Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 85
Filtrer
1.
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
2.
Article de Anglais | MEDLINE | ID: mdl-37874224

RÉSUMÉ

In June 2023, the Purine and Pyrimidine Society (PPS) organized the 20th biennial symposium on Purine and Pyrimidine metabolism (PP23). The symposium was organized in Los Angeles, California, USA, by Pr Caius Radu affiliated to UCLA. The scientific program covered various topics such as inborn errors, cancer, immunity, enzymatic reactions, drug development etc and was presented at 9 sessions over three days. The current issue of Nucleosides, Nucleotides & Nucleic Acids is a special issue covering proceedings from PP23-presentations and other PPS-related manuscripts, and in this editorial, we will give an overview of the scientific program of the meeting.

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 Virol ; 97(2): e0160022, 2023 02 28.
Article de Anglais | MEDLINE | ID: mdl-36757205

RÉSUMÉ

Infection by Kaposi sarcoma-associated herpesvirus (KSHV) can cause severe consequences, such as cancers and lymphoproliferative diseases. Whole inactivated viruses (WIV) with chemically destroyed genetic materials have been used as antigens in several licensed vaccines. During KSHV productive replication, virus-like vesicles (VLVs) that lack capsids and viral genomes are generated along with virions. Here, we investigated the immunogenicity of KSHV VLVs produced from a viral mutant that was defective in capsid formation and DNA packaging. Mice immunized with adjuvanted VLVs generated KSHV-specific T cell and antibody responses. Neutralization of KSHV infection by the VLV immune serum was low but was markedly enhanced in the presence of the complement system. Complement-enhanced neutralization and complement deposition on KSHV-infected cells was dependent on antibodies targeting viral open reading frame 4 (ORF4). However, limited complement-mediated enhancement was detected in the sera of a small cohort of KSHV-infected humans which contained few neutralizing antibodies. Therefore, vaccination that induces antibody effector functions can potentially improve infection-induced humoral immunity. Overall, our study highlights a potential benefit of engaging complement-mediated antibody functions in future KSHV vaccine development. IMPORTANCE KSHV is a virus that can lead to cancer after infection. A vaccine that prevents KSHV infection or transmission would be helpful in preventing the development of these cancers. We investigated KSHV VLV as an immunogen for vaccination. We determined that antibodies targeting the viral protein ORF4 induced by VLV immunization could engage the complement system and neutralize viral infection. However, ORF4-specific antibodies were seldom detected in the sera of KSHV-infected humans. Moreover, these human sera did not potently trigger complement-mediated neutralization, indicating an improvement that immunization can confer. Our study suggests a new antibody-mediated mechanism to control KSHV infection and underscores the benefit of activating the complement system in a future KSHV vaccine.


Sujet(s)
Anticorps neutralisants , Herpèsvirus humain de type 8 , Animaux , Humains , Souris , Anticorps neutralisants/immunologie , Infections à Herpesviridae , Herpèsvirus humain de type 8/immunologie , Cadres ouverts de lecture/immunologie , Vaccination , Protéines virales/immunologie
6.
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
7.
Cancer Discov ; 13(2): 298-311, 2023 02 06.
Article de Anglais | MEDLINE | ID: mdl-36472553

RÉSUMÉ

Mutations in the KRAS oncogene are found in more than 90% of patients with pancreatic ductal adenocarcinoma (PDAC), with Gly-to-Asp mutations (KRASG12D) being the most common. Here, we tested the efficacy of a small-molecule KRASG12D inhibitor, MRTX1133, in implantable and autochthonous PDAC models with an intact immune system. In vitro studies validated the specificity and potency of MRTX1133. In vivo, MRTX1133 prompted deep tumor regressions in all models tested, including complete or near-complete remissions after 14 days. Concomitant with tumor cell apoptosis and proliferative arrest, drug treatment led to marked shifts in the tumor microenvironment (TME), including changes in fibroblasts, matrix, and macrophages. T cells were necessary for MRTX1133's full antitumor effect, and T-cell depletion accelerated tumor regrowth after therapy. These results validate the specificity, potency, and efficacy of MRTX1133 in immunocompetent KRASG12D-mutant PDAC models, providing a rationale for clinical testing and a platform for further investigation of combination therapies. SIGNIFICANCE: Pharmacologic inhibition of KRASG12D in pancreatic cancer models with an intact immune system stimulates specific, potent, and durable tumor regressions. In the absence of overt toxicity, these results suggest that this and similar inhibitors should be tested as potential, high-impact novel therapies for patients with PDAC. See related commentary by Redding and Grabocka, p. 260. This article is highlighted in the In This Issue feature, p. 247.


Sujet(s)
Carcinome du canal pancréatique , Tumeurs du pancréas , Humains , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Mutation , Lignée cellulaire tumorale , Protéines proto-oncogènes p21(ras)/génétique , Microenvironnement tumoral , Tumeurs du pancréas
8.
Immunology ; 168(1): 152-169, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-35986643

RÉSUMÉ

Multiple sclerosis (MS) is an autoimmune disease driven by lymphocyte activation against myelin autoantigens in the central nervous system leading to demyelination and neurodegeneration. The deoxyribonucleoside salvage pathway with the rate-limiting enzyme deoxycytidine kinase (dCK) captures extracellular deoxyribonucleosides for use in intracellular deoxyribonucleotide metabolism. Previous studies have shown that deoxyribonucleoside salvage activity is enriched in lymphocytes and required for early lymphocyte development. However, specific roles for the deoxyribonucleoside salvage pathway and dCK in autoimmune diseases such as MS are unknown. Here we demonstrate that dCK activity is necessary for the development of clinical symptoms in the MOG35-55 and MOG1-125 experimental autoimmune encephalomyelitis (EAE) mouse models of MS. During EAE disease, deoxyribonucleoside salvage activity is elevated in the spleen and lymph nodes. Targeting dCK with the small molecule dCK inhibitor TRE-515 limits disease severity when treatments are started at disease induction or when symptoms first appear. EAE mice treated with TRE-515 have significantly fewer infiltrating leukocytes in the spinal cord, and TRE-515 blocks activation-induced B and T cell proliferation and MOG35-55 -specific T cell expansion without affecting innate immune cells or naïve T and B cell populations. Our results demonstrate that targeting dCK limits symptoms in EAE mice and suggest that dCK activity is required for MOG35-55 -specific lymphocyte activation-induced proliferation.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , Sclérose en plaques , Animaux , Souris , Deoxycytidine kinase/génétique , Lymphocytes/métabolisme , Modèles animaux de maladie humaine , Souris de lignée C57BL
9.
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
10.
Mol Cancer Ther ; 21(6): 936-947, 2022 06 01.
Article de Anglais | MEDLINE | ID: mdl-35313331

RÉSUMÉ

WNT signaling promotes pancreatic ductal adenocarcinoma (PDAC) through diverse effects on proliferation, differentiation, survival, and stemness. A subset of PDAC with inactivating mutations in ring finger protein 43 (RNF43) show growth dependency on autocrine WNT ligand signaling and are susceptible to agents that block WNT ligand acylation by Porcupine O-acyltransferase, which is required for proper WNT ligand processing and secretion. For this study, global transcriptomic, proteomic, and metabolomic analyses were performed to explore the therapeutic response of RNF43-mutant PDAC to the Porcupine inhibitor (PORCNi) LGK974. LGK974 disrupted cellular bioenergetics and mitochondrial function through actions that included rapid mitochondrial depolarization, reduced mitochondrial content, and inhibition of oxidative phosphorylation and tricarboxylic acid cycle. LGK974 also broadly altered transcriptional activity, downregulating genes involved in cell cycle, nucleotide metabolism, and ribosomal biogenesis and upregulating genes involved in epithelial-mesenchymal transition, hypoxia, endocytosis, and lysosomes. Autophagy and lysosomal activity were augmented in response to LGK974, which synergistically inhibited tumor cell viability in combination with chloroquine. Autocrine WNT ligand signaling dictates metabolic dependencies in RNF43-mutant PDAC through a combination of transcription dependent and independent effects linked to mitochondrial health and function. Metabolic adaptations to mitochondrial damage and bioenergetic stress represent potential targetable liabilities in combination with PORCNi for the treatment of WNT ligand-addicted PDAC.


Sujet(s)
Carcinome du canal pancréatique , Tumeurs du pancréas , Voie de signalisation Wnt , Acyltransferases/antagonistes et inhibiteurs , Acyltransferases/génétique , Acyltransferases/métabolisme , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire , Homéostasie , Humains , Ligands , Protéines membranaires/antagonistes et inhibiteurs , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Mitochondries/métabolisme , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Protéomique , Tumeurs du pancréas
11.
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
12.
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
13.
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
14.
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
15.
J Nucl Med ; 62(7): 989-995, 2021 07 01.
Article de Anglais | MEDLINE | ID: mdl-33277393

RÉSUMÉ

Prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT) is effective against prostate cancer (PCa), but all patients relapse eventually. Poor understanding of the underlying resistance mechanisms represents a key barrier to development of more effective RLT. We investigate the proteome and phosphoproteome in a mouse model of PCa to identify signaling adaptations triggered by PSMA RLT. Methods: Therapeutic efficacy of PSMA RLT was assessed by tumor volume measurements, time to progression, and survival in C4-2 or C4-2 TP53-/- tumor-bearing nonobese diabetic scid γ-mice. Two days after RLT, the proteome and phosphoproteome were analyzed by mass spectrometry. Results: PSMA RLT significantly improved disease control in a dose-dependent manner. Proteome and phosphoproteome datasets revealed activation of genotoxic stress response pathways, including deregulation of DNA damage/replication stress response, TP53, androgen receptor, phosphatidylinositol-3-kinase/AKT, and MYC signaling. C4-2 TP53-/- tumors were less sensitive to PSMA RLT than were parental counterparts, supporting a role for TP53 in mediating RLT responsiveness. Conclusion: We identified signaling alterations that may mediate resistance to PSMA RLT in a PCa mouse model. Our data enable the development of rational synergistic RLT-combination therapies to improve outcomes for PCa patients.


Sujet(s)
Tumeurs prostatiques résistantes à la castration , Humains , Mâle , Prostate , Antigène spécifique de la prostate
16.
J Nucl Med ; 62(2): 228-231, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-32646877

RÉSUMÉ

Prostate-specific membrane antigen (PSMA)-targeted radionuclide therapy (RNT) may increase tumor immunogenicity. We aimed at exploiting this effect by combining RNT with immunotherapy in a mouse model of prostate cancer (PC). Methods: C57BL/6-mice bearing syngeneic RM1-PGLS tumors were treated with 225Ac-PSMA617, an anti-PD-1 antibody, or both. Therapeutic efficacy was assessed by tumor volume measurements (CT), time to progression (TTP), and survival. Results: PSMA RNT or anti-PD-1 alone tended to prolong TTP (isotype control, 25 d; anti-PD-1, 33.5 d [P = 0.0153]; RNT, 30 d [P = 0.1038]) and survival (control, 28 d; anti-PD-1, 37 d [P = 0.0098]; RNT, 32 d [P = 0.1018]). Combining PSMA RNT and anti-PD-1 significantly improved disease control compared with either monotherapy. TTP was extended to 47.5 d (P ≤ 0.0199 vs. monotherapies), and survival to 51.5 d (P ≤ 0.0251 vs. monotherapies). Conclusion: PSMA RNT and PD-1 blockade synergistically improve therapeutic outcomes in our PC model, supporting the evaluation of RNT and immunotherapy combinations for PC patients.


Sujet(s)
Dipeptides/usage thérapeutique , Composés hétéromonocycliques/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Immunothérapie/méthodes , Tumeurs de la prostate/immunologie , Tumeurs de la prostate/radiothérapie , Actinium , Animaux , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Humains , Mâle , Souris , Souris de lignée C57BL , Récepteur-1 de mort cellulaire programmée/immunologie , Antigène spécifique de la prostate , Tumeurs de la prostate/anatomopathologie
17.
Proc Natl Acad Sci U S A ; 117(26): 15172-15181, 2020 06 30.
Article de Anglais | MEDLINE | ID: mdl-32532924

RÉSUMÉ

Hu11B6 is a monoclonal antibody that internalizes in cells expressing androgen receptor (AR)-regulated prostate-specific enzyme human kallikrein-related peptidase 2 (hK2; KLK2). In multiple rodent models, Actinium-225-labeled hu11B6-IgG1 ([225Ac]hu11B6-IgG1) has shown promising treatment efficacy. In the present study, we investigated options to enhance and optimize [225Ac]hu11B6 treatment. First, we evaluated the possibility of exploiting IgG3, the IgG subclass with superior activation of complement and ability to mediate FC-γ-receptor binding, for immunotherapeutically enhanced hK2 targeted α-radioimmunotherapy. Second, we compared the therapeutic efficacy of a single high activity vs. fractionated activity. Finally, we used RNA sequencing to analyze the genomic signatures of prostate cancer that progressed after targeted α-therapy. [225Ac]hu11B6-IgG3 was a functionally enhanced alternative to [225Ac]hu11B6-IgG1 but offered no improvement of therapeutic efficacy. Progression-free survival was slightly increased with a single high activity compared to fractionated activity. Tumor-free animals succumbing after treatment revealed no evidence of treatment-associated toxicity. In addition to up-regulation of canonical aggressive prostate cancer genes, such as MMP7, ETV1, NTS, and SCHLAP1, we also noted a significant decrease in both KLK3 (prostate-specific antigen ) and FOLH1 (prostate-specific membrane antigen) but not in AR and KLK2, demonstrating efficacy of sequential [225Ac]hu11B6 in a mouse model.


Sujet(s)
Actinium/usage thérapeutique , Immunoconjugués/usage thérapeutique , Antigène spécifique de la prostate/immunologie , Tumeurs de la prostate/thérapie , Kallicréines tissulaires/métabolisme , Particules alpha , Animaux , Marqueurs biologiques tumoraux , Humains , Mâle , Souris , Souris nude , Tumeurs expérimentales/thérapie
18.
Theranostics ; 10(6): 2612-2620, 2020.
Article de Anglais | MEDLINE | ID: mdl-32194823

RÉSUMÉ

225Ac-PSMA-617 targeted-therapy has demonstrated efficacy in 75-85% of patients; however, responses are not durable. We aimed to establish translatable mouse models of disseminated prostate cancer (PCa) to evaluate effectiveness of 225Ac-PSMA-617 at various disease stages. Methods: C4-2, C4-2B, or 22Rv1 cells were injected into the left ventricle of male NSG mice. Disease progression was monitored using bioluminescence imaging (BLI). For treatment, mice were injected with 40 kBq 225Ac-PSMA-617 at one (early treatment cohort) or three weeks (late treatment cohort) post-inoculation. Treatment efficacy was monitored by BLI of whole-body tumor burden. Mice were sacrificed based on body conditioning score. Results: C4-2 cells yielded metastases in liver, lungs, spleen, stomach, bones, and brain - achieving a clinically relevant model of widespread metastatic disease. The disease burden in the early treatment cohort was stable over 27 weeks in 5/9 mice and progressive in 4/9 mice. These mice were sacrificed due to brain metastases. Median survival of the late treatment cohort was superior to controls (13 vs. 7 weeks; p<0.0001) but inferior to that in the early treatment cohort (13 vs. 27 weeks; p<0.001). Late cohort mice succumbed to extensive liver involvement. The 22Rv1 and C4-2B systemic models were not used for treatment due to high kidney metastatic burden or low take rate, respectively. Conclusion: C4-2 cells reproduced metastatic cancer spread most relevantly. Early treatment with 225Ac-PSMA-617 prevented liver metastases and led to significant survival benefit. Late treatment improved survival without reducing tumor burden in the liver, the main site of metastasis. The current findings suggest that early 225Ac-PSMA-617 intervention is more efficacious in the setting of widespread metastatic PCa.


Sujet(s)
Actinium/usage thérapeutique , Dipeptides/usage thérapeutique , Composés hétéromonocycliques/usage thérapeutique , Tumeurs prostatiques résistantes à la castration/traitement médicamenteux , Radiopharmaceutiques/usage thérapeutique , Particules alpha/usage thérapeutique , Animaux , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Études de faisabilité , Humains , Mâle , Souris , Antigène spécifique de la prostate
19.
Clin Cancer Res ; 26(12): 2946-2955, 2020 06 15.
Article de Anglais | MEDLINE | ID: mdl-31932492

RÉSUMÉ

PURPOSE: Prostate-specific membrane antigen (PSMA) targeting radioligands deliver radiation to PSMA-expressing cells. However, the relationship between PSMA levels and intralesion heterogeneity of PSMA expression, and cytotoxic radiation by radioligand therapy (RLT) is unknown. Here we investigate RLT efficacy as function of PSMA levels/cell, and the fraction of PSMA+ cells in a tumor. EXPERIMENTAL DESIGN: RM1 cells expressing different levels of PSMA (PSMA-, PSMA+, PSMA++, PSMA+++; study 1) or a mix of PSMA+ and PSMA- RM1 (study 2, 4) or PC-3/PC-3-PIP (study 3) cells at various ratios were injected into mice. Mice received 177Lu- (studies 1-3) or 225Ac- (study 4) PSMA617. Tumor growth was monitored. Two days post-RLT, tumors were resected in a subset of mice. Radioligand uptake and DNA damage were quantified. RESULTS: 177Lu-PSMA617 efficacy increased with increasing PSMA levels (study 1) and fractions of PSMA positive cells (studies 2, 3) in both, the RM1 and PC-3-PIP models. In tumors resected 2 days post-RLT, PSMA expression correlated with 177Lu-PSMA617 uptake and the degree of DNA damage. Compared with 177Lu-PSMA617, 225Ac-PSMA617 improved overall antitumor effectiveness and tended to enhance the differences in therapeutic efficacy between experimental groups. CONCLUSIONS: In the current models, both the degree of PSMA expression and the fraction of PSMA+ cells correlate with 177Lu-/225Ac-PSMA617 tumor uptake and DNA damage, and thus, RLT efficacy. Low or heterogeneous PSMA expression represents a resistance mechanism to RLT.See related commentary by Ravi Kumar and Hofman, p. 2774.


Sujet(s)
Antigènes de surface , Antigène spécifique de la prostate , Animaux , Antigènes de surface/métabolisme , Altération de l'ADN , Ligands , Mâle , Souris , Antigène spécifique de la prostate/métabolisme , Tumeurs de la prostate , Cellules cancéreuses en culture
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...