Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Anal Chem ; 94(17): 6548-6556, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35438976

ABSTRACT

Early diagnosis of cancer is essential for the efficacy of treatment. Our group recently developed RAPPID, a bioluminescent immunoassay platform capable of measuring a wide panel of biomarkers directly in solution. Here, we developed and systematically screened different RAPPID sensors for sensitive detection of the soluble fraction of Axl (sAxl), a cell surface receptor that is overexpressed in several types of cancer. The best-performing RAPPID sensor, with a limit of detection of 8 pM and a >9-fold maximal change in emission ratio, was applied to measure Axl in three different contexts: clinically relevant sAxl levels (∼0.5 and ∼1 nM) in diluted blood plasma, proteolytically cleaved Axl in the cell culture medium of A431 and HeLa cancer cells, and Axl on the membrane of A431 cells. We further extended the sensor toolbox by developing dual-color RAPPID for simultaneous detection of Axl and EGFR on A431 and HeLa cells, as well as an AND-gate RAPPID that measures the concurrent presence of these two cell surface receptors on the same cell. These new RAPPID sensors provide attractive alternatives for more laborious protein detection and quantification methods such as FACS and immunostainings, due to their simple practical implantation and low intrinsic background signal.


Subject(s)
Neoplasms , Receptor Protein-Tyrosine Kinases , Biomarkers, Tumor , Early Detection of Cancer , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism
2.
J Biol Chem ; 288(35): 25173-25182, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23836909

ABSTRACT

Chemokine receptors and their ligands play a prominent role in immune regulation but many have also been implicated in inflammatory diseases such as multiple sclerosis, rheumatoid arthritis, allograft rejection after transplantation, and also in cancer metastasis. Most approaches to therapeutically target the chemokine system involve targeting of chemokine receptors with low molecular weight antagonists. Here we describe the selection and characterization of an unprecedented large and diverse panel of neutralizing Nanobodies (single domain camelid antibodies fragment) directed against several chemokines. We show that the Nanobodies directed against CCL2 (MCP-1), CCL5 (RANTES), CXCL11 (I-TAC), and CXCL12 (SDF-1α) bind the chemokines with high affinity (at nanomolar concentration), thereby blocking receptor binding, inhibiting chemokine-induced receptor activation as well as chemotaxis. Together, we show that neutralizing Nanobodies can be selected efficiently for effective and specific therapeutic treatment against a wide range of immune and inflammatory diseases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Chemokines/metabolism , Single-Domain Antibodies/pharmacology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Camelids, New World , Chemokines/chemistry , Chemokines/genetics , Chemokines/immunology , Humans , Immune System Diseases/drug therapy , Immune System Diseases/immunology , Inflammation/drug therapy , Inflammation/immunology , Mice , NIH 3T3 Cells , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology
3.
Purinergic Signal ; 9(1): 91-100, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23054444

ABSTRACT

The concept of functional selectivity offers great potential for the development of drugs that selectively activate a specific intracellular signaling pathway. During the last few years, it has become possible to systematically analyse compound libraries on G protein-coupled receptors (GPCRs) for this 'biased' form of signaling. We screened over 800 compounds targeting the class of adenosine A(1) receptors using a ß-arrestin-mediated signaling assay in U2OS cells as a G protein-independent readout for GPCR activation. A selection of compounds was further analysed in a G protein-mediated GTPγS assay. Additionally, receptor affinity of these compounds was determined in a radioligand binding assay with the agonist [(3)H]CCPA. Of all compounds tested, only LUF5589 9 might be considered as functionally selective for the G protein-dependent pathway, particularly in view of a likely overestimation of ß-arrestin signaling in the U2OS cells. Altogether, our study shows that functionally selective ligands for the adenosine A(1) receptor are rare, if existing at all. A thorough analysis of biased signaling on other GPCRs also reveals that only very few compounds can be considered functionally selective. This might indicate that the concept of functional selectivity is less common than speculated.


Subject(s)
Receptor, Adenosine A1/drug effects , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Arrestin/physiology , CHO Cells , Cell Line , Cell Membrane/drug effects , Cricetinae , Cricetulus , Data Interpretation, Statistical , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Ligands , Radioligand Assay , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
4.
EBioMedicine ; 93: 104663, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37379657

ABSTRACT

BACKGROUND: HexaBody®-CD38 (GEN3014) is a hexamerization-enhanced human IgG1 that binds CD38 with high affinity. The E430G mutation in its Fc domain facilitates the natural process of antibody hexamer formation upon binding to the cell surface, resulting in increased binding of C1q and potentiated complement-dependent cytotoxicity (CDC). METHODS: Co-crystallization studies were performed to identify the binding interface of HexaBody-CD38 and CD38. HexaBody-CD38-induced CDC, antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), trogocytosis, and apoptosis were assessed using flow cytometry assays using tumour cell lines, and MM patient samples (CDC). CD38 enzymatic activity was measured using fluorescence spectroscopy. Anti-tumour activity of HexaBody-CD38 was assessed in patient-derived xenograft mouse models in vivo. FINDINGS: HexaBody-CD38 binds a unique epitope on CD38 and induced potent CDC in multiple myeloma (MM), acute myeloid leukaemia (AML), and B-cell non-Hodgkin lymphoma (B-NHL) cells. Anti-tumour activity was confirmed in patient-derived xenograft models in vivo. Sensitivity to HexaBody-CD38 correlated with CD38 expression level and was inversely correlated with expression of complement regulatory proteins. Compared to daratumumab, HexaBody-CD38 showed enhanced CDC in cell lines with lower levels of CD38 expression, without increasing lysis of healthy leukocytes. More effective CDC was also confirmed in primary MM cells. Furthermore, HexaBody-CD38 efficiently induced ADCC, ADCP, trogocytosis, and apoptosis after Fc-crosslinking. Moreover, HexaBody-CD38 strongly inhibited CD38 cyclase activity, which is hypothesized to relieve immune suppression in the tumour microenvironment. INTERPRETATION: Based on these preclinical studies, a clinical trial was initiated to assess the clinical safety of HexaBody-CD38 in patients with MM. FUNDING: Genmab.


Subject(s)
Antineoplastic Agents , Multiple Myeloma , Humans , Animals , Mice , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Antibody-Dependent Cell Cytotoxicity , Cell Line, Tumor , Complement System Proteins/metabolism , Tumor Microenvironment
5.
Life Sci Alliance ; 5(11)2022 Nov.
Article in English | MEDLINE | ID: mdl-36271507

ABSTRACT

CD3 bispecific antibodies (bsAbs) show great promise as anticancer therapeutics. Here, we show in-depth mechanistic studies of a CD3 bsAb in solid cancer, using DuoBody-CD3x5T4. Cross-linking T cells with tumor cells expressing the oncofetal antigen 5T4 was required to induce cytotoxicity. Naive and memory CD4+ and CD8+ T cells were equally effective at mediating cytotoxicity, and DuoBody-CD3x5T4 induced partial differentiation of naive T-cell subsets into memory-like cells. Tumor cell kill was associated with T-cell activation, proliferation, and production of cytokines, granzyme B, and perforin. Genetic knockout of FAS or IFNGR1 in 5T4+ tumor cells abrogated tumor cell kill. In the presence of 5T4+ tumor cells, bystander kill of 5T4- but not of 5T4-IFNGR1- tumor cells was observed. In humanized xenograft models, DuoBody-CD3x5T4 antitumor activity was associated with intratumoral and peripheral blood T-cell activation. Lastly, in dissociated patient-derived tumor samples, DuoBody-CD3x5T4 activated tumor-infiltrating lymphocytes and induced tumor-cell cytotoxicity, even when most tumor-infiltrating lymphocytes expressed PD-1. These data provide an in-depth view on the mechanism of action of a CD3 bsAb in preclinical models of solid cancer.


Subject(s)
Antibodies, Bispecific , Neoplasms , Humans , Antibodies, Bispecific/pharmacology , CD8-Positive T-Lymphocytes , Granzymes/pharmacology , CD3 Complex/pharmacology , Cytotoxicity, Immunologic , Perforin/pharmacology , Programmed Cell Death 1 Receptor , Neoplasms/drug therapy , Cytokines
6.
J Immunother Cancer ; 10(6)2022 06.
Article in English | MEDLINE | ID: mdl-35688554

ABSTRACT

BACKGROUND: Despite the preclinical promise of CD40 and 4-1BB as immuno-oncology targets, clinical efforts evaluating CD40 and 4-1BB agonists as monotherapy have found limited success. DuoBody-CD40×4-1BB (GEN1042/BNT312) is a novel investigational Fc-inert bispecific antibody for dual targeting and conditional stimulation of CD40 and 4-1BB to enhance priming and reactivation of tumor-specific immunity in patients with cancer. METHODS: Characterization of DuoBody-CD40×4-1BB in vitro was performed in a broad range of functional immune cell assays, including cell-based reporter assays, T-cell proliferation assays, mixed-lymphocyte reactions and tumor-infiltrating lymphocyte assays, as well as live-cell imaging. The in vivo activity of DuoBody-CD40×4-1BB was assessed in blood samples from patients with advanced solid tumors that were treated with DuoBody-CD40×4-1BB in the dose-escalation phase of the first-in-human clinical trial (NCT04083599). RESULTS: DuoBody-CD40×4-1BB exhibited conditional CD40 and 4-1BB agonist activity that was strictly dependent on crosslinking of both targets. Thereby, DuoBody-CD40×4-1BB strengthened the dendritic cell (DC)/T-cell immunological synapse, induced DC maturation, enhanced T-cell proliferation and effector functions in vitro and enhanced expansion of patient-derived tumor-infiltrating lymphocytes ex vivo. The addition of PD-1 blocking antibodies resulted in potentiation of T-cell activation and effector functions in vitro compared with either monotherapy, providing combination rationale. Furthermore, in a first-in-human clinical trial, DuoBody-CD40×4-1BB mediated clear immune modulation of peripheral antigen presenting cells and T cells in patients with advanced solid tumors. CONCLUSION: DuoBody-CD40×4-1BB is capable of enhancing antitumor immunity by modulating DC and T-cell functions and shows biological activity in patients with advanced solid tumors. These findings demonstrate that targeting of these two pathways with an Fc-inert bispecific antibody may be an efficacious approach to (re)activate tumor-specific immunity and support the clinical investigation of DuoBody-CD40×4-1BB for the treatment of cancer.


Subject(s)
Antibodies, Bispecific , Neoplasms , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , CD40 Antigens/metabolism , Clinical Trials as Topic , Humans , Lymphocyte Activation , Neoplasms/therapy , T-Lymphocytes
7.
Oncoimmunology ; 11(1): 2030135, 2022.
Article in English | MEDLINE | ID: mdl-35186440

ABSTRACT

Immune checkpoint inhibitors (ICI) targeting the PD-1/PD-L1 axis have changed the treatment paradigm for advanced solid tumors; however, many patients experience treatment resistance. In preclinical models 4-1BB co-stimulation synergizes with ICI by activating cytotoxic T- and NK-cell-mediated anti-tumor immunity. Here we characterize the mechanism of action of a mouse-reactive Fc-inert PD-L1×4-1BB bispecific antibody (mbsAb-PD-L1×4-1BB) and provide proof-of-concept for enhanced anti-tumor activity. In reporter assays mbsAb-PD-L1×4-1BB exhibited conditional 4-1BB agonist activity that was dependent on simultaneous binding to PD-L1. mbsAb-PD-L1×4-1BB further blocked the PD-L1/PD-1 interaction independently of 4-1BB binding. By combining both mechanisms, mbsAb-PD-L1×4-1BB strongly enhanced T-cell proliferation, cytokine production and antigen-specific cytotoxicity using primary mouse cells in vitro. Furthermore, mbsAb-PD-L1×4-1BB exhibited potent anti-tumor activity in the CT26 and MC38 models in vivo, leading to the rejection of CT26 tumors that were unresponsive to PD-L1 blockade alone. Anti-tumor activity was associated with increased tumor-specific CD8+ T cells and reduced regulatory T cells within the tumor microenvironment and tumor-draining lymph nodes. In immunocompetent tumor-free mice, mbsAb-PD-L1×4-1BB treatment neither induced T-cell infiltration into the liver nor elevated liver enzymes in the blood. Dual targeting of PD-L1 and 4-1BB with a bispecific antibody may therefore address key limitations of first generation 4-1BB-agonistic antibodies, and may provide a novel approach to improve PD-1/PD-L1 checkpoint blockade.


Subject(s)
Antibodies, Bispecific , Neoplasms , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , B7-H1 Antigen , CD8-Positive T-Lymphocytes , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mice , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/therapeutic use , Tumor Microenvironment
8.
Cancer Discov ; 12(5): 1248-1265, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35176764

ABSTRACT

Checkpoint inhibitors (CPI) have revolutionized the treatment paradigm for advanced solid tumors; however, there remains an opportunity to improve response rates and outcomes. In preclinical models, 4-1BB costimulation synergizes with CPIs targeting the programmed cell death protein 1 (PD-1)/programmed cell death ligand 1 (PD-L1) axis by activating cytotoxic T-cell-mediated antitumor immunity. DuoBody-PD-L1×4-1BB (GEN1046) is an investigational, first-in-class bispecific immunotherapy agent designed to act on both pathways by combining simultaneous and complementary PD-L1 blockade and conditional 4-1BB stimulation in one molecule. GEN1046 induced T-cell proliferation, cytokine production, and antigen-specific T-cell-mediated cytotoxicity superior to clinically approved PD-(L)1 antibodies in human T-cell cultures and exerted potent antitumor activity in transplantable mouse tumor models. In dose escalation of the ongoing first-in-human study in heavily pretreated patients with advanced refractory solid tumors (NCT03917381), GEN1046 demonstrated pharmacodynamic immune effects in peripheral blood consistent with its mechanism of action, manageable safety, and early clinical activity [disease control rate: 65.6% (40/61)], including patients resistant to prior PD-(L)1 immunotherapy. SIGNIFICANCE: DuoBody-PD-L1×4-1BB (GEN1046) is a first-in-class bispecific immunotherapy with a manageable safety profile and encouraging preclinical and early clinical activity. With its ability to confer clinical benefit in tumors typically less sensitive to CPIs, GEN1046 may fill a clinical gap in CPI-relapsed or refractory disease or as a combination therapy with CPIs. See related commentary by Li et al., p. 1184. This article is highlighted in the In This Issue feature, p. 1171.


Subject(s)
Antibodies, Bispecific , Neoplasms , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , B7-H1 Antigen , Disease Models, Animal , Humans , Immunotherapy/methods , Mice , Neoplasms/drug therapy , T-Lymphocytes
9.
Purinergic Signal ; 7(2): 171-92, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21544511

ABSTRACT

Adenosine receptors are plasma membrane proteins that transduce an extracellular signal into the interior of the cell. Basically every mammalian cell expresses at least one of the four adenosine receptor subtypes. Recent insight in signal transduction cascades teaches us that the current classification of receptor ligands into agonists, antagonists, and inverse agonists relies very much on the experimental setup that was used. Upon activation of the receptors by the ubiquitous endogenous ligand adenosine they engage classical G protein-mediated pathways, resulting in production of second messengers and activation of kinases. Besides this well-described G protein-mediated signaling pathway, adenosine receptors activate scaffold proteins such as ß-arrestins. Using innovative and sensitive experimental tools, it has been possible to detect ligands that preferentially stimulate the ß-arrestin pathway over the G protein-mediated signal transduction route, or vice versa. This phenomenon is referred to as functional selectivity or biased signaling and implies that an antagonist for one pathway may be a full agonist for the other signaling route. Functional selectivity makes it necessary to redefine the functional properties of currently used adenosine receptor ligands and opens possibilities for new and more selective ligands. This review focuses on the current knowledge of functionally selective adenosine receptor ligands and on G protein-independent signaling of adenosine receptors through scaffold proteins.

10.
Bioorg Med Chem ; 19(11): 3384-93, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21570852

ABSTRACT

Small-molecule ligands for the CXCR3 chemokine receptor receive considerable attention as a means to interrogate the roles of CXCR3 in vitro and in vivo and/or to potentially treat various conditions such as inflammatory diseases and cancer. We have synthesized and explored a novel class of small-molecule antagonists for CXCR3. A medium-throughput screen revealed an adamantane-guanidine as a hit. The guanidine unit was replaced by a small quaternary ammonium group, leading to ca. 5-fold increase in affinity. Substitution of the adamantane group by a myrtenyl moiety further increased affinity, while the benzyl group was decorated with an additional (substituted) aryl ring. This led to the identification of several bisaryl-based ligands with CXCR3 affinities of around 100 nM and with the ability to antagonize the functional activity of CXCL10.


Subject(s)
Biphenyl Compounds/chemistry , Polycyclic Compounds/chemistry , Quaternary Ammonium Compounds/chemistry , Receptors, CXCR3/antagonists & inhibitors , Cell Line , Guanidine/chemistry , Humans , Protein Binding , Quaternary Ammonium Compounds/chemical synthesis , Quaternary Ammonium Compounds/pharmacology , Receptors, CXCR3/metabolism , Salts/chemistry , Structure-Activity Relationship
11.
Glia ; 58(12): 1465-76, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20648639

ABSTRACT

Sphingolipids are a class of biologically active lipids that have a role in multiple biological processes including inflammation. Sphingolipids exert their functions by direct signaling or through signaling by their specific receptors. Phosphorylated FTY720 (FTY720P) is a sphingosine 1-phosphate (S1P) analogue that is currently in trial for treatment of multiple sclerosis (MS), which targets all S1P receptors but S1P(2). To date, however, it remains unknown whether FTY720P may exert direct anti-inflammatory effects within the central nervous system (CNS), because data concerning S1P receptor expression and regulation under pathological conditions in the human brain are lacking. To investigate potential regulation of S1P receptors in the human brain during MS, we performed immunohistochemical analysis of S1P receptor 1 and 3 expression in well-characterized MS lesions. A strong increase in S1P receptor 1 and 3 expression on reactive astrocytes was detected in active and chronic inactive MS lesions. In addition, we treated primary cultures of human astrocytes with the proinflammatory cytokine tumor necrosis factor-alpha to identify the regulation of S1P(1/3) on astrocytes under pathological conditions. Importantly, we demonstrate that FTY720P exerts an anti-inflammatory action on human astrocytes by limiting secretion of proinflammatory cytokines. Our data demonstrate that reactive astrocytes in MS lesions and cultured under proinflammatory conditions strongly enhance expression of S1P receptors 1 and 3. Results from this study indicate that astrocytes may act as a yet-unknown target within the CNS for the anti-inflammatory effects observed after FTY720P administration in the treatment of MS.


Subject(s)
Multiple Sclerosis/physiopathology , Receptors, Lysosphingolipid/metabolism , Up-Regulation/physiology , Adult , Aged , Aged, 80 and over , Astrocytes/metabolism , Brain/cytology , Cells, Cultured , Chemokines/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Female , Fingolimod Hydrochloride , Humans , Immunosuppressive Agents/pharmacology , Male , Middle Aged , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/genetics , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors , T-Lymphocytes/metabolism , Up-Regulation/drug effects
12.
Bioorg Med Chem Lett ; 19(8): 2252-7, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19299127

ABSTRACT

A CXCR3 pocket capable of accommodating polycycloaliphatics was explored using a modular synthetic strategy. The systematic studies reveal that the tricyclic 2-adamantane and bicyclic (iso)bornyl group are efficiently recognized by CXCR3.


Subject(s)
Polycyclic Compounds/chemical synthesis , Polycyclic Compounds/metabolism , Receptors, CXCR3/metabolism , Adamantane/chemistry , Amination , Binding Sites/physiology , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Camphanes/chemistry , Cell Line , Humans , Receptors, CXCR3/chemistry , Structure-Activity Relationship
13.
J Pharmacol Exp Ther ; 325(2): 544-55, 2008 May.
Article in English | MEDLINE | ID: mdl-18270317

ABSTRACT

The chemokine receptor CXCR3 is involved in various inflammatory diseases, such as rheumatoid arthritis, multiple sclerosis, psoriasis, and allograft rejection in transplantation patients. The CXCR3 ligands CXCL9, CXCL10, and CXCL11 are expressed at sites of inflammation, and they attract CXCR3-bearing lymphocytes, thus contributing to the inflammatory process. In this study, we characterize five nonpeptidergic compounds of different chemical classes that block the action of CXCL10 and CXCL11 at the human CXCR3, i.e., the 3H-pyrido[2,3-d]pyrimidin-4-one derivatives N-1R-[3-(4-ethoxy-phenyl)-4-oxo-3,4-dihydro-pyrido[2,3-d]pyrimidin-2-yl]-ethyl-N-pyridin-3-ylmethyl-2-(4-fluoro-3-trifluoromethyl-phenyl)-acetamide (VUF10472/NBI-74330) and N-1R-[3-(4-ethoxy-phenyl)-4-oxo-3,4-dihydro-pyrido[2,3-d]pyrimidin-2-yl]-ethyl-N-pyridin-3-ylmethyl-2-(4-trifluoromethoxy-phenyl)-acetamide (VUF10085/AMG-487), the 3H-quinazolin-4-one decanoic acid {1-[3-(4-cyano-phenyl)-4-oxo-3,4-dihydro-quinazolin-2-yl]-ethyl}-(2-dimethylamino-ethyl)-amide (VUF5834), the imidazolium compound 1,3-bis-[2-(3,4-dichloro-phenyl)-2-oxo-ethyl]-3H-imidazol-1-ium bromide (VUF10132), and the quaternary ammonium anilide N,N-dimethyl-N-[4-[[[2-(4-methylphenyl)-6,7-dihydro-5H-benzocyclohepten-8-yl]-carbonyl]amino]benzyl] tetrahydro-2H-pyran-4-aminium chloride (TAK-779). To understand the action of these CXCR3 antagonists in various animal models of disease, the compounds were also tested at rat and mouse CXCR3, as well as at CXCR3 from rhesus macaque, which was cloned and characterized for the first time in this study. Except for TAK-779, all compounds show slightly lower affinity for rodent CXCR3 than for primate CXCR3. In addition, we have characterized the molecular mechanism of action of the various antagonists at the human CXCR3 receptor. All tested compounds act as noncompetitive antagonists at CXCR3. Moreover, this noncompetitive behavior is accompanied by inverse agonistic properties of all five compounds as determined on an identified constitutively active mutant of CXCR3, CXCR3 N3.35A. It is interesting to note that all compounds except TAK-779 act as full inverse agonists at CXCR3 N3.35A. TAK-779 shows weak partial inverse agonism at CXCR3 N3.35A, and it probably has a different mode of interaction with CXCR3 than the other two classes of small-molecule inverse agonists.


Subject(s)
Receptors, CXCR3/antagonists & inhibitors , Animals , COS Cells , Cell Line , Chemokine CXCL10/metabolism , Chemokine CXCL11/metabolism , Chlorocebus aethiops , Humans , Ligands , Macaca mulatta , Mice , Molecular Sequence Data , Rats , Receptors, CXCR3/agonists , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Receptors, Histamine H1/metabolism , Type C Phospholipases/metabolism
14.
Stem Cells Dev ; 23(16): 1959-74, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24708339

ABSTRACT

Mobilization of hematopoietic stem and progenitor cells (HPCs) is induced by treatment with granulocyte-colony stimulating factor, chemotherapy, or irradiation. We observed that these treatments are accompanied by a release of chemotactic activity into the blood. This plasma activity is derived from the bone marrow, liver, and spleen and acts on HPCs via the chemokine receptor CXCR4. A human blood peptide library was used to characterize CXCR4-activating compounds. We identified CXCL12[22-88] and N-terminally truncated variants CXCL12[24-88], CXCL12[25-88], CXCL12[27-88], and CXCL12[29-88]. Only CXCL12[22-88] could effectively bind to CXCR4 and induce intracellular calcium flux and chemotactic migration of HPCs. CXCL12[25-88] and CXCL12[27-88] revealed neither agonistic nor antagonistic activities in vitro, whereas CXCL12[29-88] inhibited CXCL12[22-88]-induced chemotactic migration. Since binding to glycosaminoglycans (GAG) modulates the function of CXCL12, binding to heparin was analyzed. Surface plasmon resonance kinetic analysis showed that N-terminal truncation of Arg22-Pro23 increased the dissociation constant KD by one log10 stage ([22-88]: KD: 5.4 ± 2.6 µM; [24-88]: KD: 54 ± 22.4 µM). Further truncation of the N-terminus decreased the KD ([25-88] KD: 30 ± 4.8 µM; [27-88] KD: 23 ± 1.6 µM; [29-88] KD: 19 ± 5.4 µM), indicating increasing competition for heparin binding. Systemic in vivo application of CXCL12[22-88] as well as CXCL12[27-88] or CXCL12[29-88] induced a significant mobilization of HPCs in mice. Our findings indicate that plasma-derived CXCL12 variants may contribute to the regulation of HPC mobilization by modulating the binding of CXCL12[22-88] to GAGs rather than blocking the CXCR4 receptor and, therefore, may have a contributing role in HPC mobilization.


Subject(s)
Chemokine CXCL12/blood , Chemotaxis , Hematopoietic Stem Cells/physiology , Animals , Blood Platelets/metabolism , Calcium Signaling , Cells, Cultured , Hematopoietic Stem Cell Transplantation , Humans , Leukocytes/metabolism , Mice, Inbred DBA , Protein Binding , Protein Processing, Post-Translational , Proteolysis
15.
Eur J Pharmacol ; 667(1-3): 105-12, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21663738

ABSTRACT

The sphingosine-1-phosphate type 1 (S1P(1)) receptor is a new target in the treatment of auto-immune diseases as evidenced by the recent approval of FTY720 (Fingolimod). The ligand-binding pocket of the S1P(1) receptor has been generally characterised but detailed insight into ligand-specific differences is still lacking. The aim of the current study is to determine differences in ligand-induced S1P(1) receptor activation using an in silico guided site-directed mutagenesis strategy. S1P(1) mutant receptors (modifications of residues Y98(2.57), R120(3.28), F125(3.33)) were probed with a chemically diverse set of S1P(1) agonists (S1P, dihydro-S1P (dhS1P), R-, S- and racemic FTY720-P, VPC24191, SEW2871). Mutation of the R(3.28) residue generally results in a reduction of the potency of all ligands although the synthetic ligands including FTY720-P are less sensitive to these mutations. The Y(2.57)F mutation does not affect the potency of any of the ligands tested, but for all ligands except FTY720-P a significant decrease in potency is observed at the Y(2.57)A mutant. The F(3.33)A mutation significantly decreased the potency of FTY720-P and is detrimental for SEW2871 and VPC24191. The non-aromatic endogenous ligands S1P and dhS1P are less affected by this mutation. Our in silico guided mutagenesis studies identified new molecular determinants of ligand-induced S1P(1) receptor activation: 1) the flexibility of the polar head of the agonist to maintain a tight H-bond network with R(3.28) and 2) the ability of the agonist to make aromatic π-stacking interactions with F(3.33). Interestingly, FTY720-P has both chemical properties and is the only ligand that can efficiently activate the Y(2.57)A mutant.


Subject(s)
Mutation , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/genetics , Animals , Computational Biology , Drug Design , Humans , Ligands , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Conformation , Protein Stability , Receptors, Lysosphingolipid/chemistry , Receptors, Lysosphingolipid/metabolism , Sphingosine-1-Phosphate Receptors , Stereoisomerism
16.
Biochem Pharmacol ; 82(6): 658-68, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21718691

ABSTRACT

Allosteric modulators for the G(i)-coupled A(3) adenosine receptor (AR) are of considerable interest as therapeutic agents and as pharmacological tools to probe various signaling pathways. In this study, we initially characterized the effects of several imidazoquinolinamine allosteric modulators (LUF5999, LUF6000 and LUF6001) on the human A(3) AR stably expressed in CHO cells using a cyclic AMP functional assay. These modulators were found to affect efficacy and potency of the agonist Cl-IB-MECA differently. LUF5999 (2-cyclobutyl derivative) enhanced efficacy but decreased potency. LUF6000 (2-cyclohexyl derivative) enhanced efficacy without affecting potency. LUF6001 (2-H derivative) decreased both efficacy and potency. We further compared the agonist enhancing effects of LUF6000 in several other A(3) AR-mediated events. It was shown that although LUF6000 behaved somewhat differently in various signaling pathways, it was more effective in enhancing the effects of low-efficacy than of high-efficacy agonists. In an assay of cyclic AMP accumulation, LUF6000 enhanced the efficacy of all agonists examined, but in the membrane hyperpolarization assay, it only enhanced the efficacy of partial agonists. In calcium mobilization, LUF6000 did not affect the efficacy of the full agonist NECA but was able to switch the nucleoside antagonist MRS542 into a partial agonist. In translocation of ß-arrestin2, the agonist-enhancing effect LUF6000 was not pronounced. In an assay of ERK1/2 phosphorylation LUF6000 did not show any effect on the efficacy of Cl-IB-MECA. The differential effects of LUF6000 on the efficacy and potency of the agonist Cl-IB-MECA in various signaling pathway were interpreted quantitatively using a mathematical model.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Aminoquinolines/pharmacology , Imidazoles/pharmacology , Receptor, Adenosine A3/metabolism , Adenosine A3 Receptor Agonists/chemistry , Allosteric Regulation/drug effects , Aminoquinolines/chemistry , Animals , Arrestins/metabolism , CHO Cells , Calcium/metabolism , Computer Simulation , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Data Interpretation, Statistical , Dose-Response Relationship, Drug , Humans , Imidazoles/chemistry , Membrane Potentials/drug effects , Molecular Structure , Receptor, Adenosine A3/genetics , Transfection , beta-Arrestins
17.
Mol Cells ; 29(2): 99-104, 2010 Feb 28.
Article in English | MEDLINE | ID: mdl-20127285

ABSTRACT

Regulatory processes including receptor phosphorylation and intracellular trafficking, also referred to as receptor internalization, are important processes to terminate G protein-coupled receptor (GPCR) signaling. Compelling evidence now indicates that internalization of a receptor is not necessarily the endpoint of signaling, but can also be the beginning of the activation of intracellular signaling pathways. Sphingosine-1-phosphate (S1P) receptors, which are activated by the endogenous phospholipid S1P, belong to the family of GPCRs. Interestingly, there is evidence indicating differential intracellular trafficking of one of the S1P receptor subtypes, the S1P1 receptor, upon agonist activation by either S1P or the synthetic agonist FTY720-P. Moreover, the differential effect of FTY720-P on S1P1 receptor regulation has been suggested to be the mechanism of action of this drug, which is now in Phase III clinical trials for the treatment of multiple sclerosis. It is thus of importance to get a good insight into the regulation of S1P receptors. This review therefore gives a detailed overview about the current state of knowledge on S1P receptor internalization and its functional implications, including some data on nuclear signaling of S1P receptors.


Subject(s)
Intracellular Space/metabolism , Receptors, Lysosphingolipid/metabolism , Amino Acid Sequence , Animals , Endocytosis , Humans , Ligands , Molecular Sequence Data , Protein Processing, Post-Translational , Protein Transport , Receptors, Lysosphingolipid/chemistry
18.
ChemMedChem ; 3(6): 861-72, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18442035

ABSTRACT

The CXCR3 chemokine receptor was first discovered in 1996 and has been shown to play an important role in several diseases, most of which are related to inflammation. This review describes in detail the development of small CXCR3 ligands and their therapeutic potential. Classes of CXCR3 antagonists with strikingly variable core structures have emerged. Some of these compounds have confirmed the beneficial role of CXCR3 antagonism in animal models of disease. One of the compounds, AMG487, progressed to Phase II clinical trials but has been withdrawn because of lack of efficacy. New antagonist classes are being developed to reveal the full therapeutic potential of CXCR3.


Subject(s)
Organic Chemicals/pharmacology , Receptors, CXCR3/agonists , Receptors, CXCR3/antagonists & inhibitors , Animals , Antibodies/chemistry , Antibodies/pharmacology , Humans , Ligands , Molecular Structure , Organic Chemicals/chemistry , Peptides/chemistry , Peptides/pharmacology , Proteins/chemistry , Proteins/pharmacology , Structure-Activity Relationship
19.
Arch Pharm (Weinheim) ; 340(6): 281-91, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17562560

ABSTRACT

CXC chemokine receptor-3 (CXCR3) is a G-protein coupled receptor (GPCR) predominantly expressed on activated T lymphocytes that promote Th1 responses. Previously, we described the 3H-quinazolin-4-one containing VUF 5834 (decanoic acid {1-[3-(4-cyano-phenyl)-4-oxo-3,4-dihydro-quinazolin-2-yl]-ethyl}-(2-dimethylamino-ethyl)-amide) as a small-molecule CXCR3 antagonist with submicromolar affinity and as a lead structure for the development of CXCR3 antagonists. More recently, the related 3H-pyrido[2,3-d]pyrimidin-4-one compounds AMG 487 and NBI-74330 have been reported as nanomolar CXCR3 antagonists and these ligands are currently under clinical investigation. The aim of this study is to link the structure-activity relationship (SAR) of the previously published class of 3H-quinazolin-4-one containing CXCR3 ligands with these novel clinical candidates. From the modification of the lead structure VUF 5834 emerged the importance of the (4-fluoro-3-(trifluoromethyl)phenyl)acetyl and the 3-methylen-pyridine as substituents to improve the affinity at the human CXCR3 receptor, whereas other features are less important. The described molecules serve as tool to investigate the role of the CXCR3 receptor in various inflammatory conditions.


Subject(s)
Pyridines/chemical synthesis , Pyrimidinones/chemical synthesis , Quinazolinones/chemical synthesis , Receptors, Chemokine/antagonists & inhibitors , Structure-Activity Relationship , Cell Line , Humans , Molecular Structure , Pyridines/pharmacology , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Quinazolinones/chemistry , Quinazolinones/pharmacology , Radioligand Assay , Receptors, CXCR3 , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism
20.
Mol Pharmacol ; 69(3): 888-98, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16332987

ABSTRACT

The human herpes virus 6 (HHV-6)-encoded chemokine receptor U51 constitutively activates phospholipase C (PLC) and inhibits cAMP-responsive element (CRE)-mediated gene transcription via the activation of G(q/11) proteins. Yet, chemokines known to bind U51 differentially regulate U51 coupling to G proteins. CCL5/RANTES induced pertussis toxin (PTX)-insensitive increases in PLC activity and changes in intracellular free calcium concentration ([Ca2+]i), whereas both CCL2/MCP-1 and CCL11/eotaxin failed to stimulate PLC activity or increase [Ca2+]i. In contrast, all three chemokines counteracted the effects of U51 on CRE activity via the activation of PTX-sensitive G(i/o) proteins. For each of the tested chemokines, coexpression of U51 with a variety of G alpha subunits, however, revealed a distinct profile for preferred G-protein coupling, which could be shifted by modulation of the relative expression of G proteins. These findings are consistent with a chemokine-selective trafficking of receptor stimulus to distinct G proteins and suggest that the constitutive activity of U51 and the chemokine-induced signaling involve different active states of the receptor. By virtue of its ability to constitutively activate signaling pathways, its G-protein promiscuity, and the chemokine-directed trafficking of receptor stimulus, U51 can be considered a sensitive and versatile virally encoded signaling device, potentially of importance in HHV-6-related pathologies.


Subject(s)
Chemokines/pharmacology , GTP-Binding Protein alpha Subunits/metabolism , Gene Expression Regulation , Receptors, Chemokine/agonists , Receptors, Chemokine/metabolism , Receptors, Virus/agonists , Receptors, Virus/metabolism , Animals , COS Cells , Chemokine CCL11 , Chemokine CCL2/pharmacology , Chemokine CCL5 , Chemokines, CC/pharmacology , Chlorocebus aethiops , Cyclic AMP/metabolism , Genes, Reporter , Herpesvirus 6, Human/pathogenicity , Humans , Ligands , Receptors, Chemokine/genetics , Receptors, Virus/genetics , Response Elements , Signal Transduction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL