Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 538(7626): 477-482, 2016 10 27.
Article in English | MEDLINE | ID: mdl-27760111

ABSTRACT

Avoidance of apoptosis is critical for the development and sustained growth of tumours. The pro-survival protein myeloid cell leukemia 1 (MCL1) is overexpressed in many cancers, but the development of small molecules targeting this protein that are amenable for clinical testing has been challenging. Here we describe S63845, a small molecule that specifically binds with high affinity to the BH3-binding groove of MCL1. Our mechanistic studies demonstrate that S63845 potently kills MCL1-dependent cancer cells, including multiple myeloma, leukaemia and lymphoma cells, by activating the BAX/BAK-dependent mitochondrial apoptotic pathway. In vivo, S63845 shows potent anti-tumour activity with an acceptable safety margin as a single agent in several cancers. Moreover, MCL1 inhibition, either alone or in combination with other anti-cancer drugs, proved effective against several solid cancer-derived cell lines. These results point towards MCL1 as a target for the treatment of a wide range of tumours.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Models, Biological , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/pathology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Thiophenes/pharmacology , Thiophenes/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Female , Humans , Leukemia/drug therapy , Leukemia/metabolism , Leukemia/pathology , Lymphoma/drug therapy , Lymphoma/metabolism , Lymphoma/pathology , Male , Mice , Models, Molecular , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Myeloid Cell Leukemia Sequence 1 Protein/chemistry , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms/metabolism , Pyrimidines/administration & dosage , Thiophenes/administration & dosage , Xenograft Model Antitumor Assays , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
2.
Lancet Oncol ; 21(6): e297, 2020 06.
Article in English | MEDLINE | ID: mdl-32502449
3.
N Engl J Med ; 376(1): 96-7, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28052222
5.
JCO Precis Oncol ; 8: e2300117, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38207228

ABSTRACT

Greater collaboration needed to realize potential of molecular profiling initiatives for pediatric cancers.


Subject(s)
Neoplasms , Humans , Child , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/therapy , Precision Medicine
6.
JAMA Netw Open ; 6(7): e2324977, 2023 07 03.
Article in English | MEDLINE | ID: mdl-37505498

ABSTRACT

Importance: The development of oncology drugs is expensive and beset by a high attrition rate. Analysis of the costs and causes of translational failure may help to reduce attrition and permit the more appropriate use of resources to reduce mortality from cancer. Objective: To analyze the causes of failure and expenses incurred in clinical trials of novel oncology drugs, with the example of insulin-like growth factor-1 receptor (IGF-1R) inhibitors, none of which was approved for use in oncology practice. Design, Setting, and Participants: In this cross-sectional study, inhibitors of the IGF-1R and their clinical trials for use in oncology practice between January 1, 2000, and July 31, 2021, were identified by searching PubMed and ClinicalTrials.gov. A proprietary commercial database was interrogated to provide expenses incurred in these trials. If data were not available, estimates were made of expenses using mean values from the proprietary database. A search revealed studies of the effects of IGF-1R inhibitors in preclinical in vivo assays, permitting calculation of the percentage of tumor growth inhibition. Archival data on the clinical trials of IGF-1R inhibitors and proprietary estimates of their expenses were examined, together with an analysis of preclinical data on IGF-1R inhibitors obtained from the published literature. Main Outcomes and Measures: Expenses associated with research and development of IGF-1R inhibitors. Results: Sixteen inhibitors of IGF-1R studied in 183 clinical trials were found. None of the trials, in a wide range of tumor types, showed efficacy permitting drug approval. More than 12 000 patients entered trials of IGF-1R inhibitors in oncology indications in 2003 to 2021. These trials incurred aggregate research and development expenses estimated at between $1.6 billion and $2.3 billion. Analysis of the results of preclinical in vivo assays of IGF-1R inhibitors that supported subsequent clinical investigations showed mixed activity and protocols that poorly reflected the treatment of advanced metastatic tumors in humans. Conclusions and Relevance: Failed drug development in oncology incurs substantial expense. At an industry level, an estimated $50 billion to $60 billion is spent annually on failed oncology trials. Improved target validation and more appropriate preclinical models are required to reduce attrition, with more attention to decision-making before launching clinical trials. A more appropriate use of resources may better reduce cancer mortality.


Subject(s)
Insulin-Like Growth Factor I , Neoplasms , Humans , Cross-Sectional Studies , Insulin-Like Growth Factor I/antagonists & inhibitors , Neoplasms/drug therapy
7.
Nat Rev Drug Discov ; 21(12): 915-931, 2022 12.
Article in English | MEDLINE | ID: mdl-36195754

ABSTRACT

Successful drug discovery is like finding oases of safety and efficacy in chemical and biological deserts. Screens in disease models, and other decision tools used in drug research and development (R&D), point towards oases when they score therapeutic candidates in a way that correlates with clinical utility in humans. Otherwise, they probably lead in the wrong direction. This line of thought can be quantified by using decision theory, in which 'predictive validity' is the correlation coefficient between the output of a decision tool and clinical utility across therapeutic candidates. Analyses based on this approach reveal that the detectability of good candidates is extremely sensitive to predictive validity, because the deserts are big and oases small. Both history and decision theory suggest that predictive validity is under-managed in drug R&D, not least because it is so hard to measure before projects succeed or fail later in the process. This article explains the influence of predictive validity on R&D productivity and discusses methods to evaluate and improve it, with the aim of supporting the application of more effective decision tools and catalysing investment in their creation.


Subject(s)
Drug Discovery , Efficiency , Humans , Drug Discovery/methods
8.
Proc Natl Acad Sci U S A ; 105(6): 2169-74, 2008 Feb 12.
Article in English | MEDLINE | ID: mdl-18250306

ABSTRACT

Maturation of neuronal synapses is thought to involve mitochondria. Bcl-xL protein inhibits mitochondria-mediated apoptosis but may have other functions in healthy adult neurons in which Bcl-xL is abundant. Here, we report that overexpression of Bcl-xL postsynaptically increases frequency and amplitude of spontaneous miniature synaptic currents in rat hippocampal neurons in culture. Bcl-xL, overexpressed either pre or postsynaptically, increases synapse number, the number and size of synaptic vesicle clusters, and mitochondrial localization to vesicle clusters and synapses, likely accounting for the changes in miniature synaptic currents. Conversely, knockdown of Bcl-xL or inhibiting it with ABT-737 decreases these morphological parameters. The mitochondrial fission protein, dynamin-related protein 1 (Drp1), is a GTPase known to localize to synapses and affect synaptic function and structure. The effects of Bcl-xL appear mediated through Drp1 because overexpression of Drp1 increases synaptic markers, and overexpression of the dominant-negative dnDrp1-K38A decreases them. Furthermore, Bcl-xL coimmunoprecipitates with Drp1 in tissue lysates, and in a recombinant system, Bcl-xL protein stimulates GTPase activity of Drp1. These findings suggest that Bcl-xL positively regulates Drp1 to alter mitochondrial function in a manner that stimulates synapse formation.


Subject(s)
Dynamins/physiology , Hippocampus/metabolism , Synapses , bcl-X Protein/physiology , Animals , Cells, Cultured , Hippocampus/cytology , Mitochondria/metabolism , Rats , Synaptic Transmission
9.
Eur J Cancer ; 150: 95-98, 2021 06.
Article in English | MEDLINE | ID: mdl-33892410

ABSTRACT

Two recent policy documents by the European Union, 'Europe's Beating Cancer Plan' and its accompanying 'Conquering Cancer: Mission Possible' (CCMP), articulate broad policies aimed at reducing cancer mortality across Europe, for example, by promoting prevention and early detection. The focus for cancer treatment in these manifestos is the expansion of personalised cancer medicine (PCM). However, the CCMP document suggests that the uptake of PCM is "hampered by uncertainty about its outcomes". What are these outcomes and why this uncertainty? We address the limits of PCM in pathology-driven and pathology-agnostic PCM, briefly discussing the results of umbrella and basket trials. We suggest that the complexity, plasticity and genetic heterogeneity of advanced cancers will continue to thwart the impact of PCM, limiting it to specific pathologies, or rare subsets of them. Caution regarding the advancement of PCM is justified, and policymakers should be wary of the hype of lobbyists, who do not acknowledge the limits of PCM.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Precision Medicine , Antineoplastic Agents/adverse effects , Drug Resistance, Neoplasm , Europe , European Union , Genetic Heterogeneity , Genetic Predisposition to Disease , Humans , Molecular Targeted Therapy , Neoplasms/pathology , Patient Safety , Phenotype , Precision Medicine/adverse effects , Risk Assessment
10.
Breast Cancer Res ; 10(6): R101, 2008.
Article in English | MEDLINE | ID: mdl-19055754

ABSTRACT

INTRODUCTION: Basal-like carcinomas (BLCs) and human epidermal growth factor receptor 2 overexpressing (HER2+) carcinomas are the subgroups of breast cancers that have the most aggressive clinical behaviour. In contrast to HER2+ carcinomas, no targeted therapy is currently available for the treatment of patients with BLCs. In order to discover potential therapeutic targets, we aimed to discover deregulated signalling pathways in human BLCs. METHODS: In this study, we focused on the oncogenic phosphatidylinositol 3-kinase (PI3K) pathway in 13 BLCs, and compared it with a control series of 11 hormonal receptor negative- and grade III-matched HER2+ carcinomas. The two tumour populations were first characterised by immunohistochemistry and gene expression. The PI3K pathway was then investigated by gene copy-number analysis, gene expression profiling and at a proteomic level using reverse-phase protein array technology and tissue microarray. The effects of the PI3K inhibition pathway on proliferation and apoptosis was further analysed in three human basal-like cell lines. RESULTS: The PI3K pathway was found to be activated in BLCs and up-regulated compared with HER2+ tumours as shown by a significantly increased activation of the downstream targets Akt and mTOR (mammalian target of rapamycin). BLCs expressed significantly lower levels of the tumour suppressor PTEN and PTEN levels were significantly negatively correlated with Akt activity within that population. PTEN protein expression correlated significantly with PTEN DNA copy number and more importantly, reduced PTEN DNA copy numbers were observed specifically in BLCs. Similar to human samples, basal-like cell lines exhibited an activation of PI3K/Akt pathway and low/lack PTEN expression. Both PI3K and mTOR inhibitors led to basal-like cell growth arrest. However, apoptosis was specifically observed after PI3K inhibition. CONCLUSIONS: These data provide insight into the molecular pathogenesis of BLCs and implicate the PTEN-dependent activated Akt signalling pathway as a potential therapeutic target for the management of patients with poor prognosis BLCs.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Neoplasms, Basal Cell/genetics , Neoplasms, Basal Cell/metabolism , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Apoptosis , Blotting, Western , Breast Neoplasms/pathology , Cell Proliferation , Enzyme Activation , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Dosage , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Neoplasms, Basal Cell/pathology , Oligonucleotide Array Sequence Analysis , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Array Analysis , Protein Kinases/genetics , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , TOR Serine-Threonine Kinases , Tissue Array Analysis , Tumor Cells, Cultured
11.
Curr Opin Genet Dev ; 12(1): 67-72, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11790557

ABSTRACT

Avoidance of apoptosis is a cornerstone of tumourigenesis. The functions of key molecules that either sense DNA damage or that commit cells to die are lost during tumorigenesis. Frequently, during tumourigenesis, cells increase their survival signals. The initiation of apoptosis by DNA damage signals was shown to absolutely depend on the expression of either Bax or Bak. Single oncogenes, such as BCR-ABL, which provide survival signals, were argued to be sufficient to support the oncogenic state; suppression-induced apoptosis and tumour regression in vivo. The concept of tumour maintenance by a single oncogene, inhibiting apoptosis, is of interest.


Subject(s)
Apoptosis/physiology , Neoplasms/etiology , Animals , Cell Survival/physiology , DNA Damage , Fusion Proteins, bcr-abl/physiology , Gene Expression , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplasms/genetics , Neoplasms/pathology , Oncogenes , Proto-Oncogene Proteins c-abl/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Proto-Oncogene Proteins c-myc/physiology , Signal Transduction
12.
Oncotarget ; 9(28): 20075-20088, 2018 Apr 13.
Article in English | MEDLINE | ID: mdl-29732004

ABSTRACT

Escape from apoptosis is one of the major hallmarks of cancer cells. The B-cell Lymphoma 2 (BCL-2) gene family encodes pro-apoptotic and anti-apoptotic proteins that are key regulators of the apoptotic process. Overexpression of the pro-survival member BCL-2 is a well-established mechanism contributing to oncogenesis and chemoresistance in several cancers, including lymphoma and leukemia. Thus, BCL-2 has become an attractive target for therapeutic strategy in cancer, as demonstrated by the recent approval of ABT-199 (Venclexta™) in relapsed or refractory Chronic Lymphocytic Leukemia with 17p deletion. Here, we describe a novel orally bioavailable BCL-2 selective and potent inhibitor called S55746 (also known as BCL201). S55746 occupies the hydrophobic groove of BCL-2. Its selectivity profile demonstrates no significant binding to MCL-1, BFL-1 (BCL2A1/A1) and poor affinity for BCL-XL. Accordingly, S55746 has no cytotoxic activity on BCL-XL-dependent cells, such as platelets. In a panel of hematological cell lines, S55746 induces hallmarks of apoptosis including externalization of phosphatidylserine, caspase-3 activation and PARP cleavage. Ex vivo, S55746 induces apoptosis in the low nanomolar range in primary Chronic Lymphocytic Leukemia and Mantle Cell Lymphoma patient samples. Finally, S55746 administered by oral route daily in mice demonstrated robust anti-tumor efficacy in two hematological xenograft models with no weight lost and no change in behavior. Taken together, these data demonstrate that S55746 is a novel, well-tolerated BH3-mimetic targeting selectively and potently the BCL-2 protein.

13.
Mol Cancer Ther ; 4(1): 71-80, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15657355

ABSTRACT

The majority of DNA-binding small molecules known thus far stabilize duplex DNA against heat denaturation. A high, drug-induced increase in the melting temperature (Tm) of DNA is generally viewed as a good criterion to select DNA ligands and is a common feature of several anticancer drugs such as intercalators (e.g., anthracyclines) and alkylators (e.g., ecteinascidin 743). The reverse situation (destabilization of DNA to facilitate its denaturation) may be an attractive option for the identification of therapeutic agents acting on the DNA structure. We have identified the tumor-active benzoacronycine derivative S23906-1 [(+/-)-cis-1,2-diacetoxy-6-methoxy-3,3,14-trimethyl-1,2,3,14-tetrahydro-7H-benzo[b]pyrano[3,2]acridin-7-one] as a potent DNA alkylating agent endowed with a helicase-like activity. Using complementary molecular approaches, we show that covalent binding to DNA of the diacetate compound S23906-1 and its monoacetate analogue S28687-1 induces a marked destabilization of the double helix with the formation of alkylated ssDNA. The DNA-bonding properties and effects on DNA structure of a series of benzoacronycine derivatives, including the dicarbamate analogue S29385-1, were studied using complementary biochemical (electromobility shift assay, nuclease S1 mapping) and spectroscopic (fluorescence and Tm measurements) approaches. Alkylation of guanines in DNA by S28687-1 leads to a local denaturation of DNA, which becomes susceptible to cleavage by nuclease S1 and significantly decreases the Tm of DNA. The drug also directly alkylates single-strand DNA, but mass spectrometry experiments indicate that guanines in duplexes are largely preferred over single-stranded structures. This molecular study expands the repertoire of DNA-binding mechanisms and provides a new dimension for DNA recognition by small molecules.


Subject(s)
Acronine/analogs & derivatives , Acronine/toxicity , Antineoplastic Agents/toxicity , DNA, Single-Stranded/drug effects , DNA/chemistry , DNA/drug effects , Nucleic Acid Conformation/drug effects , Alkylating Agents/toxicity , Anthracyclines/toxicity , DNA, Single-Stranded/chemistry , Intercalating Agents/toxicity , Kinetics , Nucleic Acid Denaturation , Oligodeoxyribonucleotides/chemistry
14.
J Neurosci ; 23(23): 8423-31, 2003 Sep 10.
Article in English | MEDLINE | ID: mdl-12968005

ABSTRACT

BCL-2 family proteins are known to regulate cell death during development by influencing the permeability of mitochondrial membranes. The anti-apoptotic BCL-2 family protein BCL-xL is highly expressed in the adult brain and localizes to mitochondria in the presynaptic terminal of the adult squid stellate ganglion. Application of recombinant BCL-xL through a patch pipette to mitochondria inside the giant presynaptic terminal triggered multiconductance channel activity in mitochondrial membranes. Furthermore, injection of full-length BCL-xL protein into the presynaptic terminal enhanced postsynaptic responses and enhanced the rate of recovery from synaptic depression, whereas a recombinant pro-apoptotic cleavage product of BCL-xL attenuated postsynaptic responses. The effect of BCL-xL on synaptic responses persisted in the presence of a blocker of mitochondrial calcium uptake and was mimicked by injection of ATP into the terminal. These studies indicate that the permeability of outer mitochondrial membranes influences synaptic transmission, and they raise the possibility that modulation of mitochondrial conductance by BCL-2 family proteins affects synaptic stability.


Subject(s)
Neurons/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Synaptic Transmission/physiology , Adenosine Triphosphate/pharmacology , Animals , Coloring Agents/pharmacology , Decapodiformes , Electric Stimulation/methods , In Vitro Techniques , Ion Channels/metabolism , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Proto-Oncogene Proteins c-bcl-2/pharmacology , Recovery of Function/drug effects , Recovery of Function/physiology , Ruthenium Red/pharmacology , Synaptic Transmission/drug effects , bcl-X Protein
15.
Sci Rep ; 5: 17187, 2015 Dec 09.
Article in English | MEDLINE | ID: mdl-26647838

ABSTRACT

Precision-cut slices of in vivo tumours permit interrogation in vitro of heterogeneous cells from solid tumours together with their native microenvironment. They offer a low throughput but high content in vitro experimental platform. Using mouse models as surrogates for three common human solid tumours, we describe a standardised workflow for systematic comparison of tumour slice cultivation methods and a tissue microarray-based method to archive them. Cultivated slices were compared to their in vivo source tissue using immunohistochemical and transcriptional biomarkers, particularly of cellular stress. Mechanical slicing induced minimal stress. Cultivation of tumour slices required organotypic support materials and atmospheric oxygen for maintenance of integrity and was associated with significant temporal and loco-regional changes in protein expression, for example HIF-1α. We recommend adherence to the robust workflow described, with recognition of temporal-spatial changes in protein expression before interrogation of tumour slices by pharmacological or other means.


Subject(s)
Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Animals , Biomarkers , Cell Line, Tumor , Gene Expression , Heterografts , Humans , Immunohistochemistry/methods , Mice , Oxygen/metabolism , Principal Component Analysis , Real-Time Polymerase Chain Reaction , Signal Transduction , Stress, Physiological , Tissue Array Analysis , Tissue Culture Techniques
16.
J Med Chem ; 46(14): 3072-82, 2003 Jul 03.
Article in English | MEDLINE | ID: mdl-12825945

ABSTRACT

The cytotoxic and antitumor activities of cis-1,2-diacyloxy-6-methoxy-3,3,14-trimethyl-1,2,3,14-tetrahydro-7H-benzo[b]pyrano[3,2-h]acridin-7-one derivatives 3, 6-9 were strongly correlated with their ability to give covalent adducts with purified, as well as genomic, DNA. Such adducts involve reaction between the exocyclic N-2 amino group of guanines exposed in the minor groove of double helical DNA and the leaving ester group at the benzylic position 1 of the drug. A transesterification process of the ester group from position 2 to position 1 in aqueous medium accounted for the intense activity of the cis-1-hydroxy-2-acyloxy-6-methoxy-3,3,14-trimethyl-1,2,3,14-tetrahydro-7H-benzo[b]pyrano[3,2-h]acridin-7-one derivatives 10-13. Compounds without acyloxy or hydroxy group at position 1, such as 15, 17, 18, and 22, were inert with respect to DNA and almost devoid of significant cytotoxic activity. Condensation of 5-amino-2,2-dimethyl-2H-chromene (26) with 3-bromo-2-naphthoic acid (27), followed by cyclization, gave access to 6-demethoxy analogues. Diacetate 32 and cyclic carbonate 33, both belonging to the latter series, were less reactive toward DNA and less cytotoxic than their 6-methoxy counterparts 3 and 34. DNA alkylation appears thus to play an important role in the antitumor properties of benzo[b]pyrano[3,2-h]acridin-7-one derivatives.


Subject(s)
Acridines/chemistry , Acronine/analogs & derivatives , Acronine/chemistry , Antineoplastic Agents/chemistry , Benzopyrans/chemistry , Acridines/pharmacology , Acronine/pharmacology , Alkylation , Animals , Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Cell Division/drug effects , DNA/chemistry , DNA/metabolism , DNA Adducts/chemistry , DNA Adducts/metabolism , Drug Screening Assays, Antitumor , Esters , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasm Transplantation , Structure-Activity Relationship , Tumor Cells, Cultured
17.
Biochem Pharmacol ; 63(8): 1443-52, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11996885

ABSTRACT

The benzoacronycine derivative S23906-1 is a highly potent antitumor agent with a broad spectrum of activity against different human solid tumor xenografts. The marked cytotoxic potential of this drug may be the result of its interaction with DNA but the precise mechanism of action remains unclear at present. We have investigated the induction of apoptosis in human promyelocytic leukemia HL-60 and murine melanoma B16 cells treated with S23906-1. With both cell lines, the drug induces cell cycle perturbations (G2/M arrest) and triggers apoptosis as revealed by the externalization of Annexin V-targeted PS residues at the periphery of the cells. But the biochemical pathways leading to apoptosis are different for the two cancer cell lines. In HL-60 cells, the drug induces significant variations of the Delta Psi(mt), measured by flow cytometry using the fluorochromes JC-1 and cm-X-ros. Activation of caspase-3 and chromatin condensation in HL-60 cells exposed to submicromolar concentrations of S23906-1 for 24hr were also clearly seen by flow cytometry and confocal microscopy experiments. In contrast, the extent of apoptosis induced by S23906-1 was found to be much more limited in B16 cells. No significant variations of Delta Psi(mt) and no cleavage of the fluorescent caspase-3 substrate GDEVDGI (PhiPhiLux-G(1)D(2) probe) could be detected by cytometry in B16 cells exposed to S23906-1. In addition, we characterized the mitochondrial production of reactive oxygen species (ROS) using the probe dihydroethidine (HE) and the variations of the mitochondrial mass using the cardiolipin-interacting probe nonyl acridine orange (NAO). S23906-1 stimulates the production of ROS in both cell lines but the number of mitochondria seems to increase only in drug-treated B16 cells. Collectively these findings identify S23906-1 as a potent inducer of cell apoptosis in the leukemia cells and to a lower extent in the melanoma cells. The results help to understand the downstream cytotoxic actions of this new anticancer agent which is currently undergoing preclinical development.


Subject(s)
Acronine/analogs & derivatives , Acronine/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis , Leukemia/pathology , Melanoma, Experimental/pathology , Acronine/chemistry , Caspases/metabolism , Cell Cycle/drug effects , Cell Membrane/drug effects , Enzyme Activation/drug effects , HL-60 Cells , Humans , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/physiology , Reactive Oxygen Species/metabolism
18.
Biotechnol J ; 9(9): 1115-28, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25174503

ABSTRACT

Cancers are complex and heterogeneous pathological "organs" in a dynamic interplay with their host. Models of human cancer in vitro, used in cancer biology and drug discovery, are generally highly reductionist. These cancer models do not incorporate complexity or heterogeneity. This raises the question as to whether the cancer models' biochemical circuitry (not their genome) represents, with sufficient fidelity, a tumor in situ. Around 95% of new anticancer drugs eventually fail in clinical trial, despite robust indications of activity in existing in vitro pre-clinical models. Innovative models are required that better capture tumor biology. An important feature of all tissues, and tumors, is that cells grow in three dimensions. Advances in generating and characterizing simple and complex (with added stromal components) three-dimensional in vitro models (3D models) are reviewed in this article. The application of stirred bioreactors to permit both scale-up/scale-down of these cancer models and, importantly, methods to permit controlled changes in environment (pH, nutrients, and oxygen) are also described. The challenges of generating thin tumor slices, their utility, and potential advantages and disadvantages are discussed. These in vitro/ex vivo models represent a distinct move to capture the realities of tumor biology in situ, but significant characterization work still remains to be done in order to show that their biochemical circuitry accurately reflects that of a tumor.


Subject(s)
Cell Culture Techniques/methods , In Vitro Techniques/methods , Neoplasms/pathology , Animals , Bioreactors , Cell Biology , Humans , Models, Biological
19.
Mol Cancer Ther ; 12(9): 1749-62, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23804704

ABSTRACT

Aberrant activity of the receptor tyrosine kinases MET, AXL, and FGFR1/2/3 has been associated with tumor progression in a wide variety of human malignancies, notably in instances of primary or acquired resistance to existing or emerging anticancer therapies. This study describes the preclinical characterization of S49076, a novel, potent inhibitor of MET, AXL/MER, and FGFR1/2/3. S49076 potently blocked cellular phosphorylation of MET, AXL, and FGFRs and inhibited downstream signaling in vitro and in vivo. In cell models, S49076 inhibited the proliferation of MET- and FGFR2-dependent gastric cancer cells, blocked MET-driven migration of lung carcinoma cells, and inhibited colony formation of hepatocarcinoma cells expressing FGFR1/2 and AXL. In tumor xenograft models, a good pharmacokinetic/pharmacodynamic relationship for MET and FGFR2 inhibition following oral administration of S49076 was established and correlated well with impact on tumor growth. MET, AXL, and the FGFRs have all been implicated in resistance to VEGF/VEGFR inhibitors such as bevacizumab. Accordingly, combination of S49076 with bevacizumab in colon carcinoma xenograft models led to near total inhibition of tumor growth. Moreover, S49076 alone caused tumor growth arrest in bevacizumab-resistant tumors. On the basis of these preclinical studies showing a favorable and novel pharmacologic profile of S49076, a phase I study is currently underway in patients with advanced solid tumors. Mol Cancer Ther; 12(9); 1749-62. ©2013 AACR.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Indoles/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Thiazolidinediones/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Bevacizumab , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Indoles/chemistry , Mice , Mice, Inbred BALB C , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Thiazolidinediones/chemistry , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
20.
Nat Neurosci ; 15(4): 574-80, 2012 Feb 26.
Article in English | MEDLINE | ID: mdl-22366758

ABSTRACT

Transient global ischemia in rats induces delayed death of hippocampal CA1 neurons. Early events include caspase activation, cleavage of anti-death Bcl-2 family proteins and large mitochondrial channel activity. However, whether these events have a causal role in ischemia-induced neuronal death is unclear. We found that the Bcl-2 and Bcl-x(L) inhibitor ABT-737, which enhances death of tumor cells, protected rats against neuronal death in a clinically relevant model of brain ischemia. Bcl-x(L) is prominently expressed in adult neurons and can be cleaved by caspases to generate a pro-death fragment, ΔN-Bcl-x(L). We found that ABT-737 administered before or after ischemia inhibited ΔN-Bcl-x(L)-induced mitochondrial channel activity and neuronal death. To establish a causal role for ΔN-Bcl-x(L), we generated knock-in mice expressing a caspase-resistant form of Bcl-x(L). The knock-in mice exhibited markedly reduced mitochondrial channel activity and reduced vulnerability to ischemia-induced neuronal death. These findings suggest that truncated Bcl-x(L) could be a potentially important therapeutic target in ischemic brain injury.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/pathology , Neurons/metabolism , Neurons/pathology , bcl-X Protein/physiology , Animals , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Brain Ischemia/prevention & control , Cell Death/drug effects , Cell Death/genetics , Cells, Cultured , Female , Gene Knock-In Techniques , Male , Mice , Mice, Knockout , Neurons/drug effects , Nitrophenols/pharmacology , Nitrophenols/therapeutic use , Organ Culture Techniques , Piperazines/pharmacology , Piperazines/therapeutic use , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , bcl-X Protein/biosynthesis , bcl-X Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL