Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 330
Filter
1.
J Cancer Res Clin Oncol ; 150(7): 348, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39002018

ABSTRACT

BACKGROUND & AIMS: Patients with intrahepatic cholangiocarcinoma (iCCA) respond poorly to immune checkpoint blockades (ICBs). In this study, we aimed to dissect the potential mechanisms underlying poor response to ICBs and explore a rational ICB-based combination therapy in iCCA. METHODS: scRNA-seq dataset GSE151530 was analyzed to investigate the differentially expressed genes in malignant cells following ICBs therapy. RNA-seq analysis and western blot assays were performed to examine the upstream and downstream signaling pathways of CD73. Subcutaneous tumor xenograft models were utilized to investigate the impact of CD73 on iCCA growth. Plasmid AKT/NICD-induced spontaneous murine iCCAs were used to explore the therapeutic efficacy of CD73 enzymatic inhibitor AB680 combined with PD-1 blockade. Time-of-flight mass cytometry (CyTOF) was conducted to identify the tumor-infiltrating immune cell populations and their functional changes in murine iCCAs treated with AB680 in combination with PD-1 antibody. RESULTS: scRNA-seq analysis identified elevated CD73 expression in malignant cells in response to ICBs therapy. Mechanistically, ICBs therapy upregulated CD73 expression in malignant cells via TNF-α/NF-κB signaling pathway. In vivo studies revealed that CD73 inhibition suppressed the growth of subcutaneous tumors, and achieved synergistic depression effects with gemcitabine and cisplatin (GC). Adenosine produced by CD73 activates AKT/GSK3ß/ß-catenin signaling axis in iCCA cells. CD73 inhibitor AB680 potentiates anti-tumor efficacy of PD-1 antibody in murine iCCAs. CyTOF analysis showed that AB680 combined with anti-PD-1 therapy promoted the infiltration of CD8+ T, CD4+ T cells, and NK cells in murine iCCAs, while simultaneously decreased the proportions of macrophages and neutrophils. Moreover, AB680 combined with anti-PD-1 significantly upregulated the expression of Granzyme B, Tbet and co-stimulatory molecule ICOS in infiltrating CD8+ T cells. CONCLUSIONS: CD73 inhibitor AB680 limits tumor progression and potentiates therapeutic efficacy of GC chemotherapy or anti-PD-1 treatment in iCCA. AB680 combined with anti-PD-1 therapy effectively elicits anti-tumor immune response.


Subject(s)
5'-Nucleotidase , Bile Duct Neoplasms , Cholangiocarcinoma , Immune Checkpoint Inhibitors , Programmed Cell Death 1 Receptor , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/pathology , Cholangiocarcinoma/immunology , Animals , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/metabolism , Mice , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Xenograft Model Antitumor Assays , Cell Line, Tumor , Disease Progression
2.
J Med Chem ; 67(11): 9686-9708, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38809692

ABSTRACT

High extracellular concentrations of adenosine triphosphate (ATP) in the tumor microenvironment generate adenosine by sequential dephosphorylation of CD39 and CD73, resulting in potent immunosuppression to inhibit T cell and natural killer (NK) cell function. CD73, as the determining enzyme for adenosine production, has been shown to correlate with poor clinical tumor prognosis. Conventional inhibitors as analogues of adenosine 5'-monophosphate (AMP) may have a risk of further metabolism to adenosine analogues. Here, we report a new series of malonic acid non-nucleoside inhibitors coordinating with zinc ions of CD73. Compound 12f was found to be a superior CD73 inhibitor (IC50 = 60 nM) by structural optimization, and its pharmacokinetic properties were investigated. In mouse tumor models, compound 12f showed excellent efficacy and reversal of immunosuppression in combination with chemotherapeutic agents or checkpoint inhibitors, suggesting that it deserves further development as a novel CD73 inhibitor.


Subject(s)
5'-Nucleotidase , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/metabolism , Animals , Humans , Mice , Malonates/pharmacology , Malonates/chemistry , Malonates/chemical synthesis , Zinc/chemistry , Zinc/metabolism , Structure-Activity Relationship , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Drug Discovery , Cell Line, Tumor
3.
J Med Chem ; 67(11): 9447-9464, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38748913

ABSTRACT

In this work, a series of bifunctional PD-L1/CD73 (cluster of differentiation 73) small-molecule inhibitors were designed and synthesized. Among them, CC-5 showed the strongest PD-L1 inhibitory effects with an IC50 of 6 nM and potent anti-CD73 activity with an IC50 of 0.773 µM. The high PD-L1/CD73 inhibitory activity of CC-5 was further confirmed by SPR assays with KD of 182 nM for human PD-L1 and 101 nM for CD73, respectively. Importantly, CC-5 significantly suppressed tumor growth in a CT26 and B16-F10 tumor model with TGI of 64.3% and 39.6%, respectively. Immunohistochemical (IHC) and flow cytometry analysis of tumor-infiltrating lymphocytes (TILs) indicated that CC-5 exerted anticancer effects via activating the tumor immune microenvironment. Collectively, CC-5 represents the first dual PD-L1/CD73 inhibitor worthy of further research as a bifunctional immunotherapeutic agent.


Subject(s)
5'-Nucleotidase , B7-H1 Antigen , Immunotherapy , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/metabolism , Humans , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Animals , Mice , Immunotherapy/methods , Cell Line, Tumor , Tumor Microenvironment/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemical synthesis , Drug Discovery , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Mice, Inbred C57BL , Cell Proliferation/drug effects , Structure-Activity Relationship , Mice, Inbred BALB C , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/chemistry , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/chemical synthesis
4.
J Nanobiotechnology ; 22(1): 214, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689291

ABSTRACT

Combination of tumor immunotherapy with photothermal therapy (PTT) is a feasible tactic to overcome the drawback of immunotherapy such as poor immune response. Via triggering the immunogenic cells death (ICD), PTT can stimulate the activity of immune cells, but meanwhile, the level of adenosine is elevated via the CD73-induced decomposition of ATP which is overexpressed accompanying with the PTT process, resulting in negative feedback to impair the immune stimulation. Herein, we developed a novel biomimetic photothermal nanodrug to specifically block CD73 for inhibition of adenosine production and more efficient priming of the suppressive immune microenvironments. The nanodrug, named as AptEM@CBA, is constructed by encapsulation of photothermal agent black phosphorus quantum dots (BPQDs) and selective CD73 inhibitor α, ß-Methyleneadenosine 5'-diphosphate (AMPCP) in chitosan nanogels, which are further covered with aptamer AS1411 modified erythrocyte membrane (EM) for biomimetic camouflage. With AS1411 induced active targeting and EM induced long blood circulation time, the enrichment of the nanodrug tumor sites is promoted. The photothermal treatment promotes the maturation of dendritic cells. Meanwhile, the release of AMPCP suppress the adenosine generation via CD73 blockade, alleviating the impairment of adenosine to dendritic cells and suppressing regulatory T cells, synergically stimulate the activity of T cells. The combination of CD73 blockade with PTT, not only suppresses the growth of primary implanted tumors, but also boosts strong antitumor immunity to inhibit the growth of distal tumors, providing good potential for tumor photoimmunotherapy.


Subject(s)
5'-Nucleotidase , Adenosine Diphosphate , Adenosine , Immunotherapy , Photothermal Therapy , Animals , Humans , Mice , 5'-Nucleotidase/antagonists & inhibitors , Adenosine/chemistry , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine Diphosphate/analogs & derivatives , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Biomimetics/methods , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/immunology , Immunotherapy/methods , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanoparticles/chemistry , Neoplasms/therapy , Neoplasms/drug therapy , Photothermal Therapy/methods , Quantum Dots/chemistry , Tumor Microenvironment/drug effects , Male
5.
J Clin Invest ; 134(10)2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38530357

ABSTRACT

Despite widespread utilization of immunotherapy, treating immune-cold tumors remains a challenge. Multiomic analyses and experimental validation identified the OTUD4/CD73 proteolytic axis as a promising target in treating immune-suppressive triple negative breast cancer (TNBC). Mechanistically, deubiquitylation of CD73 by OTUD4 counteracted its ubiquitylation by TRIM21, resulting in CD73 stabilization inhibiting tumor immune responses. We further demonstrated the importance of TGF-ß signaling for orchestrating the OTUD4/CD73 proteolytic axis within tumor cells. Spatial transcriptomics profiling discovered spatially resolved features of interacting malignant and immune cells pertaining to expression levels of OTUD4 and CD73. In addition, ST80, a newly developed inhibitor, specifically disrupted proteolytic interaction between CD73 and OTUD4, leading to reinvigoration of cytotoxic CD8+ T cell activities. In preclinical models of TNBC, ST80 treatment sensitized refractory tumors to anti-PD-L1 therapy. Collectively, our findings uncover what we believe to be a novel strategy for targeting the immunosuppressive OTUD4/CD73 proteolytic axis in treating immune-suppressive breast cancers with the inhibitor ST80.


Subject(s)
5'-Nucleotidase , Proteolysis , Triple Negative Breast Neoplasms , Animals , Female , Humans , Mice , 5'-Nucleotidase/genetics , 5'-Nucleotidase/immunology , 5'-Nucleotidase/antagonists & inhibitors , Cell Line, Tumor , GPI-Linked Proteins/immunology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Ubiquitination , Ubiquitin-Specific Proteases
6.
J Thorac Oncol ; 18(5): 650-656, 2023 05.
Article in English | MEDLINE | ID: mdl-36641093

ABSTRACT

INTRODUCTION: CD73 is overexpressed in EGFR-mutated NSCLC and may promote immune evasion, suggesting potential for combining CD73 blockers with EGFR tyrosine kinase inhibitors (TKIs). This phase 1b-2 study (NCT03381274) evaluated the anti-CD73 antibody oleclumab plus the third-generation EGFR TKI osimertinib in advanced EGFR-mutated NSCLC. METHODS: Patients had tissue T790M-negative NSCLC with TKI-sensitive EGFR mutations after progression on a first- or second-generation EGFR TKI and were osimertinib naive. They received osimertinib 80 mg orally once daily plus oleclumab 1500 mg (dose level 1 [DL1]) or 3000 mg (DL2) intravenously every 2 weeks. Primary end points included safety and objective response rate by Response Evaluation Criteria in Solid Tumors version 1.1. RESULTS: By July 9, 2021, five patients received DL1 and 21 received DL2. Of these patients, 60.0% and 85.7% had any-grade treatment-related adverse events (TRAEs) and 20.0% and 14.3% had grade 3 TRAEs, respectively. No dose-limiting toxicities, serious TRAEs, or deaths occurred. Four patients were T790M positive on retrospective circulating tumor DNA (ctDNA) testing; three had objective partial responses. In patients who were T790M negative in tumor and ctDNA, objective response rate was 25.0% at DL1 and 11.8% at DL2 (all partial responses); response durations at DL2 were 14.8 and 16.6 months. In patients receiving DL2, excluding those who were T790M positive by ctDNA, median progression-free survival was 7.4 months, and median overall survival was 24.8 months. DL2 was the recommended phase 2 dose. CONCLUSIONS: Oleclumab plus osimertinib was found to have moderate activity with acceptable tolerability in previously treated patients with advanced EGFR-mutated NSCLC.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Aniline Compounds , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Protein Kinase Inhibitors/pharmacology , Retrospective Studies , 5'-Nucleotidase/antagonists & inhibitors
7.
Steroids ; 188: 109112, 2022 12.
Article in English | MEDLINE | ID: mdl-36150476

ABSTRACT

The ecto-5'-nucleotidase (CD73) is an important enzyme in the adenosine pathway and catalyzes the extracellular hydrolysis of adenosine monophosphate (AMP) yielding adenosine which is involved in the inflammation and immunosuppression. Inhibitors of CD73 have potential as novel immunotherapy agents for the treatment of cancer and infection. In this study, we discovered a series of fluorinated betulinic acid derivatives as potent CD73 inhibitors by a fluorine scanning strategy. Among these, three compounds ZM522, ZM553 and ZM557 exhibited inhibitory activity with IC50 values of 0.56 uM, 0.74 uM and 0.47 uM, respectively. In addition, these compounds showed a 7-fold, 5-fold and 8-fold increase in activity compared to the positive control drug α, ß-methylene adenosine diphosphate (APCP) against the human CD73 enzyme. Two of these (ZM522 and ZM553) also exhibited effective interferon gamma (INF-γ) elevation and indicated the regulation of rescued T cell activation. Therefore, our study provides both a lead optimization strategy and potential compounds for further development of small molecule CD73 inhibitors.


Subject(s)
5'-Nucleotidase , Fluorine , Humans , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/metabolism , Adenosine , Pentacyclic Triterpenes/chemistry , Betulinic Acid
8.
Cell Mol Life Sci ; 79(3): 152, 2022 Feb 25.
Article in English | MEDLINE | ID: mdl-35212809

ABSTRACT

ATP and adenosine have emerged as important signaling molecules involved in vascular remodeling, retinal functioning and neurovascular coupling in the mammalian eye. However, little is known about the regulatory mechanisms of purinergic signaling in the eye. Here, we used three-dimensional multiplexed imaging, in situ enzyme histochemistry, flow cytometric analysis, and single cell transcriptomics to characterize the whole pattern of purine metabolism in mouse and human eyes. This study identified ecto-nucleoside triphosphate diphosphohydrolase-1 (NTPDase1/CD39), NTPDase2, and ecto-5'-nucleotidase/CD73 as major ocular ecto-nucleotidases, which are selectively expressed in the photoreceptor layer (CD73), optic nerve head, retinal vasculature and microglia (CD39), as well as in neuronal processes and cornea (CD39, NTPDase2). Specifically, microglial cells can create a spatially arranged network in the retinal parenchyma by extending and retracting their branched CD39high/CD73low processes and forming local "purinergic junctions" with CD39low/CD73- neuronal cell bodies and CD39high/CD73- retinal blood vessels. The relevance of the CD73-adenosine pathway was confirmed by flash electroretinography showing that pharmacological inhibition of adenosine production by injection of highly selective CD73 inhibitor PSB-12489 in the vitreous cavity of dark-adapted mouse eyes rendered the animals hypersensitive to prolonged bright light, manifested as decreased a-wave and b-wave amplitudes. The impaired electrical responses of retinal cells in PSB-12489-treated mice were not accompanied by decrease in total thickness of the retina or death of photoreceptors and retinal ganglion cells. Our study thus defines ocular adenosine metabolism as a complex and spatially integrated network and further characterizes the critical role of CD73 in maintaining the functional activity of retinal cells.


Subject(s)
5'-Nucleotidase/metabolism , Adenosine/metabolism , Light , Retina/radiation effects , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/genetics , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis/drug effects , Apoptosis/radiation effects , Apyrase/genetics , Apyrase/metabolism , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microglia/metabolism , Photoreceptor Cells/metabolism , Retina/metabolism , Retina/physiology , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism
9.
JCI Insight ; 7(3)2022 02 08.
Article in English | MEDLINE | ID: mdl-35132961

ABSTRACT

Treatment with anti-PD-1 and anti-PD-L1 therapies has shown durable clinical benefit in non-small cell lung cancer (NSCLC). However, patients with NSCLC with epidermal growth factor receptor (EGFR) mutations do not respond as well to treatment as patients without an EGFR mutation. We show that EGFR-mutated NSCLC expressed higher levels of CD73 compared with EGFR WT tumors and that CD73 expression was regulated by EGFR signaling. EGFR-mutated cell lines were significantly more resistant to T cell killing compared with WT cell lines through suppression of T cell proliferation and function. In a xenograft mouse model of EGFR-mutated NSCLC, neither anti-PD-L1 nor anti-CD73 antibody alone inhibited tumor growth compared with the isotype control. In contrast, the combination of both antibodies significantly inhibited tumor growth, increased the number of tumor-infiltrating CD8+ T cells, and enhanced IFN-γ and TNF-α production of these T cells. Consistently, there were increases in gene expression that corresponded to inflammation and T cell function in tumors treated with the combination of anti-PD-L1 and anti-CD73. Together, these results further support the combination of anti-CD73 and anti-PD-L1 therapies in treating EGFR-mutated NSCLC, while suggesting that increased T cell activity may play a role in response to therapy.


Subject(s)
5'-Nucleotidase , CD8-Positive T-Lymphocytes , Carcinoma, Non-Small-Cell Lung , ErbB Receptors , Immune Checkpoint Inhibitors , Lung Neoplasms , Mutation , Animals , Female , Humans , Mice , 5'-Nucleotidase/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , DNA Mutational Analysis , DNA, Neoplasm/genetics , Drug Therapy, Combination , ErbB Receptors/genetics , ErbB Receptors/metabolism , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, SCID , Neoplasms, Experimental , Signal Transduction
10.
J Med Chem ; 65(3): 2409-2433, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35080883

ABSTRACT

We recently reported N4-substituted 3-methylcytidine-5'-α,ß-methylenediphosphates as CD73 inhibitors, potentially useful in cancer immunotherapy. We now expand the structure-activity relationship of pyrimidine nucleotides as human CD73 inhibitors. 4-Chloro (MRS4598 16; Ki = 0.673 nM) and 4-iodo (MRS4620 18; Ki = 0.436 nM) substitution of the N4-benzyloxy group decreased Ki by ∼20-fold. Primary alkylamine derivatives coupled through a p-amido group with a varying methylene chain length (24 and 25) were functionalized congeners, for subsequent conjugation to carrier or reporter moieties. X-ray structures of hCD73 with two inhibitors indicated a ribose ring conformational adaptation, and the benzyloxyimino group (E configuration) binds to the same region (between the C-terminal and N-terminal domains) as N4-benzyl groups in adenine inhibitors. Molecular dynamics identified stabilizing interactions and predicted conformational diversity. Thus, by N4-benzyloxy substitution, we have greatly enhanced the inhibitory potency and added functionality enabling molecular probes. Their potential as anticancer drugs was confirmed by blocking CD73 activity in tumor tissues in situ.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , Cytosine Nucleotides/pharmacology , Diphosphonates/pharmacology , Enzyme Inhibitors/pharmacology , 5'-Nucleotidase/metabolism , Adult , Cytosine Nucleotides/chemical synthesis , Cytosine Nucleotides/metabolism , Diphosphonates/chemical synthesis , Diphosphonates/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Humans , Male , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Neoplasms/enzymology , Palatine Tonsil/enzymology , Protein Binding , Structure-Activity Relationship
11.
J Neurochem ; 160(3): 305-324, 2022 02.
Article in English | MEDLINE | ID: mdl-34905223

ABSTRACT

Extracellular adenosine plays prominent roles in the brain in both physiological and pathological conditions. Adenosine can be generated following the degradation of extracellular nucleotides by various types of ectonucleotidases. Several ectonucleotidases are present in the brain parenchyma: ecto-nucleotide triphosphate diphosphohydrolases 1 and 3 (NTPDase 1 and 3), ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (NPP 1), ecto-5'-nucleotidase (eN), and tissue non-specific alkaline phosphatase (TNAP, whose function in the brain has received little attention). Here we examined, in a living brain preparation, the role of these ectonucleotidases in generating extracellular adenosine. We recorded local field potentials evoked by electrical stimulation of the lateral olfactory tract in the mouse piriform cortex in vitro. Variations in adenosine level were evaluated by measuring changes in presynaptic inhibition generated by adenosine A1 receptors (A1Rs) activation. A1R-mediated presynaptic inhibition was present endogenously and was enhanced by bath-applied AMP and ATP. We hypothesized that inhibiting ectonucleotidases would reduce extracellular adenosine concentration, which would result in a weakening of presynaptic inhibition. However, inhibiting TNAP had no effect in controlling endogenous adenosine action and no effect on presynaptic inhibition induced by bath-applied AMP. Furthermore, contrary to our expectation, inhibiting TNAP reinforced, rather than reduced, presynaptic inhibition induced by bath-applied ATP. Similarly, inhibition of NTPDase 1 and 3, NPP1, and eN induced stronger, rather than weaker, presynaptic inhibition, both in endogenous condition and with bath-applied ATP and AMP. Consequently, attempts to suppress the functions of extracellular adenosine by blocking its extracellular synthesis in living brain tissue could have functional impacts opposite to those anticipated.


Subject(s)
Cerebral Cortex/drug effects , Enzyme Inhibitors/pharmacology , Nucleotidases/antagonists & inhibitors , Synaptic Transmission/drug effects , 5'-Nucleotidase/antagonists & inhibitors , Adenosine/metabolism , Adenosine A1 Receptor Agonists/pharmacology , Adenosine Monophosphate/pharmacology , Adenosine Triphosphate/pharmacology , Alkaline Phosphatase/antagonists & inhibitors , Animals , Electric Stimulation , Evoked Potentials/drug effects , Female , Mice , Mice, Inbred C57BL , Olfactory Bulb/drug effects , Receptor, Adenosine A1/drug effects , Receptor, Adenosine A1/metabolism
12.
Int J Mol Sci ; 22(23)2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34884548

ABSTRACT

Radiotherapy and chemotherapy are conventional cancer treatments. Around 60% of all patients who are diagnosed with cancer receive radio- or chemotherapy in combination with surgery during their disease. Only a few patients respond to the blockage of immune checkpoints alone, or in combination therapy, because their tumours might not be immunogenic. Under these circumstances, an increasing level of extracellular adenosine via the activation of ecto-5'-nucleotidase (CD73) and consequent adenosine receptor signalling is a typical mechanism that tumours use to evade immune surveillance. CD73 is responsible for the conversion of adenosine monophosphate to adenosine. CD73 is overexpressed in various tumour types. Hence, targetting CD73's signalling is important for the reversal of adenosine-facilitated immune suppression. In this study, we selected a potent series of the non-nucleotide small molecule inhibitors of CD73. Molecular docking studies were performed in order to examine the binding mode of the inhibitors inside the active site of CD73 and 3D-QSAR was used to study the structure-activity relationship. The obtained CoMFA (q2 = 0.844, ONC = 5, r2 = 0.947) and CoMSIA (q2 = 0.804, ONC = 4, r2 = 0.954) models showed reasonable statistical values. The 3D-QSAR contour map analysis revealed useful structural characteristics that were needed to modify non-nucleotide small molecule inhibitors. We used the structural information from the overall docking and 3D-QSAR results to design new, potent CD73 non-nucleotide inhibitors. The newly designed CD73 inhibitors exhibited higher activity (predicted pIC50) than the most active compound of all of the derivatives that were selected for this study. Further experimental studies are needed in order to validate the new CD73 inhibitors.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Molecular Docking Simulation , Quantitative Structure-Activity Relationship , Binding Sites , Catalytic Domain , Computer Simulation , GPI-Linked Proteins/antagonists & inhibitors , Humans , Models, Molecular , Molecular Structure , Protein Binding
13.
Nat Biomed Eng ; 5(11): 1306-1319, 2021 11.
Article in English | MEDLINE | ID: mdl-34725506

ABSTRACT

Antigen release resulting from the death of tumour cells induced by chemotherapies and targeted therapies can augment the antitumour responses induced by immune checkpoint blockade (ICB). However, tumours responding to ICB therapies often become resistant to them. Here we show that the specific targeting of tumour cells promotes the growth of tumour-cell variants that are resistant to ICB, and that the acquired resistance can be overcome via the concurrent depletion of tumour cells and of major types of immunosuppressive cell via a monoclonal antibody binding the enzyme CD73, which we identified as highly expressed on tumour cells and on regulatory T cells, myeloid-derived suppressor cells and tumour-associated macrophages, but not on cytolytic T lymphocytes, natural killer cells and dendritic cells. In mice with murine tumours, the systemic administration of anti-PD1 antibodies and anti-CD73 antibodies conjugated to a near-infrared dye prevented near-infrared-irradiated tumours from acquiring resistance to ICB and resulted in the eradication of advanced tumours. The elimination of immunosuppressive cells may overcome acquired resistance to ICB across a range of tumour types and combination therapies.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Drug Resistance, Neoplasm , Neoplasms , Programmed Cell Death 1 Receptor , 5'-Nucleotidase/antagonists & inhibitors , Animals , Killer Cells, Natural , Mice , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes, Regulatory
14.
Arch Pharm (Weinheim) ; 354(12): e2100300, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34697820

ABSTRACT

The antithrombotic prodrugs ticlopidine and clopidogrel are thienotetrahydro-pyridine derivatives that are metabolized in the liver to produce thiols that irreversibly block adenosine diphosphate (ADP)-activated P2Y12 receptors on thrombocytes. In their native, nonmetabolized form, both drugs were reported to act as inhibitors of ectonucleoside triphosphate diphosphohydrolase-1 (NTPDase1, CD39). CD39 catalyzes the extracellular hydrolysis of nucleoside tri- and diphosphates, mainly adenosine 5'-triphosphate (ATP) and ADP, yielding adenosine monophosphate, which is further hydrolyzed by ecto-5'-nucleotidase (CD73) to produce adenosine. While ATP has proinflammatory effects, adenosine is a potent anti-inflammatory, immunosuppressive agent. Inhibitors of CD39 and CD73 have potential as novel checkpoint inhibitors for the immunotherapy of cancer and infection. In the present study, we investigated 2-substituted thienotetrahydropyridine derivatives, structurally related to ticlopidine, as CD39 inhibitors. Due to their substituent on the 2-position, they will not be metabolically transformed into reactive thiols and can, therefore, be expected to be devoid of P2Y12 receptor-antagonistic activity in vivo. Several of the investigated 2-substituted thienotetrahydropyridine derivatives showed concentration-dependent inhibition of CD39. The most potent derivative, 32, showed similar CD39-inhibitory potency to ticlopidine, both acting as allosteric inhibitors. Compound 32 showed an improved selectivity profile: While ticlopidine blocked several NTPDase isoenzymes, 32 was characterized as a novel dual inhibitor of CD39 and CD73.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , Apyrase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Thienopyridines/pharmacology , Allosteric Regulation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , GPI-Linked Proteins/antagonists & inhibitors , Humans , Structure-Activity Relationship , Thienopyridines/chemical synthesis , Thienopyridines/chemistry , Ticlopidine/pharmacology
15.
Mol Pharm ; 18(11): 4015-4028, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34648293

ABSTRACT

Combination immunotherapy is a promising strategy to remove the inhibitory effect of the tumor microenvironment on immune effector cells, improving the efficacy of immune checkpoint inhibitor treatment in bladder cancer. However, it is challenging to deliver multiple drugs to the tumor tissue effectively and simultaneously to ensure optimal therapeutic effects. Macrophage-derived exosome-mimetic nanovesicles (EMVs) were designed and validated as a nanoplatform for coloading and delivery of the CD73 inhibitor (AB680) and the monoclonal antibody to programmed cell death ligand 1 (aPDL1). The tumor-targeting, biosafety, and therapeutic effects of these nanocomplexes (AB680@EMVs-aPDL1), as a combined immunotherapy strategy for bladder cancer, were assessed in vitro and in vivo. Our results indicate that the nanodrug system was highly stable, provided adequate biosafety, and enhanced tumor targeting in a mouse model of bladder cancer. Moreover, the CD73 inhibitor reduced extracellular adenosine production, and the combination therapy significantly promoted the activation and infiltration of cytotoxic T-lymphocytes, which helped to optimally suppress tumor growth and extend median survival in vivo. Therefore, using EMVs to deliver a combination of aPDL1 and the CD73 inhibitor may be a useful combined immunotherapy strategy for treating bladder cancer.


Subject(s)
Exosomes/chemistry , Immune Checkpoint Inhibitors/administration & dosage , Nanoparticle Drug Delivery System , T-Lymphocytes, Cytotoxic/drug effects , Urinary Bladder Neoplasms/drug therapy , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/immunology , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Cell Line, Tumor , Disease Models, Animal , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/immunology , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/cytology , Male , Mice , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology
16.
Mol Cancer Ther ; 20(11): 2250-2261, 2021 11.
Article in English | MEDLINE | ID: mdl-34482286

ABSTRACT

Despite some impressive clinical results with immune checkpoint inhibitors, the majority of patients with cancer do not respond to these agents, in part due to immunosuppressive mechanisms in the tumor microenvironment. High levels of adenosine in tumors can suppress immune cell function, and strategies to target the pathway involved in its production have emerged. CD73 is a key enzyme involved in adenosine production. This led us to identify a novel humanized antagonistic CD73 antibody, mAb19, with distinct binding properties. mAb19 potently inhibits the enzymatic activity of CD73 in vitro, resulting in an inhibition of adenosine formation and enhanced T-cell activation. We then investigated the therapeutic potential of combining CD73 antagonism with other immune modulatory and chemotherapeutic agents. Combination of mAb19 with a PD-1 inhibitor increased T-cell activation in vitro Interestingly, this effect could be further enhanced with an agonist of the adenosine receptor ADORA3. Adenosine levels were found to be elevated upon doxorubicin treatment in vivo, which could be blocked by CD73 inhibition. Combining CD73 antagonism with doxorubicin resulted in superior responses in vivo Furthermore, a retrospective analysis of rectal cancer patient samples demonstrated an upregulation of the adenosine pathway upon chemoradiation, providing further rationale for combining CD73 inhibition with chemotherapeutic agents.This study demonstrates the ability of a novel CD73 antibody to enhance T-cell function through the potent suppression of adenosine levels. In addition, the data highlight combination opportunities with standard of care therapies as well as with an ADORA3 receptor agonist to treat patients with solid tumors.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , Adenosine/therapeutic use , Immunosuppression Therapy/methods , Adenosine/pharmacology , Animals , Female , Humans , Mice , Tumor Microenvironment
17.
Int J Mol Sci ; 22(18)2021 Sep 11.
Article in English | MEDLINE | ID: mdl-34575993

ABSTRACT

In previous studies using isolated, paced guinea pig left atria, we observed that FSCPX, known as a selective A1 adenosine receptor antagonist, paradoxically increased the direct negative inotropic response to A1 adenosine receptor agonists (determined using concentration/effect (E/c) curves) if NBTI, a nucleoside transport inhibitor, was present. Based on mathematical modeling, we hypothesized that FSCPX blunted the cardiac interstitial adenosine accumulation in response to nucleoside transport blockade, probably by inhibiting CD39 and/or CD73, which are the two main enzymes of the interstitial adenosine production in the heart. The goal of the present study was to test this hypothesis. In vitro CD39 and CD73 inhibitor assays were carried out; furthermore, E/c curves were constructed in isolated, paced rat and guinea pig left atria using adenosine, CHA and CPA (two A1 adenosine receptor agonists), FSCPX, NBTI and NBMPR (two nucleoside transport inhibitors), and PSB-12379 (a CD73 inhibitor), measuring the contractile force. We found that FSCPX did not show any inhibitory effect during the in vitro enzyme assays. However, we successfully reproduced the paradox effect of FSCPX in the rat model, mimicked the "paradox" effect of FSCPX with PSB-12379, and demonstrated the lipophilia of FSCPX, which could explain the negative outcome of inhibitor assays with CD39 and CD73 dissolved in a water-based solution. Taken together, these three pieces of indirect evidence are strong enough to indicate that FSCPX possesses an additional action besides the A1 adenosine receptor antagonism, which action may be the inhibition of an ectonucleotidase. Incidentally, we found that POM-1 inhibited CD73, in addition to CD39.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , Adenosine A1 Receptor Antagonists/pharmacology , Apyrase/antagonists & inhibitors , Receptor, Adenosine A1/metabolism , Xanthines/pharmacology , 5'-Nucleotidase/metabolism , Animals , Antigens, CD/metabolism , Apyrase/metabolism , Guinea Pigs , Male , Rats , Rats, Wistar
18.
Cancer Lett ; 520: 385-399, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34407431

ABSTRACT

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editor-in-Chief. Following the publication of the above article, the Editor was notified that an error occurred in which all images were published with incorrect versions. The Editor has taken the decision that the manuscript is no longer acceptable in its current form, nor with a corrigendum, as the extensive changes to the figures and publication would lead to ambiguity for our readers. We have therefore made the decision to retract this manuscript from Cancer Letters with the possibility of resubmission and republication of the manuscript in its corrected form after peer review.


Subject(s)
5'-Nucleotidase/genetics , AC133 Antigen/genetics , B7-H1 Antigen/genetics , Small Cell Lung Carcinoma/therapy , 5'-Nucleotidase/antagonists & inhibitors , AC133 Antigen/immunology , Animals , Antibodies, Anti-Idiotypic/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Cell Line, Tumor , Female , Heterografts , Humans , Immunotherapy, Adoptive/trends , Male , Mice , Neoplasm Metastasis , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/immunology , Small Cell Lung Carcinoma/pathology , T-Lymphocytes/immunology , Tumor Burden
19.
Expert Opin Ther Pat ; 31(10): 867-876, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33909515

ABSTRACT

INTRODUCTION: Hydrolysis of AMP to adenosine and inorganic phosphate is catalyzed by 5´-ectonucleotidase, e5NT, alias CD73, a metalloenzyme incorporating two zinc ions at its active site. e5NT is involved in crucial physiological and pathological processes, such as immune ho meostasis, inflammation, and tumor progression. CD73 inhibitors belonging to the monoclonal antibodies (MAbs) and small molecules started to be considered as candidates for the immunotherapy of tumors. AREAS COVERED: We review the drug design landscape in the scientific and patent literature on CD73 inhibitors from 2017 to the present. Small-molecule inhibitors were mostly discussed, although the MAbs are also considered. EXPERT OPINION: Considerable advances have been reported in the design of nucleotide/nucleoside-based CD73 inhibitors, after the X-ray crystal structure of the enzyme in complex with the non-hydrolyzable ADP analog, adenosine (α,ß)-methylene diphosphate (AMPCP), was reported. A large number of highly effective such inhibitors are now available, through modifications of the nucleobase, sugar and zinc-binding groups of the lead. Few classes of non-nucleotide inhibitors were also reported, including flavones, anthraquinone ssulfonates, and primary sulfonamides. A highly potent ssmall-molecule CD73 inhibitor, AB680, is presently in the early phase of clinical trials as immunotherapeutic agents against various types of cancer.


Subject(s)
5'-Nucleotidase/antagonists & inhibitors , Immunotherapy/methods , Neoplasms/drug therapy , 5'-Nucleotidase/chemistry , 5'-Nucleotidase/immunology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Design , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/immunology , Humans , Neoplasms/immunology , Neoplasms/pathology , Patents as Topic , Structure-Activity Relationship
20.
Anticancer Res ; 41(3): 1231-1242, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33788714

ABSTRACT

BACKGROUND/AIM: Successful therapy of EGFR-mutant NSCLC remains a challenging task despite initial benefits with the usage of EGFR tyrosine kinase inhibitors. Cancer immunotherapy has shown promising results in certain tumors, but response rate in EGFR-mutant NCLC is low, because these tumors are thought to have weak immunogenicity. MATERIALS AND METHODS: We used data from in vivo NSCLC databases as well as from in vitro cell culture experiments to investigate the regulation of CD73 by EGFR. RESULTS: EGFR expression was correlated with CD73 expression in patients' datasets, with EGFR-mutant tumors showing higher expression than their EGFR wildtype counterparts. Treatment of EGFR-mutant NSCLC cell lines with EGFR TKI reduced expression of CD73 at both mRNA and protein level. Among EGFR downstream signaling pathways, the Ras-Raf-ERK pathway was involved in the regulation of CD73 expression directly via ERK1/2 without the engagement of RSKs or MSKs. CONCLUSION: The results of this study may provide novel therapeutic strategies for the treatment of oncogene-driven NSCLC.


Subject(s)
5'-Nucleotidase/physiology , Carcinoma, Non-Small-Cell Lung/drug therapy , Extracellular Signal-Regulated MAP Kinases/physiology , Lung Neoplasms/drug therapy , MAP Kinase Signaling System , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/genetics , Carcinoma, Non-Small-Cell Lung/physiopathology , Cell Line, Tumor , ErbB Receptors/physiology , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/physiology , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/physiopathology , MAP Kinase Signaling System/physiology , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL