cancer tissue where apoptosis has been identified as an important mechanism for tissue homeostasis is one rational approach for innovative cancer therapy. In this review, we focus on the relevance of the tumor suppressor p53 for apoptosis-induction and successful cancer therapy outlining the importance of an intact caspase machinery for apoptosis execution. Strategies are discussed to overcome treatment resistance and a high apoptotic threshold in human malignancies where apoptosis is the dominant mode of cell death and the status of p53 is an important determinant for apoptosis induction.


Subject(s)
Apoptosis/genetics , Caspases/physiology , Genes, p53/physiology , Genetic Therapy/methods , Genetic Vectors/therapeutic use , Neoplasm Proteins/physiology , Neoplasms/therapy , Adenoviridae/genetics , Animals , Apoptosis/physiology , Cell Cycle/physiology , DNA Repair , Enzyme Activation , Enzyme Inhibitors/therapeutic use , Genes, p53/genetics , Genetic Vectors/genetics , Humans , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/radiotherapy , Staurosporine/therapeutic use
17.
Cancer Res ; 61(2): 732-8, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11212276

ABSTRACT

The cellular response to ionizing radiation is governed by the DNA-damage recognition process but is also modulated by cytoplasmic signal transduction cascades that are part of the cellular stress response. Growth-promoting protein kinase C activity antagonizes irradiation-induced cell death, and, therefore, protein kinase C inhibitors might be potent radiosensitizers. The antiproliferative and radiosensitizing effect of the novel N-benzoylated staurosporine analogue PKC412 was tested in vitro against genetically defined p53-wild type (+/+) and p53-deficient (-/-) murine fibrosarcoma cells and in vivo against radioresistant p53-/- murine fibrosarcoma and human colon adenocarcinoma tumor xenograft (SW480, p53-mutated). PKC412 sensitized both p53+/+ and p53-/- tumor cells in vitro and in vivo for treatment with ionizing radiation but with a different mechanism of radiosensitization depending on the p53 status. In p53+/+, cells combined treatment with PKC412 and ionizing radiation drastically induced apoptotic cell death, whereas no apoptosis induction could be observed in p53-deficient cells in vitro and in histological tumor sections. Combined treatment resulted in an increased G2 cell cycle distribution in p53-/- cells at PKC412 concentrations that did not alter cell cycle distribution when applied alone. In vivo, a minimal treatment regimen during 4 consecutive days of PKC412 (4 x 100 mg/kg) in combination with ionizing radiation (4 x 3 Gy) exerted a substantial tumor growth delay for both p53-disfunctional tumor xenografts and showed that the clinically relevant protein kinase C inhibitor PKC412 is a promising new radiosensitizer with a potentially broad therapeutic window.


Subject(s)
Enzyme Inhibitors/pharmacology , Protein Kinase C/antagonists & inhibitors , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Tumor Suppressor Protein p53/physiology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cells, Cultured , Combined Modality Therapy , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Genotype , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Neoplasms, Experimental/radiotherapy , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
18.
Cell Growth Differ ; 11(9): 491-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11007454

ABSTRACT

Caspases are a family of cysteine proteases that constitute the apoptotic cell death machinery. We report the importance of the cytochrome c-mediated caspase-9 death pathway for radiosensitization by the protein kinase C (PKC) inhibitors staurosporine (STP) and PKC-412. In our genetically defined tumor cells, treatment with low doses of STP or the conventional PKC-specific inhibitor PKC-412 in combination with irradiation (5 Gy) potently reduced viability, enhanced mitochondrial cytochrome c release into the cytosol, and specifically stimulated the initiator caspase-9. Whereas treatment with each agent alone had a minimal effect, combined treatment resulted in enhanced caspase-3 activation. This was prevented by broad-range and specific caspase-9 inhibitors and absent in caspase-9-deficient cells. The tumor suppressor p53 was required for apoptosis induction by combined treatment but was dispensable for dose-dependent STP-induced caspase activation. These results demonstrate the requirement for an intact caspase-9 pathway for apoptosis-based radiosensitization by PKC inhibitors and show that STP induces apoptosis independent of p53.


Subject(s)
Apoptosis/drug effects , Apoptosis/radiation effects , Caspases/drug effects , Caspases/radiation effects , Cytochrome c Group/drug effects , Cytochrome c Group/radiation effects , Protein Kinase C/antagonists & inhibitors , Radiation Tolerance/drug effects , Staurosporine/analogs & derivatives , Animals , Apoptosis/physiology , Caspase 9 , Caspases/metabolism , Cytochrome c Group/metabolism , Enzyme Inhibitors/pharmacology , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/radiotherapy , Protein Kinase C/metabolism , Radiation Tolerance/physiology , Staurosporine/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/radiation effects , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
19.
Anticancer Drug Des ; 15(1): 17-28, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10888033

ABSTRACT

The staurosporine derivative PKC412 was originally identified as an inhibitor of protein kinase C (PKC) and subsequently shown to inhibit other kinases including the kinase insert domain receptor (KDR) (vascular endothelial growth factor receptor, VEGF-R2), the receptor of platelet-derived growth factor, and the receptor for the stem cell factor, c-kit. PKC412 showed a broad antiproliferative activity against various tumor and normal cell lines in vitro, and was able to reverse the Pgp-mediated multidrug resistance of tumor cells in vitro. Exposure of cells to PKC412 resulted in a dose-dependent increase in the G2/M phase of the cell cycle concomitant with increased polyploidy, apoptosis and enhanced sensitivity to ionizing radiation. PKC412 displayed a potent antitumor activity as single agent and was able to potentiate the antitumor activity of some of the clinically used cytotoxins (Taxol and doxorubicin) in vivo. The combined treatment of PKC412 with loco-regional ionizing irradiation showed significant antitumor activity against tumors which are resistant to both ionizing radiation and chemotherapeutic agents (dysfunctional p53). The finding that PKC412 is an inhibitor of the VEGF-mediated cellular signaling via inhibition of KDR and PKC in vitro is consistent with the in vivo inhibition of VEGF-dependent angiogenesis in a growth factor implant model. Orally administered PKC412 also strongly inhibited retinal neovascularization as well as laser-induced choroidal neovascularization in murine models. In summary, PKC412 may suppress tumor growth by inhibiting tumor angiogenesis in addition to directly-inhibiting tumor cell proliferation via its effects on PKC and/or other protein kinases. PKC412 is currently in Phase I clinical trials for treatment of advanced cancer as well as for the treatment of ischemic retinopathy.


Subject(s)
Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Protein Kinase C/antagonists & inhibitors , Staurosporine/analogs & derivatives , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Blood Proteins/metabolism , Cell Cycle/drug effects , Clinical Trials, Phase I as Topic , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/therapeutic use , Humans , Mice , Neoplasms/pathology , Protein Binding , Staurosporine/metabolism , Staurosporine/pharmacology , Staurosporine/therapeutic use , Tumor Cells, Cultured
20.
Br J Cancer ; 80(5-6): 693-8, 1999 May.
Article in English | MEDLINE | ID: mdl-10360645

ABSTRACT

p53 mutations are among the most common genetic alterations in human cancer and are frequently described in intrinsic or acquired radio- and chemotherapy resistance. Radiation-induced cell kill is not only mediated by DNA damage but also by the activation of signal transduction cascades generated at the plasma membrane like the sphingomyelin pathway. We used genetically defined wild-type p53 or p53-deficient mouse fibrosarcoma cells to investigate the p53-dependence of tumour response upon activation of the sphingomyelin pathway. Treatment of the tumour cells with neutral sphingomyelinase drastically reduced the amount of wild-type p53 fibrosarcoma cell proliferation over 72 h in a clear dose-response (0.2-1.0 U ml(-1) nSMase). Sphingomyelinase had no effect on cell proliferation in tumour cells lacking p53. Similarly, cell proliferation was abolished by C2-ceramide (5-20 microM) only in wild-type p53 cells. FACS-analysis revealed that C2-ceramide induced massive p53-dependent apoptosis (40-50% after 12-24 h) and cell cycle analysis showed a transient G1 arrest in p53-deficient tumour cells 12-24 h after C2-ceramide exposure. These results suggest that ceramide-induced apoptosis in tumour cells can be dependent on the status of p53 and imply that p53 is also important for stress-induced apoptotic signal transduction cascades generated at the plasma membrane.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Fibrosarcoma/genetics , Fibrosarcoma/pathology , Sphingosine/analogs & derivatives , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Fibrosarcoma/drug therapy , Growth Inhibitors/pharmacology , Mice , Mice, Knockout , Signal Transduction/drug effects , Signal Transduction/genetics , Sphingomyelin Phosphodiesterase/pharmacology , Sphingomyelins/physiology , Sphingosine/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...