Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
bioRxiv ; 2024 Apr 13.
Article de Anglais | MEDLINE | ID: mdl-38645232

RÉSUMÉ

Adenocarcinomas from multiple tissues can converge to treatment-resistant small cell neuroendocrine (SCN) cancers comprised of ASCL1, POU2F3, NEUROD1, and YAP1 subtypes. We investigated how mitochondrial metabolism influences SCN cancer (SCNC) progression. Extensive bioinformatics analyses encompassing thousands of patient tumors and human cancer cell lines uncovered enhanced expression of PGC-1α, a potent regulator of mitochondrial oxidative phosphorylation (OXPHOS), across several SCNC types. PGC-1α correlated tightly with increased expression of the lineage marker ASCL1 through a positive feedback mechanism. Analyses using a human prostate tissue-based SCN transformation system showed that the ASCL1 subtype has heightened PGC-1α expression and OXPHOS activity. PGC-1α inhibition diminished OXPHOS, reduced SCNC cell proliferation, and blocked SCN prostate tumor formation. PGC-1α overexpression enhanced OXPHOS, tripled the SCN prostate tumor formation rate, and promoted commitment to the ASCL1 lineage. These findings reveal the metabolic heterogeneity among SCNC subtypes and identify PGC-1α-induced OXPHOS as a regulator of SCNC lineage plasticity.

2.
Elife ; 122024 Mar 21.
Article de Anglais | MEDLINE | ID: mdl-38512723

RÉSUMÉ

Host-directed therapy (HDT) is an emerging approach to overcome antimicrobial resistance in pathogenic microorganisms. Specifically, HDT targets host-encoded factors required for pathogen replication and survival without interfering with microbial growth or metabolism, thereby eliminating the risk of resistance development. By applying HDT and a drug repurposing approach, we demonstrate that (R)-DI-87, a clinical-stage anticancer drug and potent inhibitor of mammalian deoxycytidine kinase (dCK), mitigates Staphylococcus aureus abscess formation in organ tissues upon invasive bloodstream infection. Mechanistically, (R)-DI-87 shields phagocytes from staphylococcal death-effector deoxyribonucleosides that target dCK and the mammalian purine salvage pathway-apoptosis axis. In this manner, (R)-DI-87-mediated protection of immune cells amplifies macrophage infiltration into deep-seated abscesses, a phenomenon coupled with enhanced pathogen control, ameliorated immunopathology, and reduced disease severity. Thus, pharmaceutical blockade of dCK represents an advanced anti-infective intervention strategy against which staphylococci cannot develop resistance and may help to fight fatal infectious diseases in hospitalized patients.


Sujet(s)
Anti-infectieux , Infections à staphylocoques , Animaux , Humains , Staphylococcus aureus , Deoxycytidine kinase , Abcès/anatomopathologie , Mammifères
3.
bioRxiv ; 2023 Dec 08.
Article de Anglais | MEDLINE | ID: mdl-37645972

RÉSUMÉ

Host-directed therapy (HDT) is an emerging approach to overcome antimicrobial resistance in pathogenic microorganisms. Specifically, HDT targets host-encoded factors required for pathogen replication and survival without interfering with microbial growth or metabolism, thereby eliminating the risk of resistance development. By applying HDT and a drug repurposing approach, we demonstrate that (R)-DI-87, a clinical-stage anti-cancer drug and potent inhibitor of mammalian deoxycytidine kinase (dCK), mitigates Staphylococcus aureus abscess formation in organ tissues upon invasive bloodstream infection. Mechanistically, (R)-DI-87 shields phagocytes from staphylococcal death-effector deoxyribonucleosides that target dCK and the mammalian purine salvage pathway-apoptosis axis. In this manner, (R)-DI-87-mediated protection of immune cells amplifies macrophage infiltration into deep-seated abscesses, a phenomenon coupled with enhanced pathogen control, ameliorated immunopathology, and reduced disease severity. Thus, pharmaceutical blockade of dCK represents an advanced anti-infective intervention strategy against which staphylococci cannot develop resistance and may help to fight fatal infectious diseases in hospitalized patients.

4.
Clin Cancer Res ; 29(16): 3130-3141, 2023 08 15.
Article de Anglais | MEDLINE | ID: mdl-37195712

RÉSUMÉ

PURPOSE: Stimulator of interferon genes (STING) agonists are currently in development for treatment of solid tumors, including pancreatic ductal adenocarcinoma (PDAC). Response rates to STING agonists alone have been promising yet modest, and combination therapies will likely be required to elicit their full potency. We sought to identify combination therapies and mechanisms that augment the tumor cell-intrinsic effect of therapeutically relevant STING agonists apart from their known effects on tumor immunity. EXPERIMENTAL DESIGN: We screened 430 kinase inhibitors to identify synergistic effectors of tumor cell death with diABZI, an intravenously administered and systemically available STING agonist. We deciphered the mechanisms of synergy with STING agonism that cause tumor cell death in vitro and tumor regression in vivo. RESULTS: We found that MEK inhibitors caused the greatest synergy with diABZI and that this effect was most pronounced in cells with high STING expression. MEK inhibition enhanced the ability of STING agonism to induce type I IFN-dependent cell death in vitro and tumor regression in vivo. We parsed NFκB-dependent and NFκB-independent mechanisms that mediate STING-driven type I IFN production and show that MEK signaling inhibits this effect by suppressing NFκB activation. CONCLUSIONS: Our results highlight the cytotoxic effects of STING agonism on PDAC cells that are independent of tumor immunity and that these therapeutic benefits of STING agonism can be synergistically enhanced by MEK inhibition.


Sujet(s)
Antinéoplasiques , Carcinome du canal pancréatique , Interféron de type I , Tumeurs du pancréas , Humains , Antinéoplasiques/pharmacologie , Transduction du signal , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/génétique , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/génétique , Mitogen-Activated Protein Kinase Kinases/métabolisme
5.
J Nucl Med ; 64(1): 117-123, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-35738905

RÉSUMÉ

Stimulator of interferon genes (STING) is a mediator of immune recognition of cytosolic DNA, which plays important roles in cancer, cytotoxic therapies, and infections with certain pathogens. Although pharmacologic STING activation stimulates potent antitumor immune responses in animal models, clinically applicable pharmacodynamic biomarkers that inform of the magnitude, duration, and location of immune activation elicited by systemic STING agonists are yet to be described. We investigated whether systemic STING activation induces metabolic alterations in immune cells that can be visualized by PET imaging. Methods: C57BL/6 mice were treated with systemic STING agonists and imaged with 18F-FDG PET after 24 h. Splenocytes were harvested 6 h after STING agonist administration and analyzed by single-cell RNA sequencing and flow cytometry. 18F-FDG uptake in total splenocytes and immunomagnetically enriched splenic B and T lymphocytes from STING agonist-treated mice was measured by γ-counting. In mice bearing prostate or pancreas cancer tumors, the effects of STING agonist treatment on 18F-FDG uptake, T-lymphocyte activation marker levels, and tumor growth were evaluated. Results: Systemic delivery of structurally distinct STING agonists in mice significantly increased 18F-FDG uptake in the spleen. The average spleen SUVmax in control mice was 1.90 (range, 1.56-2.34), compared with 4.55 (range, 3.35-6.20) in STING agonist-treated mice (P < 0.0001). Single-cell transcriptional and flow cytometry analyses of immune cells from systemic STING agonist-treated mice revealed enrichment of a glycolytic transcriptional signature in both T and B lymphocytes that correlated with the induction of immune cell activation markers. In tumor-bearing mice, STING agonist administration significantly delayed tumor growth and increased 18F-FDG uptake in secondary lymphoid organs. Conclusion: These findings reveal hitherto unknown functional links between STING signaling and immunometabolism and suggest that 18F-FDG PET may provide a widely applicable approach toward measuring the pharmacodynamic effects of systemic STING agonists at a whole-body level and guiding their clinical development.


Sujet(s)
Fluorodésoxyglucose F18 , Activation des lymphocytes , Mâle , Animaux , Souris , Fluorodésoxyglucose F18/métabolisme , Souris de lignée C57BL , Tomographie par émission de positons , Transduction du signal
6.
J Clin Invest ; 132(16)2022 08 15.
Article de Anglais | MEDLINE | ID: mdl-35653193

RÉSUMÉ

Purine nucleoside phosphorylase (PNP) enables the breakdown and recycling of guanine nucleosides. PNP insufficiency in humans is paradoxically associated with both immunodeficiency and autoimmunity, but the mechanistic basis for these outcomes is incompletely understood. Here, we identify two immune lineage-dependent consequences of PNP inactivation dictated by distinct gene interactions. During T cell development, PNP inactivation is synthetically lethal with downregulation of the dNTP triphosphohydrolase SAMHD1. This interaction requires deoxycytidine kinase activity and is antagonized by microenvironmental deoxycytidine. In B lymphocytes and macrophages, PNP regulates Toll-like receptor 7 signaling by controlling the levels of its (deoxy)guanosine nucleoside ligands. Overriding this regulatory mechanism promotes germinal center formation in the absence of exogenous antigen and accelerates disease in a mouse model of autoimmunity. This work reveals that one purine metabolism gene protects against immunodeficiency and autoimmunity via independent mechanisms operating in distinct immune lineages and identifies PNP as a potentially novel metabolic immune checkpoint.


Sujet(s)
Déficits immunitaires , Purine nucleoside phosphorylase , Animaux , Auto-immunité , Humains , Souris , Nucléoside purique , Purine nucleoside phosphorylase/génétique , Purine nucleoside phosphorylase/métabolisme , Lymphocytes T , Récepteur de type Toll-7
7.
Cell Rep ; 38(2): 110236, 2022 01 11.
Article de Anglais | MEDLINE | ID: mdl-35021095

RÉSUMÉ

We determine that type I interferon (IFN) response biomarkers are enriched in a subset of pancreatic ductal adenocarcinoma (PDAC) tumors; however, actionable vulnerabilities associated with IFN signaling have not been systematically defined. Integration of a phosphoproteomic analysis and a chemical genomics synergy screen reveals that IFN activates the replication stress response kinase ataxia telangiectasia and Rad3-related protein (ATR) in PDAC cells and sensitizes them to ATR inhibitors. IFN triggers cell-cycle arrest in S-phase, which is accompanied by nucleotide pool insufficiency and nucleoside efflux. In combination with IFN, ATR inhibitors induce lethal DNA damage and downregulate nucleotide biosynthesis. ATR inhibition limits the growth of PDAC tumors in which IFN signaling is driven by stimulator of interferon genes (STING). These results identify a cross talk between IFN, DNA replication stress response networks, and nucleotide metabolism while providing the rationale for targeted therapeutic interventions that leverage IFN signaling in tumors.


Sujet(s)
Carcinome du canal pancréatique/métabolisme , Interféron de type I/métabolisme , Adénocarcinome/métabolisme , Adénocarcinome/anatomopathologie , Animaux , Protéines mutées dans l'ataxie-télangiectasie/antagonistes et inhibiteurs , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Altération de l'ADN/effets des médicaments et des substances chimiques , Femelle , Humains , Interféron de type I/pharmacologie , Mâle , Protéines membranaires/métabolisme , Souris , Souris de lignée NOD , Nucléotides/antagonistes et inhibiteurs , Nucléotides/biosynthèse , Nucléotides/métabolisme , Tumeurs du pancréas/anatomopathologie , Inhibiteurs de protéines kinases/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Tumeurs du pancréas
8.
J Clin Invest ; 132(2)2022 01 18.
Article de Anglais | MEDLINE | ID: mdl-34813507

RÉSUMÉ

Various populations of cells are recruited to the heart after cardiac injury, but little is known about whether cardiomyocytes directly regulate heart repair. Using a murine model of ischemic cardiac injury, we demonstrate that cardiomyocytes play a pivotal role in heart repair by regulating nucleotide metabolism and fates of nonmyocytes. Cardiac injury induced the expression of the ectonucleotidase ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1), which hydrolyzes extracellular ATP to form AMP. In response to AMP, cardiomyocytes released adenine and specific ribonucleosides that disrupted pyrimidine biosynthesis at the orotidine monophosphate (OMP) synthesis step and induced genotoxic stress and p53-mediated cell death of cycling nonmyocytes. As nonmyocytes are critical for heart repair, we showed that rescue of pyrimidine biosynthesis by administration of uridine or by genetic targeting of the ENPP1/AMP pathway enhanced repair after cardiac injury. We identified ENPP1 inhibitors using small molecule screening and showed that systemic administration of an ENPP1 inhibitor after heart injury rescued pyrimidine biosynthesis in nonmyocyte cells and augmented cardiac repair and postinfarct heart function. These observations demonstrate that the cardiac muscle cell regulates pyrimidine metabolism in nonmuscle cells by releasing adenine and specific nucleosides after heart injury and provide insight into how intercellular regulation of pyrimidine biosynthesis can be targeted and monitored for augmenting tissue repair.


Sujet(s)
Myocarde/métabolisme , Myocytes cardiaques/métabolisme , Phosphodiesterases/métabolisme , Pyrimidines/biosynthèse , Pyrophosphatases/métabolisme , Régénération , Transduction du signal , AMP/génétique , AMP/métabolisme , Adénosine triphosphate/génétique , Adénosine triphosphate/métabolisme , Animaux , Lésions traumatiques du coeur/génétique , Lésions traumatiques du coeur/métabolisme , Souris , Phosphodiesterases/génétique , Pyrophosphatases/génétique
9.
J Nucl Med ; 63(5): 708-712, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-34593596

RÉSUMÉ

The purpose of this study was to evaluate 18F-FLT PET/CT as an early prognostic imaging biomarker of long-term overall survival and disease-specific survival (DSS) in soft-tissue sarcoma (STS) patients treated with neoadjuvant therapy (NAT) and surgical resection. Methods: This was a 10-y follow-up of a previous single-center, single-arm prospective clinical trial. Patients underwent 18F-FLT PET/CT before treatment (PET1) and after NAT (PET2). Posttreatment pathology specimens were assessed for tumor necrosis or fibrosis and for Ki-67 and thymidine kinase 1 expression. Maximally selected cutoffs for PET and histopathologic factors were applied. Survival was calculated from the date of subject consent to the date of death or last follow-up. Results: The study population consisted of 26 patients who underwent PET1; 16 of the 26 with primary STS underwent PET2. Thirteen deaths occurred during a median follow-up of 104 mo. In the overall cohort, overall survival was longer in patients with a low than a high PET1 tumor SUVmax (dichotomized by an SUVmax of ≥8.5 vs. <8.5: not yet reached vs. 49.7 mo; P = 0.0064). DSS showed a trend toward significance (P = 0.096). In a subanalysis of primary STS, DSS was significantly longer in patients with a low PET1 tumor SUVmax (dichotomized by an SUVmax of ≥8 vs. <8; P = 0.034). There were no significant 18F-FLT PET response thresholds corresponding to DSS or overall survival after NAT at PET2. Conclusion:18F-FLT PET may serve as a prognostic baseline imaging biomarker for DSS in patients with primary STS.


Sujet(s)
Tomographie par émission de positons couplée à la tomodensitométrie , Sarcomes , Marqueurs biologiques , Fluorodésoxyglucose F18 , Humains , Tomographie par émission de positons couplée à la tomodensitométrie/méthodes , Tomographie par émission de positons , Pronostic , Études prospectives , Radiopharmaceutiques/usage thérapeutique , Sarcomes/imagerie diagnostique , Sarcomes/thérapie
10.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Article de Anglais | MEDLINE | ID: mdl-34480004

RÉSUMÉ

Type I interferons (IFNs) are critical effectors of emerging cancer immunotherapies designed to activate pattern recognition receptors (PRRs). A challenge in the clinical translation of these agents is the lack of noninvasive pharmacodynamic biomarkers that indicate increased intratumoral IFN signaling following PRR activation. Positron emission tomography (PET) imaging enables the visualization of tissue metabolic activity, but whether IFN signaling-induced alterations in tumor cell metabolism can be detected using PET has not been investigated. We found that IFN signaling augments pancreatic ductal adenocarcinoma (PDAC) cell nucleotide metabolism via transcriptional induction of metabolism-associated genes including thymidine phosphorylase (TYMP). TYMP catalyzes the first step in the catabolism of thymidine, which competitively inhibits intratumoral accumulation of the nucleoside analog PET probe 3'-deoxy-3'-[18F]fluorothymidine ([18F]FLT). Accordingly, IFN treatment up-regulates cancer cell [18F]FLT uptake in the presence of thymidine, and this effect is dependent upon TYMP expression. In vivo, genetic activation of stimulator of interferon genes (STING), a PRR highly expressed in PDAC, enhances the [18F]FLT avidity of xenograft tumors. Additionally, small molecule STING agonists trigger IFN signaling-dependent TYMP expression in PDAC cells and increase tumor [18F]FLT uptake in vivo following systemic treatment. These findings indicate that [18F]FLT accumulation in tumors is sensitive to IFN signaling and that [18F]FLT PET may serve as a pharmacodynamic biomarker for STING agonist-based therapies in PDAC and possibly other malignancies characterized by elevated STING expression.


Sujet(s)
Didéoxynucléosides/administration et posologie , Radio-isotopes du fluor/administration et posologie , Interféron de type I/métabolisme , Protéines membranaires/métabolisme , Tumeurs du pancréas/métabolisme , Tomographie par émission de positons/méthodes , Animaux , Lignée cellulaire tumorale , Femelle , Humains , Mâle , Souris , Souris de lignée NOD , Tumeurs du pancréas/anatomopathologie , Transduction du signal , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Article de Anglais | MEDLINE | ID: mdl-33597293

RÉSUMÉ

Emerging evidence suggests that intratumoral interferon (IFN) signaling can trigger targetable vulnerabilities. A hallmark of pancreatic ductal adenocarcinoma (PDAC) is its extensively reprogrammed metabolic network, in which nicotinamide adenine dinucleotide (NAD) and its reduced form, NADH, are critical cofactors. Here, we show that IFN signaling, present in a subset of PDAC tumors, substantially lowers NAD(H) levels through up-regulating the expression of NAD-consuming enzymes PARP9, PARP10, and PARP14. Their individual contributions to this mechanism in PDAC have not been previously delineated. Nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme in the NAD salvage pathway, a dominant source of NAD in cancer cells. We found that IFN-induced NAD consumption increased dependence upon NAMPT for its role in recycling NAM to salvage NAD pools, thus sensitizing PDAC cells to pharmacologic NAMPT inhibition. Their combination decreased PDAC cell proliferation and invasion in vitro and suppressed orthotopic tumor growth and liver metastases in vivo.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Cytokines/antagonistes et inhibiteurs , Régulation de l'expression des gènes tumoraux , Interféron de type I/métabolisme , NAD/déficit , Nicotinamide phosphoribosyltransferase/antagonistes et inhibiteurs , Tumeurs du pancréas/anatomopathologie , Animaux , Apoptose , Marqueurs biologiques tumoraux/génétique , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme , Prolifération cellulaire , Cytokines/génétique , Cytokines/métabolisme , Humains , Interféron de type I/génétique , Mâle , Souris , Souris de lignée NOD , Souris SCID , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Nicotinamide phosphoribosyltransferase/génétique , Nicotinamide phosphoribosyltransferase/métabolisme , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Poly(ADP-ribose) polymerases/génétique , Poly(ADP-ribose) polymerases/métabolisme , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Transduction du signal , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
12.
Theranostics ; 10(2): 829-840, 2020.
Article de Anglais | MEDLINE | ID: mdl-31903153

RÉSUMÉ

Arginine (Arg) deprivation is a promising therapeutic approach for tumors with low argininosuccinate synthetase 1 (ASS1) expression. However, its efficacy as a single agent therapy needs to be improved as resistance is frequently observed. Methods: A tissue microarray was performed to assess ASS1 expression in surgical specimens of pancreatic ductal adenocarcinoma (PDAC) and its correlation with disease prognosis. An RNA-Seq analysis examined the role of ASS1 in regulating the global gene transcriptome. A high throughput screen of FDA-approved oncology drugs identified synthetic lethality between histone deacetylase (HDAC) inhibitors and Arg deprivation in PDAC cells with low ASS1 expression. We examined HDAC inhibitor panobinostat (PAN) and Arg deprivation in a panel of human PDAC cell lines, in ASS1-high and -knockdown/knockout isogenic models, in both anchorage-dependent and -independent cultures, and in multicellular complex cultures that model the PDAC tumor microenvironment. We examined the effects of combined Arg deprivation and PAN on DNA damage and the protein levels of key DNA repair enzymes. We also evaluated the efficacy of PAN and ADI-PEG20 (an Arg-degrading agent currently in Phase 2 clinical trials) in xenograft models with ASS1-low and -high PDAC tumors. Results: Low ASS1 protein level is a negative prognostic indicator in PDAC. Arg deprivation in ASS1-deficient PDAC cells upregulated asparagine synthetase (ASNS) which redirected aspartate (Asp) from being used for de novo nucleotide biosynthesis, thus causing nucleotide insufficiency and impairing cell cycle S-phase progression. Comprehensively validated, HDAC inhibitors and Arg deprivation showed synthetic lethality in ASS1-low PDAC cells. Mechanistically, combined Arg deprivation and HDAC inhibition triggered degradation of a key DNA repair enzyme C-terminal-binding protein interacting protein (CtIP), resulting in DNA damage and apoptosis. In addition, S-phase-retained ASS1-low PDAC cells (due to Arg deprivation) were also sensitized to DNA damage, thus yielding effective cell death. Compared to single agents, the combination of PAN and ADI-PEG20 showed better efficacy in suppressing ASS1-low PDAC tumor growth in mouse xenograft models. Conclusion: The combination of PAN and ADI-PEG20 is a rational translational therapeutic strategy for treating ASS1-low PDAC tumors through synergistic induction of DNA damage.


Sujet(s)
Arginine/déficit , Argininosuccinate synthase/métabolisme , Carcinome du canal pancréatique/traitement médicamenteux , Histone deacetylases/composition chimique , Hydrolases/pharmacologie , Tumeurs du pancréas/traitement médicamenteux , Panobinostat/pharmacologie , Polyéthylène glycols/pharmacologie , Animaux , Antinéoplasiques/pharmacologie , Argininosuccinate synthase/génétique , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Lignée cellulaire tumorale , Femelle , Inhibiteurs de désacétylase d'histone/pharmacologie , Humains , Mâle , Souris , Souris de lignée NOD , Souris SCID , Adulte d'âge moyen , Thérapie moléculaire ciblée/méthodes , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Pronostic , Mutations synthétiques létales
13.
RSC Med Chem ; 11(3): 392-410, 2020 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-33479645

RÉSUMÉ

A potent class of isoquinoline-based α-N-heterocyclic carboxaldehyde thiosemicarbazone (HCT) compounds has been rediscovered; based upon this scaffold, three series of antiproliferative agents were synthesized through iterative rounds of methylation and fluorination modifications, with anticancer activities being potentiated by physiologically relevant levels of copper. The lead compound, HCT-13, was highly potent against a panel of pancreatic, small cell lung carcinoma, prostate cancer, and leukemia models, with IC50 values in the low-to-mid nanomolar range. Density functional theory (DFT) calculations showed that fluorination at the 6-position of HCT-13 was beneficial for ligand-copper complex formation, stability, and ease of metal-center reduction. Through a chemical genomics screen, we identify DNA damage response/replication stress response (DDR/RSR) pathways, specifically those mediated by ataxia-telangiectasia and Rad3-related protein kinase (ATR), as potential compensatory mechanism(s) of action following HCT-13 treatment. We further show that the cytotoxicity of HCT-13 is copper-dependent, that it promotes mitochondrial electron transport chain (mtETC) dysfunction, induces production of reactive oxygen species (ROS), and selectively depletes guanosine nucleotide pools. Lastly, we identify metabolic hallmarks for therapeutic target stratification and demonstrate the in vivo efficacy of HCT-13 against aggressive models of acute leukemias in mice.

14.
Cell Chem Biol ; 27(2): 197-205.e6, 2020 02 20.
Article de Anglais | MEDLINE | ID: mdl-31734178

RÉSUMÉ

Biosynthesis of the pyrimidine nucleotide uridine monophosphate (UMP) is essential for cell proliferation and is achieved by the activity of convergent de novo and salvage metabolic pathways. Here we report the development and application of a cell-based metabolic modifier screening platform that leverages the redundancy in pyrimidine metabolism for the discovery of selective UMP biosynthesis modulators. In evaluating a library of protein kinase inhibitors, we identified multiple compounds that possess nucleotide metabolism modifying activity. The JNK inhibitor JNK-IN-8 was found to potently inhibit nucleoside transport and engage ENT1. The PDK1 inhibitor OSU-03012 (also known as AR-12) and the RAF inhibitor TAK-632 were shown to inhibit the therapeutically relevant de novo pathway enzyme DHODH and their affinities were unambiguously confirmed through in vitro assays and co-crystallization with human DHODH.


Sujet(s)
Transporteur équilibrant de nucléosides de type 1/antagonistes et inhibiteurs , Oxidoreductases acting on CH-CH group donors/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/composition chimique , Nucléosides pyrimidiques/métabolisme , Sites de fixation , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Cristallographie aux rayons X , Dihydroorotate dehydrogenase , Conception de médicament , Transporteur équilibrant de nucléosides de type 1/métabolisme , Humains , Simulation de dynamique moléculaire , Oxidoreductases acting on CH-CH group donors/génétique , Oxidoreductases acting on CH-CH group donors/métabolisme , Inhibiteurs de protéines kinases/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Bibliothèques de petites molécules/composition chimique
15.
Proc Natl Acad Sci U S A ; 116(14): 6842-6847, 2019 04 02.
Article de Anglais | MEDLINE | ID: mdl-30894490

RÉSUMÉ

Functional lysosomes mediate autophagy and macropinocytosis for nutrient acquisition. Pancreatic ductal adenocarcinoma (PDAC) tumors exhibit high basal lysosomal activity, and inhibition of lysosome function suppresses PDAC cell proliferation and tumor growth. However, the codependencies induced by lysosomal inhibition in PDAC have not been systematically explored. We performed a comprehensive pharmacological inhibition screen of the protein kinome and found that replication stress response (RSR) inhibitors were synthetically lethal with chloroquine (CQ) in PDAC cells. CQ treatment reduced de novo nucleotide biosynthesis and induced replication stress. We found that CQ treatment caused mitochondrial dysfunction and depletion of aspartate, an essential precursor for de novo nucleotide synthesis, as an underlying mechanism. Supplementation with aspartate partially rescued the phenotypes induced by CQ. The synergy of CQ and the RSR inhibitor VE-822 was comprehensively validated in both 2D and 3D cultures of PDAC cell lines, a heterotypic spheroid culture with cancer-associated fibroblasts, and in vivo xenograft and syngeneic PDAC mouse models. These results indicate a codependency on functional lysosomes and RSR in PDAC and support the translational potential of the combination of CQ and RSR inhibitors.


Sujet(s)
Acide aspartique/déficit , Carcinome du canal pancréatique , Chloroquine/pharmacologie , Lysosomes/métabolisme , Mitochondries , Tumeurs du pancréas , Animaux , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Lignée cellulaire tumorale , Femelle , Humains , Lysosomes/anatomopathologie , Mâle , Souris , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Stress physiologique , Tests d'activité antitumorale sur modèle de xénogreffe
16.
Nat Commun ; 8(1): 241, 2017 08 14.
Article de Anglais | MEDLINE | ID: mdl-28808226

RÉSUMÉ

Leukemia cells rely on two nucleotide biosynthetic pathways, de novo and salvage, to produce dNTPs for DNA replication. Here, using metabolomic, proteomic, and phosphoproteomic approaches, we show that inhibition of the replication stress sensing kinase ataxia telangiectasia and Rad3-related protein (ATR) reduces the output of both de novo and salvage pathways by regulating the activity of their respective rate-limiting enzymes, ribonucleotide reductase (RNR) and deoxycytidine kinase (dCK), via distinct molecular mechanisms. Quantification of nucleotide biosynthesis in ATR-inhibited acute lymphoblastic leukemia (ALL) cells reveals substantial remaining de novo and salvage activities, and could not eliminate the disease in vivo. However, targeting these remaining activities with RNR and dCK inhibitors triggers lethal replication stress in vitro and long-term disease-free survival in mice with B-ALL, without detectable toxicity. Thus the functional interplay between alternative nucleotide biosynthetic routes and ATR provides therapeutic opportunities in leukemia and potentially other cancers.Leukemic cells depend on the nucleotide synthesis pathway to proliferate. Here the authors use metabolomics and proteomics to show that inhibition of ATR reduced the activity of these pathways thus providing a valuable therapeutic target in leukemia.


Sujet(s)
Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Nucléotides/biosynthèse , Leucémie-lymphome lymphoblastique à précurseurs B et T/métabolisme , Animaux , Protéines mutées dans l'ataxie-télangiectasie/génétique , Voies de biosynthèse , Réplication de l'ADN , Deoxycytidine kinase/génétique , Deoxycytidine kinase/métabolisme , Femelle , Humains , Souris , Souris de lignée C57BL , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Ribonucleotide reductases/génétique , Ribonucleotide reductases/métabolisme
17.
Proc Natl Acad Sci U S A ; 113(15): 4027-32, 2016 Apr 12.
Article de Anglais | MEDLINE | ID: mdl-27035974

RÉSUMÉ

Deoxycytidine kinase (dCK), a rate-limiting enzyme in the cytosolic deoxyribonucleoside (dN) salvage pathway, is an important therapeutic and positron emission tomography (PET) imaging target in cancer. PET probes for dCK have been developed and are effective in mice but have suboptimal specificity and sensitivity in humans. To identify a more suitable probe for clinical dCK PET imaging, we compared the selectivity of two candidate compounds-[(18)F]Clofarabine; 2-chloro-2'-deoxy-2'-[(18)F]fluoro-9-ß-d-arabinofuranosyl-adenine ([(18)F]CFA) and 2'-deoxy-2'-[(18)F]fluoro-9-ß-d-arabinofuranosyl-guanine ([(18)F]F-AraG)-for dCK and deoxyguanosine kinase (dGK), a dCK-related mitochondrial enzyme. We demonstrate that, in the tracer concentration range used for PET imaging, [(18)F]CFA is primarily a substrate for dCK, with minimal cross-reactivity. In contrast, [(18)F]F-AraG is a better substrate for dGK than for dCK. [(18)F]CFA accumulation in leukemia cells correlated with dCK expression and was abrogated by treatment with a dCK inhibitor. Although [(18)F]CFA uptake was reduced by deoxycytidine (dC) competition, this inhibition required high dC concentrations present in murine, but not human, plasma. Expression of cytidine deaminase, a dC-catabolizing enzyme, in leukemia cells both in cell culture and in mice reduced the competition between dC and [(18)F]CFA, leading to increased dCK-dependent probe accumulation. First-in-human, to our knowledge, [(18)F]CFA PET/CT studies showed probe accumulation in tissues with high dCK expression: e.g., hematopoietic bone marrow and secondary lymphoid organs. The selectivity of [(18)F]CFA for dCK and its favorable biodistribution in humans justify further studies to validate [(18)F]CFA PET as a new cancer biomarker for treatment stratification and monitoring.


Sujet(s)
Nucléotides adényliques/composition chimique , Arabinonucléosides/composition chimique , Marqueurs biologiques tumoraux/composition chimique , Deoxycytidine kinase/analyse , Deoxycytidine kinase/métabolisme , Tomographie par émission de positons/méthodes , Animaux , Antinéoplasiques/composition chimique , Lignée cellulaire tumorale , Clofarabine , Produits de contraste/composition chimique , Deoxycytidine kinase/antagonistes et inhibiteurs , Humains , Leucémies/enzymologie , Souris , Tumeurs/traitement médicamenteux , Promédicaments/composition chimique , Rats
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE