Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33947138

ABSTRACT

Ovarian clear cell carcinoma (OCCC) is a rare but chemorefractory tumor. About 50% of all OCCC patients have inactivating mutations of ARID1A, a member of the SWI/SNF chromatin-remodeling complex. Members of the SWI/SNF remodeling have emerged as regulators of the energetic metabolism of mammalian cells; however, the role of ARID1A as a modulator of the mitochondrial metabolism in OCCCs is yet to be defined. Here, we show that ARID1A loss results in increased mitochondrial metabolism and renders ARID1A-mutated cells increasingly and selectively dependent on it. The increase in mitochondrial activity following ARID1A loss is associated with increase in c-Myc expression and increased mitochondrial number and reduction of their size consistent with a higher mitochondrial cristae/outer membrane ratio. Significantly, preclinical testing of the complex I mitochondrial inhibitor IACS-010759 showed it extends overall survival in a preclinical model of ARID1A-mutated OCCC. These findings provide for the targeting mitochondrial activity in ARID1A-mutated OCCCs.


Subject(s)
Adenocarcinoma, Clear Cell/drug therapy , Antineoplastic Agents/therapeutic use , DNA-Binding Proteins/antagonists & inhibitors , Mitochondria/drug effects , Neoplasm Proteins/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Oxadiazoles/therapeutic use , Piperidines/therapeutic use , Transcription Factors/antagonists & inhibitors , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/pathology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Electron Transport Complex I/antagonists & inhibitors , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Oxadiazoles/pharmacology , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Piperidines/pharmacology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Random Allocation , Spheroids, Cellular , Transcription Factors/deficiency , Transcription Factors/genetics , Xenograft Model Antitumor Assays
2.
Invest New Drugs ; 38(5): 1448-1453, 2020 10.
Article in English | MEDLINE | ID: mdl-32125598

ABSTRACT

This phase 1 study sought to characterize the safety, tolerability, and pharmacokinetic behavior of VLX1570, a small molecule inhibitor of the deubiquitinases (DUBs) that remove sterically bulky ubiquitin chains from proteins during processing in the19S regulatory subunit of the proteasome, in patients with relapsed and refractory multiple myeloma (MM). Fourteen patients were treated with escalating doses of VLX1570 ranging from 0.05 to 1.2 mg/kg as a brief intravenous (IV) infusion on Days 1, 2, 8, 9, 15, and 16 of a 28-day cycle. Due to its poor aqueous solubility, VLX1570 was formulated in polyethylene glycol, polyoxyethylated castor oil, and polysorbate 80 and administered as a brief intravenous (IV) infusion via a central venous catheter. Anti-myeloma effects were noted at doses at or above 0.6 mg/kg, however, two patients treated at the 1.2 mg/kg dose level experienced severe, abrupt, and progressive respiratory insufficiency, which was associated with diffuse pulmonary infiltrates on imaging studies, similar to those rarely noted with bortezomib and other inhibitors of the 20S proteasome, culminating in death. Although the contribution of VLX1570's formulation to the pulmonary toxicity could not be ruled out, the severity and precipitous nature of the toxicity and the steep relationship between dose and toxicity, the study was discontinued. Despite the severe pulmonary toxicity noted with VLX1570, efforts directed at identifying DUB inhibitors with greater therapeutic indices appear warranted based on the unique mechanism of action, robustness of preclinical antitumor activity, and activity of the DUB inhibitors in MM resistant to PIs targeting the 20S proteasome subunit.


Subject(s)
Antineoplastic Agents/administration & dosage , Azepines/administration & dosage , Benzylidene Compounds/administration & dosage , Deubiquitinating Enzymes/antagonists & inhibitors , Multiple Myeloma/drug therapy , Respiratory Insufficiency/chemically induced , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Azepines/adverse effects , Azepines/pharmacokinetics , Benzylidene Compounds/adverse effects , Benzylidene Compounds/pharmacokinetics , Drug Resistance, Neoplasm , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Multiple Myeloma/metabolism , Multiple Myeloma/mortality , Recurrence , Respiratory Insufficiency/mortality
3.
Int J Mol Sci ; 21(13)2020 Jul 04.
Article in English | MEDLINE | ID: mdl-32635430

ABSTRACT

The proteasome is a validated target of cancer therapeutics. Inhibition of proteasome activity results in the activation of the unfolded protein response (UPR) characterized by phosphorylation of eukaryotic initiation factor 2α (eIF2α), global translational arrest, and increased expression of the proapoptotic CHOP (C/EBP homologous protein) protein. Defects in the UPR response has been reported to result in altered sensitivity of tumor cells to proteasome inhibitors. Here, we characterized the effects of the deubiquitinase (DUB) inhibitor VLX1570 on protein homeostasis, both at the level of the UPR and on protein translation, in acute lymphoblastic leukemia (ALL). Similar to the 20S inhibitor bortezomib, VLX1570 induced accumulation of polyubiquitinated proteins and increased expression of the chaperone Grp78/Bip in ALL cells. Both compounds induced cleavage of PARP (Poly (ADP-ribose) polymerase) in ALL cells, consistent with induction of apoptosis. However, and in contrast to bortezomib, VLX1570 treatment resulted in limited induction of the proapoptotic CHOP protein. Translational inhibition was observed by both bortezomib and VLX1570. We report that in distinction to bortezomib, suppression of translation by VXL1570 occurred at the level of elongation. Increased levels of Hsc70/Hsp70 proteins were observed on polysomes following exposure to VLX1570, possibly suggesting defects in nascent protein folding. Our findings demonstrate apoptosis induction in ALL cells that appears to be uncoupled from CHOP induction, and show that VLX1570 suppresses protein translation by a mechanism distinct from that of bortezomib.


Subject(s)
Azepines/pharmacology , Benzylidene Compounds/pharmacology , Deubiquitinating Enzymes/antagonists & inhibitors , Deubiquitinating Enzymes/metabolism , Endoplasmic Reticulum Stress/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Animals , Apoptosis/drug effects , Bortezomib/pharmacology , Cell Line, Tumor , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Protein Folding/drug effects , Transcription Factor CHOP/metabolism , Unfolded Protein Response/drug effects , Zebrafish
4.
Int J Mol Sci ; 20(1)2019 Jan 08.
Article in English | MEDLINE | ID: mdl-30626133

ABSTRACT

Ovarian cancer is the leading cause of death from gynecologic malignancy in the Western world. This is due, in part, to the fact that despite standard treatment of surgery and platinum/paclitaxel most patients recur with ultimately chemoresistant disease. Ovarian cancer is a unique form of solid tumor that develops, metastasizes and recurs in the same space, the abdominal cavity, which becomes a unique microenvironment characterized by ascites, hypoxia and low glucose levels. It is under these conditions that cancer cells adapt and switch to mitochondrial respiration, which becomes crucial to their survival, and therefore an ideal metabolic target for chemoresistant ovarian cancer. Importantly, independent of microenvironmental factors, mitochondria spatial redistribution has been associated to both tumor metastasis and chemoresistance in ovarian cancer while specific sets of genetic mutations have been shown to cause aberrant dependence on mitochondrial pathways in the most aggressive ovarian cancer subtypes. In this review we summarize on targeting mitochondria for treatment of chemoresistant ovarian cancer and current state of understanding of the role of mitochondria respiration in ovarian cancer. We feel this is an important and timely topic given that ovarian cancer remains the deadliest of the gynecological diseases, and that the mitochondrial pathway has recently emerged as critical in sustaining solid tumor progression.


Subject(s)
Drug Resistance, Neoplasm , Mitochondria/metabolism , Ovarian Neoplasms/therapy , Drug Evaluation, Preclinical , Female , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Spheroids, Cellular/pathology , Tumor Microenvironment
5.
Cancer Metastasis Rev ; 36(4): 635-653, 2017 12.
Article in English | MEDLINE | ID: mdl-29134486

ABSTRACT

Maintenance of protein homeostasis is a crucial process for the normal functioning of the cell. The regulated degradation of proteins is primarily facilitated by the ubiquitin proteasome system (UPS), a system of selective tagging of proteins with ubiquitin followed by proteasome-mediated proteolysis. The UPS is highly dynamic consisting of both ubiquitination and deubiquitination steps that modulate protein stabilization and degradation. Deregulation of protein stability is a common feature in the development and progression of numerous cancer types. Simultaneously, the elevated protein synthesis rate of cancer cells and consequential accumulation of misfolded proteins drives UPS addiction, thus sensitizing them to UPS inhibitors. This sensitivity along with the potential of stabilizing pro-apoptotic signaling pathways makes the proteasome an attractive clinical target for the development of novel therapies. Targeting of the catalytic 20S subunit of the proteasome is already a clinically validated strategy in multiple myeloma and other cancers. Spurred on by this success, promising novel inhibitors of the UPS have entered development, targeting the 20S as well as regulatory 19S subunit and inhibitors of deubiquitinating and ubiquitin ligase enzymes. In this review, we outline the manner in which deregulation of the UPS can cause cancer to develop, current clinical application of proteasome inhibitors, and the (pre-)clinical development of novel inhibitors of the UPS.


Subject(s)
Deubiquitinating Enzymes/metabolism , Neoplasms/drug therapy , Neoplasms/enzymology , Proteasome Endopeptidase Complex/metabolism , Animals , Deubiquitinating Enzymes/antagonists & inhibitors , Humans , Proteasome Inhibitors/administration & dosage
6.
Cancer Cell Int ; 18: 147, 2018.
Article in English | MEDLINE | ID: mdl-30263014

ABSTRACT

BACKGROUND: Drug screening for the identification of compounds with anticancer activity is commonly performed using cell lines cultured under normal oxygen pressure and physiological pH. However, solid tumors are characterized by a microenvironment with limited access to nutrients, reduced oxygen supply and acidosis. Tumor hypoxia and acidosis have been identified as important drivers of malignant progression and contribute to multicellular resistance to different forms of therapy. Tumor acidosis represents an important mechanism mediating drug resistance thus the identification of drugs active on acid-adapted cells may improve the efficacy of cancer therapy. METHODS: Here, we characterized human colon carcinoma cells (HCT116) chronically adapted to grow at pH 6.8 and used them to screen the Prestwick drug library for cytotoxic compounds. Analysis of gene expression profiles in parental and low pH-adapted cells showed several differences relating to cell cycle, metabolism and autophagy. RESULTS: The screen led to the identification of several compounds which were further selected for their preferential cytotoxicity towards acid-adapted cells. Amongst 11 confirmed hits, we primarily focused our investigation on the benzoporphyrin derivative Verteporfin (VP). VP significantly reduced viability in low pH-adapted HCT116 cells as compared to parental HCT116 cells and normal immortalized epithelial cells. The cytotoxic activity of VP was enhanced by light activation and acidic pH culture conditions, likely via increased acid-dependent drug uptake. VP displayed the unique property to cause light-dependent cross-linking of proteins and resulted in accumulation of polyubiquitinated proteins without inducing inhibition of the proteasome. CONCLUSIONS: Our study provides an example and a tool to identify anticancer drugs targeting acid-adapted cancer cells.

7.
Blood ; 123(5): 706-16, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24319254

ABSTRACT

Proteasome inhibitors have demonstrated that targeting protein degradation is effective therapy in multiple myeloma (MM). Here we show that deubiquitylating enzymes (DUBs) USP14 and UCHL5 are more highly expressed in MM cells than in normal plasma cells. USP14 and UCHL5 short interfering RNA knockdown decreases MM cell viability. A novel 19S regulatory particle inhibitor b-AP15 selectively blocks deubiquitylating activity of USP14 and UCHL5 without inhibiting proteasome activity. b-AP15 decreases viability in MM cell lines and patient MM cells, inhibits proliferation of MM cells even in the presence of bone marrow stroma cells, and overcomes bortezomib resistance. Anti-MM activity of b-AP15 is associated with growth arrest via downregulation of CDC25C, CDC2, and cyclin B1 as well as induction of caspase-dependent apoptosis and activation of unfolded protein response. In vivo studies using distinct human MM xenograft models show that b-AP15 is well tolerated, inhibits tumor growth, and prolongs survival. Combining b-AP15 with suberoylanilide hydroxamic acid, lenalidomide, or dexamethasone induces synergistic anti-MM activity. Our preclinical data showing efficacy of b-AP15 in MM disease models validates targeting DUBs in the ubiquitin proteasomal cascade to overcome proteasome inhibitor resistance and provides the framework for clinical evaluation of USP14/UCHL5 inhibitors to improve patient outcome in MM.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Multiple Myeloma/drug therapy , Piperidones/pharmacology , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Ubiquitin Thiolesterase/antagonists & inhibitors , Animals , Apoptosis/drug effects , Bortezomib , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Endoplasmic Reticulum Stress/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Ubiquitin Thiolesterase/genetics , Up-Regulation
8.
Drug Resist Updat ; 21-22: 20-9, 2015.
Article in English | MEDLINE | ID: mdl-26183292

ABSTRACT

Although more traditionally associated with degradation and maintenance of protein homeostasis, the ubiquitin-proteasome system (UPS) has emerged as a critical component in the regulation of cancer cell growth and survival. The development of inhibitors that block the proteolytic activities of the proteasome have highlighted its suitability as a bona fide anti-cancer drug target. However, key determinants including the development of drug resistance and dose-limiting toxicity call for the identification of alternative components of the UPS for novel drug targeting. Recently the deubiquitinases (DUBs), a diverse family of enzymes that catalyze ubiquitin removal, have attracted significant interest as targets for the development of next generation UPS inhibitors. In particular, pharmacological inhibition of the proteasomal cysteine DUBs (i.e., USP14 and UCHL5) has been shown to be particularly cytotoxic to cancer cells and inhibit tumour growth in several in vivo models. In the current review we focus on the modes of action of proteasome DUB inhibitors and discus the potential of DUB inhibitors to circumvent acquired drug resistance and provide a therapeutic option for the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Proteasome Inhibitors/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Humans , Neoplasms/enzymology , Proteasome Inhibitors/administration & dosage , Proteasome Inhibitors/adverse effects , Ubiquitin-Specific Proteases/antagonists & inhibitors
9.
Int J Cancer ; 136(4): E51-61, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25156627

ABSTRACT

The mechanism of multicellular drug resistance, defined as the reduced efficacy of chemotherapeutic drugs in solid tumors is incompletely understood. Here we report that colon carcinoma cells cultured as 3D microtissues (spheroids) display dramatic increases in the expression of a subset of type I interferon-(IFN)-stimulated genes (ISGs). A similar gene signature was associated previously with resistance to radiation and chemotherapy, prompting us to examine the underlying biological mechanisms. Analysis of spheroids formed by different tumor cell lines and studies using knock-down of gene expression showed that cell crowding leads to the induction of IFN regulatory factor-9 (IRF9) which together with STAT2 and independently of IFNs, is necessary for ISG upregulation. Increased expression of IRF9 alone was sufficient to induce the ISG subset in monolayer cells and to confer increased resistance to clinically used cytotoxic drugs. Our data reveal a novel mechanism of regulation of a subset of ISGs, leading to drug resistance in solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Apoptosis , Cell Communication , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferons/physiology , STAT2 Transcription Factor/metabolism , Transcriptional Activation
10.
Br J Haematol ; 169(3): 377-90, 2015 May.
Article in English | MEDLINE | ID: mdl-25691154

ABSTRACT

Deubiquitinase enzymes (DUBs) of the proteasomal 19S regulatory particle are emerging as important therapeutic targets in several malignancies. Here we demonstrate that inhibition of two proteasome-associated DUBs (USP14 and UCHL5) with the small molecule DUB inhibitor b-AP15, results in apoptosis of human Waldenström macroglobulinaemia (WM) cell lines and primary patient-derived WM tumour cells. Importantly, b-AP15 produced proteotoxic stress and apoptosis in WM cells that have acquired resistance to the proteasome inhibitor bortezomib. In silico modelling identified protein residues that were critical for the binding of b-AP15 with USP14 or UCHL5 and proteasome enzyme activity assays confirmed that b-AP15 does not affect the proteolytic capabilities of the 20S proteasome ß-subunits. In vitro toxicity from b-AP15 appeared to result from a build-up of ubiquitinated proteins and activation of the endoplasmic reticulum stress response in WM cells, an effect that also disrupted the mitochondria. Focused transcriptome profiling of b-AP15-treated WM cells revealed modulation of several genes regulating cell stress and NF-κB signalling, the latter whose protein translocation and downstream target activation was reduced by b-AP15 in vitro. This is the first report to define the effects and underlying mechanisms associated with inhibition of USP14 and UCHL5 DUB activity in WM tumour cells.


Subject(s)
Apoptosis/drug effects , Protease Inhibitors/pharmacology , Stress, Physiological/drug effects , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitination/drug effects , Waldenstrom Macroglobulinemia/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cluster Analysis , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation/drug effects , Gene Expression Profiling , Gene Regulatory Networks , Humans , Ligases/metabolism , Membrane Potential, Mitochondrial/drug effects , Models, Molecular , Molecular Conformation , NF-kappa B/metabolism , Piperidones/chemistry , Piperidones/pharmacology , Protease Inhibitors/chemistry , Proteasome Endopeptidase Complex/metabolism , Protein Transport , Proteolysis , Signal Transduction/drug effects , Stress, Physiological/genetics , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism , Waldenstrom Macroglobulinemia/genetics
11.
Int J Mol Sci ; 16(11): 27313-26, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26580606

ABSTRACT

The disorganized nature of tumor vasculature results in the generation of microenvironments characterized by nutrient starvation, hypoxia and accumulation of acidic metabolites. Tumor cell populations in such areas are often slowly proliferating and thus refractory to chemotherapeutical drugs that are dependent on an active cell cycle. There is an urgent need for alternative therapeutic interventions that circumvent growth dependency. The screening of drug libraries using multicellular tumor spheroids (MCTS) or glucose-starved tumor cells has led to the identification of several compounds with promising therapeutic potential and that display activity on quiescent tumor cells. Interestingly, a common theme of these drug screens is the recurrent identification of agents that affect mitochondrial function. Such data suggest that, contrary to the classical Warburg view, tumor cells in nutritionally-compromised microenvironments are dependent on mitochondrial function for energy metabolism and survival. These findings suggest that mitochondria may represent an "Achilles heel" for the survival of slowly-proliferating tumor cells and suggest strategies for the development of therapy to target these cell populations.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Resting Phase, Cell Cycle/drug effects , Animals , Cell Proliferation/drug effects , Drug Discovery , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Energy Metabolism/drug effects , Glucose/metabolism , Humans , Hypoxia , Molecular Targeted Therapy , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Oxidative Phosphorylation/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Tumor Cells, Cultured , Tumor Microenvironment/drug effects
12.
Mol Pharmacol ; 85(6): 932-45, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24714215

ABSTRACT

b-AP15 [(3E,5E)-3,5-bis[(4-nitrophenyl)methylidene]-1-(prop-2-enoyl)piperidin-4-one] is a small molecule inhibitor of the ubiquitin specific peptidase (USP) 14/ubiquitin carboxyl-terminal hydrolase (UCH) L5 deubiquitinases of the 19S proteasome that shows antitumor activity in a number of tumor models, including multiple myeloma. b-AP15 contains an α,ß-unsaturated carbonyl unit that is likely to react with intracellular nucleophiles such as cysteine thiolates by Michael addition. We found that binding of b-AP15 to USP14 is partially reversible, and that inhibition of proteasome function is reversible in cells. Despite reversible binding, tumor cells are rapidly committed to apoptosis/cell death after exposure to b-AP15. We show that b-AP15 is rapidly taken up from the medium and enriched in cells. Enrichment provides an explanation of the stronger potency of the compound in cellular assays compared with in vitro biochemical assays. Cellular uptake was impaired by 30-minute pretreatment of cells with low concentrations of N-ethylmaleimide (10 µM), suggesting that enrichment was thiol dependent. We report that in addition to inhibition of deubiquitinases, b-AP15 inhibits the selenoprotein thioredoxin reductase (TrxR). Whereas proteasome inhibition was closely associated with cell death induction, inhibition of TrxR was not. TrxR inhibition is, however, likely to contribute to triggering of oxidative stress observed with b-AP15. Furthermore, we present structure-activity, in vivo pharmacokinetic, and hepatocyte metabolism data for b-AP15. We conclude that the strong enrichment of b-AP15 in cells and a rapid commitment to apoptosis/cell death are factors that likely contribute to the strong antitumor activity of this compound.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Piperidones/pharmacology , Ubiquitin-Specific Proteases/antagonists & inhibitors , Cell Line, Tumor , Humans , Oxidative Stress/drug effects , Proteasome Endopeptidase Complex/drug effects
13.
Biochim Biophys Acta ; 1830(3): 2562-73, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23262140

ABSTRACT

BACKGROUND: Radiolabeled annexin A5 (AnxA5) is widely used for detecting phosphatidylserine exposed on cell surfaces during apoptosis. We describe here a new method for labeling AnxA5 and a size-matched control protein with short-lived carbon-11, for probing the specificity of in vivo cell death monitoring using positron emission tomography (PET) imaging. METHODS: AnxA5 and the control protein were recombinantly expressed with a C-terminal "Sel-tag", the tetrapeptide -Gly-Cys-Sec-Gly-COOH. The proteins were then labeled either fluorescently for in vitro corroborations of binding behaviors or with 11C for dynamic in vivo PET studies. RESULTS: AnxA5 demonstrated retained calcium-dependent binding to apoptotic cells after the C-terminus modification. The control protein showed no functional binding. The 11C-ligands demonstrated similar in vivo pharmacokinetic behavior in healthy mice except for higher uptake in kidney and higher intact elimination to urine of AnxA5. After inducing hepatic apoptosis, however, the uptake of labeled AnxA5 in the targeted tissue increased compared to baseline levels while that of the control protein tended to decrease. CONCLUSIONS: These data suggest that the combined use of these two tracers can facilitate differentiating specific AnxA5 binding and its changes caused by induced cell death from uptake due to non-specific permeability and retention effects at baseline or after therapy. GENERAL SIGNIFICANCE: The Sel-tag enables rapid and mild reactions with electrophilic agents giving site-specifically labeled proteins for multi-probe analyses. The combined use of 11C-labeled AnxA5 and a size-matched control protein with dynamic PET can be useful for evaluating drug effects on target as well as off-target tissues.


Subject(s)
Annexin A5/analysis , Isotope Labeling/methods , Liver/metabolism , Positron-Emission Tomography/methods , Radiopharmaceuticals/analysis , Animals , Annexin A5/chemistry , Annexin A5/metabolism , Apoptosis , Base Sequence , Biological Availability , Calcium/metabolism , Carbon Radioisotopes , Kidney/metabolism , Kinetics , Liver/diagnostic imaging , Liver/pathology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Radiography , Radiopharmaceuticals/chemical synthesis , Staining and Labeling/methods
14.
Stem Cells ; 31(8): 1715-25, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23554294

ABSTRACT

Adoptive transfer of third-party mesenchymal stromal cells (MSCs) has emerged as a promising tool for the treatment of steroid-refractory graft-versus-host disease (GVHD). Despite numerous in vitro studies and preclinical models, little is known about their effects on the patients' immune system. We assessed immune alterations in the T-cell, B-cell, natural killer cell, dendritic cell, and monocytic compartments of steroid-refractory GVHD patients 30, 90, and 180 days after MSC (n = 6) or placebo (n = 5) infusion, respectively. Infused MSCs were bioactive as suggested by the significant reduction in epithelial cell death, which represents a biomarker for acute GVHD. There were several indications that MSCs shift the patients' immune system toward a more tolerogenic profile. Most importantly, infusion of MSCs was associated with increased levels of regulatory (forkhead box P3 (FOXP3)(+) and interleukin (IL)-10(+) ) T-cells, reduced pro-inflammatory IL-17(+) T(Th17)-cells, and skewing toward type-2 T-helper cell responses. Furthermore, IL-2, which has been recently shown to exert a positive immune modulating effect in GVHD patients, was higher in the MSC patients at all evaluated time points during 6 months after MSC-infusion. Overall, our findings will contribute to the refinement of monitoring tools, for assessing MSC treatment-efficacy and increase our understanding regarding the MSCs' in vivo effects.


Subject(s)
Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Hematopoietic Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Adult , Aged , Female , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Hematologic Neoplasms/surgery , Humans , Immunity, Cellular/immunology , Immunotherapy, Adoptive/methods , Male , Middle Aged , Transplantation, Homologous
15.
Liver Int ; 34(3): 367-78, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24118944

ABSTRACT

BACKGROUND & AIMS: There is a demand for more sensitive, specific and predictive biomarkers for drug-induced liver injury (DILI) than the gold standard used today, alanine aminotransferase (ALT). The aim of this study was to qualify novel DILI biomarkers (keratin-18 markers M65/M30, microRNA-122, glutamate dehydrogenase and alpha-foetoprotein) in human DILI. METHODS: Levels of the novel biomarkers were measured by enzyme-linked immunosorbent assay or real-time quantitative reverse-transcription PCR (qRT-PCR) in two human DILI cohorts: a human volunteer study with acetaminophen and a human immunodeficiency virus (HIV)/tuberculosis (TB) study. RESULTS: In the acetaminophen study, serum M65 and microRNA-122 levels were significantly increased at an earlier time point than ALT. Furthermore, the maximal elevation of M65 and microRNA-122 exceeded the increase in ALT. In the HIV/TB study, all the analysed novel biomarkers increased after 1 week of treatment. In contrast to ALT, the novel biomarkers remained stable in a human cohort with exercise-induced muscular injury. CONCLUSIONS: M65 and microRNA-122 are potential biomarkers of DILI superior to ALT with respect to sensitivity and specificity.


Subject(s)
Alanine Transaminase/blood , Biomarkers/blood , Chemical and Drug Induced Liver Injury/diagnosis , Keratin-18/blood , MicroRNAs/blood , Peptide Fragments/blood , Chemical and Drug Induced Liver Injury/blood , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Humans , Sensitivity and Specificity
16.
NPJ Precis Oncol ; 8(1): 53, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38413842

ABSTRACT

Epithelial ovarian cancer (EOC) is the gynaecological malignancy with highest mortality. Although adjuvant treatment with carboplatin and paclitaxel leads to an objective response in ~80% of these patients, a majority will relapse within two years. Better methods for assessing long-term treatment outcomes are needed. To address this, we established safe and efficacious doses of carboplatin and paclitaxel using IGROV-1 zebrafish-CDX models. Then fluorescently-labelled cell suspensions from 83 tumour biopsies collected at exploratory laparotomy of women with suspected EOC were generated and 37 (45%) were successfully implanted in zebrafish larvae. Among these 19 of 27 pathology-confirmed EOC samples (70%) engrafted. These zebrafish patient-derived tumour xenograft (ZTX) models were treated with carboplatin or paclitaxel and tumour growth/regression and metastatic dissemination were recorded. In a subgroup of nine patients, four ZTX models regressed during carboplatin treatment. All four corresponding patients had >24 months PFS. Furthermore, both ZTX models established from two patients having <24 months PFS failed to regress during carboplatin treatment. Seven of eight models seeding <6 metastatic cells were established from patients having >24 months PFS. In eleven of fourteen patients, FIGO stage I + II or III tumours gave rise to ZTX models seeding <4 or >4 metastatic cells, respectively. In conclusion, ZTX models predicted patients having >24 or <24 months PFS, based on response/no response to carboplatin. Furthermore, high metastatic dissemination in ZTX models correlated to shorter PFS and more advanced disease at diagnosis. These preliminary results suggest that ZTX models could become a useful prognostic tool in EOC treatment planning.

17.
J Biol Chem ; 287(36): 30664-76, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22761435

ABSTRACT

Cyclic hypoxia and alterations in oncogenic signaling contribute to switch cancer cell metabolism from oxidative phosphorylation to aerobic glycolysis. A major consequence of up-regulated glycolysis is the increased production of metabolic acids responsible for the presence of acidic areas within solid tumors. Tumor acidosis is an important determinant of tumor progression and tumor pH regulation is being investigated as a therapeutic target. Autophagy is a cellular catabolic pathway leading to lysosomal degradation and recycling of proteins and organelles, currently considered an important survival mechanism in cancer cells under metabolic stress or subjected to chemotherapy. We investigated the response of human melanoma cells cultured in acidic conditions in terms of survival and autophagy regulation. Melanoma cells exposed to acidic culture conditions (7.0 < pH < 6.2) promptly accumulated LC3+ autophagic vesicles. Immunoblot analysis showed a consistent increase of LC3-II in acidic culture conditions as compared with cells at normal pH. Inhibition of lysosomal acidification by bafilomycin A1 further increased LC3-II accumulation, suggesting an active autophagic flux in cells under acidic stress. Acute exposure to acidic stress induced rapid inhibition of the mammalian target of rapamycin signaling pathway detected by decreased phosphorylation of p70S6K and increased phosphorylation of AMP-activated protein kinase, associated with decreased ATP content and reduced glucose and leucine uptake. Inhibition of autophagy by knockdown of the autophagic gene ATG5 consistently reduced melanoma cell survival in low pH conditions. These observations indicate that induction of autophagy may represent an adaptation mechanism for cancer cells exposed to an acidic environment. Our data strengthen the validity of therapeutic strategies targeting tumor pH regulation and autophagy in progressive malignancies.


Subject(s)
Autophagy , Melanoma/metabolism , Stress, Physiological , AMP-Activated Protein Kinases/metabolism , Autophagy-Related Protein 5 , Cell Hypoxia , Cell Line, Tumor , Humans , Hydrogen-Ion Concentration , Lysosomes/metabolism , Lysosomes/pathology , Melanoma/pathology , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Phosphorylation , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism
18.
Biochem Biophys Res Commun ; 431(2): 117-23, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23318177

ABSTRACT

Piperlongumine, a natural product from the plant Piperlongum, has demonstrated selective cytotoxicity to tumor cells and to show anti-tumor activity in animal models [1]. Cytotoxicity of piperlongumine has been attributed to increase in reactive oxygen species (ROS) in cancer cells. We here report that piperlongumine is an inhibitor of the ubiquitin-proteasome system (UPS). Exposure of tumor cells to piperlongumine resulted in accumulation of a reporter substrate known to be rapidly degraded by the proteasome, and of accumulation of ubiquitin conjugated proteins. However, no inhibition of 20S proteolytic activity or 19S deubiquitinating activity was observed at concentrations inducing cytotoxicity. Consistent with previous reports, piperlongumine induced strong ROS activation which correlated closely with UPS inhibition and cytotoxicity. Proteasomal blocking could not be mimicked by agents that induce oxidative stress. Our results suggest that the anti-cancer activity of piperlongumine involves inhibition of the UPS at a pre-proteasomal step, prior to deubiquitination of malfolded protein substrates at the proteasome, and that the previously reported induction of ROS is a consequence of this inhibition.


Subject(s)
Dioxolanes/pharmacology , Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Ubiquitin/antagonists & inhibitors , Cell Line, Tumor , Humans , Neoplasms/genetics , Oxidative Stress , Reactive Oxygen Species/metabolism , Transcriptome/drug effects
19.
Cancer Immunol Immunother ; 62(8): 1359-68, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23689729

ABSTRACT

The proteasome inhibitor bortezomib simultaneously renders tumor cells sensitive to killing by natural killer (NK) cells and resistant to killing by tumor-specific T cells. Here, we show that b-AP15, a novel inhibitor of proteasome deubiquitinating activity, sensitizes tumors to both NK and T cell-mediated killing. Exposure to b-AP15 significantly increased the susceptibility of tumor cell lines of various origins to NK (p < 0.0002) and T cell (p = 0.02)-mediated cytotoxicity. Treatment with b-AP15 resulted in increased tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor-2 expression (p = 0.03) and decreased cFLIP expression in tumor cells in vitro. In tumor-bearing SCID/Beige mice, treatment with b-AP15 followed by infusion of either human NK cells or tumor-specific T cells resulted in a significantly delayed tumor progression compared with mice treated with NK cells (p = 0.006), T cells (p < 0.0001) or b-AP15 alone (p = 0.003). Combined infusion of NK and T cells in tumor-bearing BALB/c mice following treatment with b-AP15 resulted in a significantly prolonged long-term survival compared with mice treated with b-AP15 and NK or T cells (p ≤ 0.01). Our findings show that b-AP15-induced sensitization to TRAIL-mediated apoptosis could be used as a novel strategy to augment the anticancer effects of adoptively infused NK and T cells in patients with cancer.


Subject(s)
Apoptosis/immunology , Killer Cells, Natural/immunology , Neoplasms/immunology , Piperidones/immunology , Protease Inhibitors/immunology , Proteasome Endopeptidase Complex/immunology , T-Lymphocytes/immunology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Combined Modality Therapy , Cytokines/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , HCT116 Cells , HeLa Cells , Humans , Immunotherapy, Adoptive , K562 Cells , Killer Cells, Natural/metabolism , Killer Cells, Natural/transplantation , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasms/pathology , Neoplasms/therapy , Piperidones/pharmacology , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , TNF-Related Apoptosis-Inducing Ligand/immunology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
20.
Invest New Drugs ; 31(3): 587-98, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23179339

ABSTRACT

Gambogic acid (GA), displays cytotoxicity towards a wide variety of tumor cells and has been shown to affect many important cell-signaling pathways. In the present work, we investigated the mechanism of action of GA by analysis of drug-induced changes in gene expression profiles and identified GA and the derivative dihydro GA as possible inhibitors of the ubiquitin-proteasome system (UPS). Both GA and dihydro GA inhibited proteasome function in cells resulting in the accumulation of polyubiquitin complexes. In vitro experiments showed that both GA and dihydro GA inhibited 20S chymotrypsin activity and the inhibitory effects of GA and dihydro GA on proteasome function corresponded with apoptosis induction and cell death. In conclusion, our results show that GA and dihydro GA exert their cytotoxic activity through inhibition of the UPS, specifically by acting as inhibitors of the chymotrypsin activity of the 20S proteasome.


Subject(s)
Antineoplastic Agents/pharmacology , Proteasome Inhibitors/pharmacology , Xanthones/pharmacology , Apoptosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , MCF-7 Cells , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL