Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
2.
Cancer Metab ; 8: 13, 2020.
Article in English | MEDLINE | ID: mdl-32647572

ABSTRACT

BACKGROUND: Humans produce heat through non-shivering thermogenesis, a metabolic process that occurs in inducible beige adipocytes expressing uncoupling protein 1 (UCP1). UCP1 dissipates the proton gradient of the mitochondrial inner membrane and converts that energy into heat. It is unclear whether cancer cells can exhibit autonomous thermogenesis. Previously, we found that the knockdown of hypoxia-inducible fatty acid binding protein 7 (FABP7) increased reactive oxygen species (ROS) in breast cancer cells. ROS are known to induce beige adipocyte differentiation. METHODS: We investigated the association of tumor hypoxia, FABP7, and UCP1 across breast cancer patients using METABRIC and TCGA data sets. Furthermore, using a breast cancer cell line, HCC1806, we tested the effect of FABP7 knockdown on cellular physiology including thermogenesis. RESULTS: We found a strong mutual exclusivity of FABP7 and UCP1 expression both in METABRIC and in TCGA, indicating major metabolic phenotypic differences. FABP7 was preferentially distributed in poorly differentiated-, estrogen receptor (ER) negative tumors. In contrast, UCP1 was highly expressed in normal ducts and well-differentiated-, ER positive-, less hypoxic tumors. In the cell line-based experiments, UCP1 and its transcriptional regulators were upregulated upon FABP7 knockdown. UCP1 was induced in about 20% of cancer cells, and the effect was increased further in hypoxia. UCP1 depolarized mitochondrial membranes at the site of expression. UCP1 induction was associated with the increase in proton leak, glycolysis, and maximal respiration, mimicking the typical energy profile of beige adipocytes. Most importantly, UCP1 induction elevated cancer cell temperature associated with increased vulnerability to hypoxia and γ-irradiation. CONCLUSIONS: We demonstrated that breast cancer cells can undergo thermogenesis through UCP1 induction. Disrupting FABP7-mediated fatty acid metabolism can unlock UCP1-mediated thermogenesis, potentially making it possible to develop therapies to target thermogenesis. Further study would be warranted to investigate the effect of rise in temperature of cancer cells on patients' outcomes and the relationship to other metabolic pathways.

3.
Br J Cancer ; 122(2): 258-265, 2020 01.
Article in English | MEDLINE | ID: mdl-31819193

ABSTRACT

BACKGROUND: Epidemiological studies suggest that metformin may reduce the incidence of cancer in patients with diabetes and multiple late phase clinical trials assessing the potential of repurposing this drug are underway. Transcriptomic profiling of tumour samples is an excellent tool to understand drug bioactivity, identify candidate biomarkers and assess for mechanisms of resistance to therapy. METHODS: Thirty-six patients with untreated primary breast cancer were recruited to a window study and transcriptomic profiling of tumour samples carried out before and after metformin treatment. RESULTS: Multiple genes that regulate fatty acid oxidation were upregulated at the transcriptomic level and there was a differential change in expression between two previously identified cohorts of patients with distinct metabolic responses. Increase in expression of a mitochondrial fatty oxidation gene composite signature correlated with change in a proliferation gene signature. In vitro assays showed that, in contrast to previous studies in models of normal cells, metformin reduces fatty acid oxidation with a subsequent accumulation of intracellular triglyceride, independent of AMPK activation. CONCLUSIONS: We propose that metformin at clinical doses targets fatty acid oxidation in cancer cells with implications for patient selection and drug combinations. CLINICAL TRIAL REGISTRATION: NCT01266486.


Subject(s)
Breast Neoplasms/drug therapy , Fatty Acids/metabolism , Metformin/pharmacology , Protein Kinases/genetics , AMP-Activated Protein Kinase Kinases , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Diabetes Mellitus/drug therapy , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Lipid Metabolism/drug effects , Lipid Peroxidation/drug effects , Mice , Mitochondria/drug effects , Oxidation-Reduction/drug effects , Transcriptome/drug effects
4.
Front Immunol ; 10: 1495, 2019.
Article in English | MEDLINE | ID: mdl-31379806

ABSTRACT

The interplay between NOD2 and TLR2 following recognition of components of the bacterial cell wall peptidoglycan is well-established, however their role in redirecting metabolic pathways in myeloid cells to degrade pathogens and mount antigen presentation remains unclear. We show NOD2 and TLR2 mediate phosphorylation of the deubiquitinase ataxin-3 via RIPK2 and TBK1. In myeloid cells ataxin-3 associates with the mitochondrial cristae protein MIC60, and is required for oxidative phosphorylation. Depletion of ataxin-3 leads to impaired induction of mitochondrial reactive oxygen species (mROS) and defective bacterial killing. A mass spectrometry analysis of NOD2/TLR2 triggered ataxin-3 deubiquitination targets revealed immunometabolic regulators, including HIF-1α and LAMTOR1 that may contribute to these effects. Thus, we define how ataxin-3 plays an essential role in NOD2 and TLR2 sensing and effector functions in myeloid cells.


Subject(s)
Ataxin-3/immunology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Nod2 Signaling Adaptor Protein/immunology , Receptor-Interacting Protein Serine-Threonine Kinase 2/immunology , Toll-Like Receptor 2/immunology , Ataxin-3/metabolism , Cell Respiration , HEK293 Cells , Humans , Immunity, Innate , Mitochondria/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Signal Transduction , THP-1 Cells , Toll-Like Receptor 2/metabolism
5.
Proc Natl Acad Sci U S A ; 116(25): 12452-12461, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31152137

ABSTRACT

Tumor hypoxia is associated with poor patient outcomes in estrogen receptor-α-positive (ERα+) breast cancer. Hypoxia is known to affect tumor growth by reprogramming metabolism and regulating amino acid (AA) uptake. Here, we show that the glutamine transporter, SNAT2, is the AA transporter most frequently induced by hypoxia in breast cancer, and is regulated by hypoxia both in vitro and in vivo in xenografts. SNAT2 induction in MCF7 cells was also regulated by ERα, but it became predominantly a hypoxia-inducible factor 1α (HIF-1α)-dependent gene under hypoxia. Relevant to this, binding sites for both HIF-1α and ERα overlap in SNAT2's cis-regulatory elements. In addition, the down-regulation of SNAT2 by the ER antagonist fulvestrant was reverted in hypoxia. Overexpression of SNAT2 in vitro to recapitulate the levels induced by hypoxia caused enhanced growth, particularly after ERα inhibition, in hypoxia, or when glutamine levels were low. SNAT2 up-regulation in vivo caused complete resistance to antiestrogen and, partially, anti-VEGF therapies. Finally, high SNAT2 expression levels correlated with hypoxia profiles and worse outcome in patients given antiestrogen therapies. Our findings show a switch in the regulation of SNAT2 between ERα and HIF-1α, leading to endocrine resistance in hypoxia. Development of drugs targeting SNAT2 may be of value for a subset of hormone-resistant breast cancer.


Subject(s)
Amino Acid Transport System A/metabolism , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/pathology , Cell Hypoxia , Drug Resistance, Neoplasm , Estrogen Receptor Modulators/therapeutic use , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Estrogen Receptor alpha/metabolism , Female , Heterografts , Humans , Mice , Tumor Microenvironment
6.
Cell Metab ; 28(5): 679-688.e4, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30244975

ABSTRACT

Late-phase clinical trials investigating metformin as a cancer therapy are underway. However, there remains controversy as to the mode of action of metformin in tumors at clinical doses. We conducted a clinical study integrating measurement of markers of systemic metabolism, dynamic FDG-PET-CT, transcriptomics, and metabolomics at paired time points to profile the bioactivity of metformin in primary breast cancer. We show metformin reduces the levels of mitochondrial metabolites, activates multiple mitochondrial metabolic pathways, and increases 18-FDG flux in tumors. Two tumor groups are identified with distinct metabolic responses, an OXPHOS transcriptional response (OTR) group for which there is an increase in OXPHOS gene transcription and an FDG response group with increased 18-FDG uptake. Increase in proliferation, as measured by a validated proliferation signature, suggested that patients in the OTR group were resistant to metformin treatment. We conclude that mitochondrial response to metformin in primary breast cancer may define anti-tumor effect.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Hypoglycemic Agents/pharmacology , Metabolic Networks and Pathways/drug effects , Metformin/pharmacology , Adult , Aged , Antineoplastic Agents/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Glucose/analogs & derivatives , Glucose/metabolism , Humans , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Middle Aged , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Positron Emission Tomography Computed Tomography , Transcriptome/drug effects
7.
Ann Neurol ; 83(5): 915-925, 2018 05.
Article in English | MEDLINE | ID: mdl-29604226

ABSTRACT

OBJECTIVE: Defective mitochondrial function attributed to optic atrophy 1 (OPA1) mutations causes primarily optic atrophy and, less commonly, neurodegenerative syndromes. The pathomechanism by which OPA1 mutations trigger diffuse loss of neurons in some, but not all, patients is unknown. Here, we used a tractable induced pluripotent stem cell (iPSC)-based model to capture the biology of OPA1 haploinsufficiency in cases presenting with classic eye disease versus syndromic parkinsonism. METHODS: iPSCs were generated from 2 patients with OPA1 haploinsufficiency and 2 controls and differentiated into dopaminergic neurons. Metabolic profile was determined by extracellular flux analysis, respiratory complex levels using immunoblotting, and complex I activity by a colorimetric assay. Mitochondria were examined by transmission electron microscopy. Mitochondrial DNA copy number and deletions were assayed using long-range PCR. Mitochondrial membrane potential was measured by tetramethylrhodamine methyl ester uptake, and mitochondrial fragmentation was assessed by confocal microscopy. Exome sequencing was used to screen for pathogenic variants. RESULTS: OPA1 haploinsufficient iPSCs differentiated into dopaminergic neurons and exhibited marked reduction in OPA1 protein levels. Loss of OPA1 caused a late defect in oxidative phosphorylation, reduced complex I levels, and activity without a significant change in the ultrastructure of mitochondria. Loss of neurons in culture recapitulated dopaminergic degeneration in syndromic disease and correlated with mitochondrial fragmentation. INTERPRETATION: OPA1 levels maintain oxidative phosphorylation in iPSC-derived neurons, at least in part, by regulating the stability of complex I. Severity of OPA1 disease associates primarily with the extent of OPA1-mediated fusion, suggesting that activation of this mechanism or identification of its genetic modifiers may have therapeutic or prognostic value. Ann Neurol 2018;83:915-925.


Subject(s)
GTP Phosphohydrolases/metabolism , Induced Pluripotent Stem Cells/metabolism , Mitochondria/metabolism , Parkinsonian Disorders/metabolism , DNA, Mitochondrial/genetics , Humans , Membrane Potential, Mitochondrial/physiology , Optic Atrophy/genetics , Oxidative Phosphorylation , Parkinsonian Disorders/genetics
8.
Immunotherapy ; 9(1): 25-32, 2017 01.
Article in English | MEDLINE | ID: mdl-28000528

ABSTRACT

AIM: IL-2 is one of the first immunomodulating cytokines to be tested in the treatment of cancer patients. The effects of this agent in the treatment of solid tumors other than renal cancer and melanoma are poorly understood. MATERIALS & METHODS: We have carried out a meta-analysis of randomized studies. We fixed the response rate as the primary outcome. RESULTS: The pooled risk ratio for an objective response with IL-2 plus chemotherapy versus chemotherapy alone was 1.43 (95% CI: 1.12-1.81; p = 0.004), in favor of colorectal cancer. CONCLUSION: Further investigation in the treatment of patients with colorectal cancer or other solid malignancies with IL-2 is required, alone or in combination with chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/therapy , Immunotherapy/methods , Interleukin-2/therapeutic use , Kidney Neoplasms/therapy , Melanoma/therapy , Animals , Colorectal Neoplasms/immunology , Drug Therapy , Humans , Kidney Neoplasms/immunology , Melanoma/immunology , Randomized Controlled Trials as Topic , Risk
9.
Genome Biol ; 17(1): 140, 2016 06 29.
Article in English | MEDLINE | ID: mdl-27358048

ABSTRACT

BACKGROUND: Altered metabolism is a hallmark of cancer. However, the role of genomic changes in metabolic genes driving the tumour metabolic shift remains to be elucidated. Here, we have investigated the genomic and transcriptomic changes underlying this shift across ten different cancer types. RESULTS: A systematic pan-cancer analysis of 6538 tumour/normal samples covering ten major cancer types identified a core metabolic signature of 44 genes that exhibit high frequency somatic copy number gains/amplifications (>20 % cases) associated with increased mRNA expression (ρ > 0.3, q < 10(-3)). Prognostic classifiers using these genes were confirmed in independent datasets for breast and kidney cancers. Interestingly, this signature is strongly associated with hypoxia, with nine out of ten cancer types showing increased expression and five out of ten cancer types showing increased gain/amplification of these genes in hypoxic tumours (P ≤ 0.01). Further validation in breast and colorectal cancer cell lines highlighted squalene epoxidase, an oxygen-requiring enzyme in cholesterol biosynthesis, as a driver of dysregulated metabolism and a key player in maintaining cell survival under hypoxia. CONCLUSIONS: This study reveals somatic genomic alterations underlying a pan-cancer metabolic shift and suggests genomic adaptation of these genes as a survival mechanism in hypoxic tumours.


Subject(s)
Energy Metabolism/genetics , Genetic Variation , Neoplasms/genetics , Neoplasms/metabolism , Tumor Hypoxia/genetics , Animals , Biomarkers, Tumor , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cluster Analysis , DNA Copy Number Variations , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genomic Instability , Genomics/methods , Heterografts , Humans , Mice , Mutation , Neoplasms/mortality , Neoplasms/pathology , Prognosis , Transcriptome
10.
Cancer Res ; 76(13): 3744-55, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27197160

ABSTRACT

Tumor hypoxia is associated clinically with therapeutic resistance and poor patient outcomes. One feature of tumor hypoxia is activated expression of carbonic anhydrase IX (CA9), a regulator of pH and tumor growth. In this study, we investigated the hypothesis that impeding the reuptake of bicarbonate produced extracellularly by CA9 could exacerbate the intracellular acidity produced by hypoxic conditions, perhaps compromising cell growth and viability as a result. In 8 of 10 cancer cell lines, we found that hypoxia induced the expression of at least one bicarbonate transporter. The most robust and frequent inductions were of the sodium-driven bicarbonate transporters SLC4A4 and SLC4A9, which rely upon both HIF1α and HIF2α activity for their expression. In cancer cell spheroids, SLC4A4 or SLC4A9 disruption by either genetic or pharmaceutical approaches acidified intracellular pH and reduced cell growth. Furthermore, treatment of spheroids with S0859, a small-molecule inhibitor of sodium-driven bicarbonate transporters, increased apoptosis in the cell lines tested. Finally, RNAi-mediated attenuation of SLC4A9 increased apoptosis in MDA-MB-231 breast cancer spheroids and dramatically reduced growth of MDA-MB-231 breast tumors or U87 gliomas in murine xenografts. Our findings suggest that disrupting pH homeostasis by blocking bicarbonate import might broadly relieve the common resistance of hypoxic tumors to anticancer therapy. Cancer Res; 76(13); 3744-55. ©2016 AACR.


Subject(s)
Bicarbonates/metabolism , Hypoxia/physiopathology , Neoplasms/metabolism , Neoplasms/prevention & control , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Apoptosis , Blotting, Western , Carbonic Anhydrase IX/genetics , Carbonic Anhydrase IX/metabolism , Cell Proliferation , Female , Humans , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasms/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sodium-Bicarbonate Symporters/genetics , Sodium-Bicarbonate Symporters/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
PLoS One ; 11(4): e0153692, 2016.
Article in English | MEDLINE | ID: mdl-27088720

ABSTRACT

The oxygen status of a tumor has significant clinical implications for treatment prognosis, with well-oxygenated subvolumes responding markedly better to radiotherapy than poorly supplied regions. Oxygen is essential for tumor growth, yet estimation of local oxygen distribution can be difficult to ascertain in situ, due to chaotic patterns of vasculature. It is possible to avoid this confounding influence by using avascular tumor models, such as tumor spheroids, a much better approximation of realistic tumor dynamics than monolayers, where oxygen supply can be described by diffusion alone. Similar to in situ tumours, spheroids exhibit an approximately sigmoidal growth curve, often approximated and fitted by logistic and Gompertzian sigmoid functions. These describe the basic rate of growth well, but do not offer an explicitly mechanistic explanation. This work examines the oxygen dynamics of spheroids and demonstrates that this growth can be derived mechanistically with cellular doubling time and oxygen consumption rate (OCR) being key parameters. The model is fitted to growth curves for a range of cell lines and derived values of OCR are validated using clinical measurement. Finally, we illustrate how changes in OCR due to gemcitabine treatment can be directly inferred using this model.


Subject(s)
Deoxycytidine/analogs & derivatives , Models, Theoretical , Neoplasms/drug therapy , Neoplasms/pathology , Oxygen Consumption/drug effects , Oxygen/metabolism , Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/pharmacology , Humans , Tumor Cells, Cultured , Gemcitabine
12.
Oncotarget ; 6(23): 19413-27, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26305601

ABSTRACT

Carbonic anhydrase IX (CAIX) is strongly induced by hypoxia and its overexpression is associated with poor therapeutic outcome in cancer. Here, we report that hypoxia promotes tumour heterogeneity through the epigenetic regulation of CAIX. Based on hypoxic CAIX expression we identify and characterize two distinct populations of tumour cells, one that has inducible expression of CAIX and one that does not. The CAIX+ve population is enriched with cells expressing cancer stem cell markers and which have high self-renewal capacity. We show that differential CAIX expression is due to differences in chromatin structure. To further investigate the relationship between chromatin organization and hypoxic induction of CAIX expression we investigated the effect of JQ1 an inhibitor of BET bromodomain proteins and A366 a selective inhibitor of the H3K9 methyltransferase G9a/GLP. We identified that these drugs were able to modulate hypoxic CAIX expression induction. This further highlights the role of epigenetic modification in adaption to hypoxia and also in regulation of heterogeneity of cells within tumours. Interestingly, we identified that the two subpopulations show a differential sensitivity to HDAC inhibitors, NaBu or SAHA, with the CAIX positive showing greater sensitivity to treatment. We propose that drugs modulating chromatin regulation of expression may be used to reduce heterogeneity induced by hypoxia and could in combination have significant clinical consequences.


Subject(s)
Antigens, Neoplasm/biosynthesis , Carbonic Anhydrases/biosynthesis , Histone Deacetylase Inhibitors/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Animals , Antigens, Neoplasm/genetics , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Line, Tumor , Enzyme Induction , Female , HCT116 Cells , Heterografts , Humans , Isoenzymes/biosynthesis , MCF-7 Cells , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/pathology
13.
Cell Rep ; 9(1): 349-365, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25263561

ABSTRACT

An in vivo model of antiangiogenic therapy allowed us to identify genes upregulated by bevacizumab treatment, including Fatty Acid Binding Protein 3 (FABP3) and FABP7, both of which are involved in fatty acid uptake. In vitro, both were induced by hypoxia in a hypoxia-inducible factor-1α (HIF-1α)-dependent manner. There was a significant lipid droplet (LD) accumulation in hypoxia that was time and O2 concentration dependent. Knockdown of endogenous expression of FABP3, FABP7, or Adipophilin (an essential LD structural component) significantly impaired LD formation under hypoxia. We showed that LD accumulation is due to FABP3/7-dependent fatty acid uptake while de novo fatty acid synthesis is repressed in hypoxia. We also showed that ATP production occurs via ß-oxidation or glycogen degradation in a cell-type-dependent manner in hypoxia-reoxygenation. Finally, inhibition of lipid storage reduced protection against reactive oxygen species toxicity, decreased the survival of cells subjected to hypoxia-reoxygenation in vitro, and strongly impaired tumorigenesis in vivo.


Subject(s)
Fatty Acids/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lipid Metabolism , Oxygen/metabolism , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Glioblastoma , Humans , Oxidation-Reduction
14.
J Mol Med (Berl) ; 91(6): 749-58, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23361368

ABSTRACT

Inhibition of vascular endothelial growth factor increases response rates to chemotherapy and progression-free survival in glioblastoma. However, resistance invariably occurs, prompting the urgent need for identification of synergizing agents. One possible strategy is to understand tumor adaptation to microenvironmental changes induced by antiangiogenic drugs and test agents that exploit this process. We used an in vivo glioblastoma-derived xenograft model of tumor escape in presence of continuous treatment with bevacizumab. U87-MG or U118-MG cells were subcutaneously implanted into either BALB/c SCID or athymic nude mice. Bevacizumab was given by intraperitoneal injection every 3 days (2.5 mg/kg/dose) and/or dichloroacetate (DCA) was administered by oral gavage twice daily (50 mg/kg/dose) when tumor volumes reached 0.3 cm(3) and continued until tumors reached approximately 1.5-2.0 cm(3). Microarray analysis of resistant U87 tumors revealed coordinated changes at the level of metabolic genes, in particular, a widening gap between glycolysis and mitochondrial respiration. There was a highly significant difference between U87-MG-implanted athymic nude mice 1 week after drug treatment. By 2 weeks of treatment, bevacizumab and DCA together dramatically blocked tumor growth compared to either drug alone. Similar results were seen in athymic nude mice implanted with U118-MG cells. We demonstrate for the first time that reversal of the bevacizumab-induced shift in metabolism using DCA is detrimental to neoplastic growth in vivo. As DCA is viewed as a promising agent targeting tumor metabolism, our data establish the timely proof of concept that combining it with antiangiogenic therapy represents a potent antineoplastic strategy.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Dichloroacetic Acid/administration & dosage , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Animals , Bevacizumab , Cell Line, Tumor , Drug Therapy, Combination , Female , Humans , Hypoxia , Mice , Mice, SCID , Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Clin Cancer Res ; 18(11): 3100-11, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22498007

ABSTRACT

PURPOSE: Bevacizumab, an anti-VEGFA antibody, inhibits the developing vasculature of tumors, but resistance is common. Antiangiogenic therapy induces hypoxia and we observed increased expression of hypoxia-regulated genes, including carbonic anhydrase IX (CAIX), in response to bevacizumab treatment in xenografts. CAIX expression correlates with poor prognosis in most tumor types and with worse outcome in bevacizumab-treated patients with metastatic colorectal cancer, malignant astrocytoma, and recurrent malignant glioma. EXPERIMENTAL DESIGN: We knocked down CAIX expression by short hairpin RNA in a colon cancer (HT29) and a glioblastoma (U87) cell line which have high hypoxic induction of CAIX and overexpressed CAIX in HCT116 cells which has low CAIX. We investigated the effect on growth rate in three-dimensional (3D) culture and in vivo, and examined the effect of CAIX knockdown in combination with bevacizumab. RESULTS: CAIX expression was associated with increased growth rate in spheroids and in vivo. Surprisingly, CAIX expression was associated with increased necrosis and apoptosis in vivo and in vitro. We found that acidity inhibits CAIX activity over the pH range found in tumors (pK = 6.84), and this may be the mechanism whereby excess acid self-limits the build-up of extracellular acid. Expression of another hypoxia inducible CA isoform, CAXII, was upregulated in 3D but not two-dimensional culture in response to CAIX knockdown. CAIX knockdown enhanced the effect of bevacizumab treatment, reducing tumor growth rate in vivo. CONCLUSION: This work provides evidence that inhibition of the hypoxic adaptation to antiangiogenic therapy enhances bevacizumab treatment and highlights the value of developing small molecules or antibodies which inhibit CAIX for combination therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, Neoplasm/metabolism , Carbonic Anhydrases/metabolism , Cell Proliferation/drug effects , Animals , Antigens, Neoplasm/genetics , Bevacizumab , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Cell Line, Tumor , Female , Gene Knockdown Techniques , Glioblastoma/metabolism , HCT116 Cells , HT29 Cells , Humans , Hydrogen-Ion Concentration , Mice , Necrosis , Neoplasm Transplantation , Transfection , Transplantation, Heterologous
16.
Radiother Oncol ; 99(3): 424-31, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21676479

ABSTRACT

BACKGROUND AND PURPOSE: Carbonic anhydrase (CA) IX expression is increased upon hypoxia and has been proposed as a therapeutic target since it has been associated with poor prognosis, tumor progression and pH regulation. The aim of this study was to evaluate the antitumor activity of a high CAIX-affinity indanesulfonamide (11c) combined with irradiation, compared with the general CA inhibitor acetazolamide (AZA). MATERIAL AND METHODS: HT-29 carcinoma cells with or without (genetic knockdown, KD) CAIX expression were incubated with 11c/AZA under different oxygen levels and proliferation, apoptosis and radiosensitivity were evaluated. 11c/AZA was administered intravenously (1×/day; 5 days) to tumor-bearing mice and tumor irradiation (10 Gy) was performed at day 3 of the injection period. Tumor growth and potential treatment toxicity were monitored (3×/week). RESULTS: Treatment with 11c/AZA alone resulted in tumor regression, which was further increased in CAIX expressing cells by combining 11c with irradiation. AZA demonstrated also an additional effect in the KD tumors when combined with irradiation. CAIX inhibition in vitro significantly reduced proliferation and increased apoptosis upon hypoxia exposure without affecting intrinsic radiosensitivity. CONCLUSIONS: Specific inhibition of CAIX activity enhanced the effect of tumor irradiation and might, therefore, be an attractive strategy to improve overall cancer treatment.


Subject(s)
Acetazolamide/pharmacology , Carbonic Anhydrase Inhibitors/pharmacology , HT29 Cells/drug effects , HT29 Cells/radiation effects , Animals , Apoptosis , Cell Hypoxia , Cell Proliferation , Flow Cytometry , Humans , Immunoblotting , Lactic Acid/metabolism , Mice , Mice, Nude , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric
17.
Biochem Pharmacol ; 79(11): 1562-72, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20100468

ABSTRACT

BNIP3 is a hypoxia-inducible BH3-only member of the Bcl-2 family of proteins that regulate apoptosis and autophagy. However the role of BNIP3 in the hypoxia response has proved difficult to define and remains controversial. In this study we show that in cancer cells, knockdown or forced expression of BNIP3 fails to modulate cell survival under hypoxic or normoxic conditions. However, we demonstrate that BNIP3 is regulated post-translationally, existing as multiple monomeric and dimeric phosphorylated forms. Upon treatment with microtubule inhibitors, but not other classes of chemotherapeutics, BNIP3 becomes hyperphosphorylated. We demonstrate that the phosphorylation of BNIP3 occurs in synchrony with phosphorylation of its binding partners Bcl-2 and Bcl-xL. Microtubule inhibitor-induced phosphorylation of these proteins occurs independently of the AKT/mTor and JNK kinase pathways and requires Mps1 mitotic checkpoint kinase activity. Inhibition of mitotic arrest in the presence of paclitaxel blocks the phosphorylation of BNIP3, Bcl-2 and Bcl-xL, demonstrating that these proteins are phosphorylated by a mitochondrially active mitotic kinase. We show that phosphorylation increases the stability of BNIP3 and that BNIP3 predominantly interacts with the phosphorylated form of Bcl-2. This study provides new insight into the post-translational functional control of these Bcl-2 family members.


Subject(s)
Cell Cycle Proteins/metabolism , Microtubules/drug effects , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival , Gene Expression Regulation/drug effects , Humans , Hypoxia , MAP Kinase Kinase 4 , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondrial Proteins , Mitosis , Phosphorylation/drug effects , Protein-Tyrosine Kinases , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/metabolism
18.
J Biol Chem ; 283(29): 20473-83, 2008 Jul 18.
Article in English | MEDLINE | ID: mdl-18482982

ABSTRACT

CA9 is a membrane-tethered, carbonic anhydrase (CA) enzyme, expressed mainly at the external surface of cells, that catalyzes reversible CO(2) hydration. Expression is greatly enhanced in many tumors, particularly in aggressive carcinomas. The functional role of CA9 in tumors is not well established. Here we show that CA9, when expressed heterologously in cultured spheroids (0.5-mm diameter, ~25,000 cells) of RT112 cells (derived from bladder carcinoma), induces a near-uniform intracellular pH (pH(i)) throughout the structure. Dynamic pH(i) changes during displacements of superfusate CO(2) concentration are also spatially coincident (within 2 s). In contrast, spheroids of wild-type RT112 cells lacking CA9 exhibit an acidic core (~0.25 pH(i) reduction) and significant time delays (~9 s) for pH(i) changes in core versus peripheral regions. pH(i) non-uniformity also occurs in CA9-expressing spheroids after selective pharmacological inhibition of the enzyme. In isolated RT112 cells, pH(i) regulation is unaffected by CA9 expression. The influence of CA9 on pH(i) is thus only evident in multicellular tissue. Diffusion-reaction modeling indicates that CA9 coordinates pH(i) spatially by facilitating CO(2) diffusion in the unstirred extracellular space of the spheroid. We suggest that pH(i) coordination may favor survival and growth of a tumor. By disrupting spatial pH(i) control, inhibition of CA9 activity may offer a novel strategy for the clinical treatment of CA9-associated tumors.


Subject(s)
Antigens, Neoplasm/chemistry , Antigens, Neoplasm/metabolism , Carbonic Anhydrases/chemistry , Carbonic Anhydrases/metabolism , Neoplasms/enzymology , Antigens, Neoplasm/genetics , Apoptosis , Carbon Dioxide/metabolism , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Cell Line, Tumor , Cell Membrane/enzymology , Gene Expression Regulation, Enzymologic , Humans , Hydrogen-Ion Concentration , Models, Molecular , Neoplasms/genetics , Protein Structure, Tertiary , Protons
20.
Endocr Relat Cancer ; 13(3): 921-30, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16954440

ABSTRACT

The purpose of this study is to investigate the role of carbonic anhydrase IX (CAIX) expression in predicting the response to epirubicin and disease-free survival (DFS) in breast cancer patients enrolled in a single institution trial of primary anthracycline and tamoxifen therapy. CAIX expression was assessed in 183 patients with T2-4 N0-1 breast cancer enrolled in a randomized trial comparing four cycles of single agent epirubicin versus epirubicin+tamoxifen as primary systemic treatment. All patients received postoperatively four cycles of the four weekly i.v. cyclophosphamide, methotrexate, 5-fluorouracil regimen. Patients with estrogen receptor (ER)-positive primary tumors received 5 years of adjuvant tamoxifen. Pretreatment, p53 (P=0.007), c-erbB2 (P<0.01), and Ki67 (P=0.02) were directly associated with CAIX expression, while bcl2 (P<0.000) and ER (P=0.000) and progesterone receptor (PgR; P<0.01) were inversely correlated. In multivariate analysis, only high p53 and low bcl2 were independently associated with CAIX positivity. CAIX immunostaining was significantly associated with poor outcome for DFS (P<0.002) and overall survival (P=0.001). In multivariate analysis, a significant interaction was found between CAIX and markers of hormone sensitivity, bcl2 (P=0.01), ER (P=0.02), PgR (P=0.02), and lymph node involvement (P=0.04), in predicting DFS. Presently, there are few clinical markers of resistance to tamoxifen treatment in ER-positive tumors. CAIX expression in breast cancer patients shows a negative predictive role of treatment efficacy in ER-positive patients on the adjuvant tamoxifen after primary chemo-endocrine therapy. Studies investigating the effects of pH on tamoxifen uptake and the effects of therapy with CA inhibitors are planned.


Subject(s)
Antigens, Neoplasm/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Carbonic Anhydrases/genetics , Epirubicin/therapeutic use , Tamoxifen/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Antigens, Neoplasm/metabolism , Antineoplastic Agents, Hormonal/therapeutic use , Biopsy , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carbonic Anhydrase IX , Carbonic Anhydrases/metabolism , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Neoplasm Metastasis , Neoplasm Staging , Postmenopause , Premenopause , Receptors, Estrogen/metabolism , Survival Analysis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...