Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
MAbs ; 16(1): 2381891, 2024.
Article in English | MEDLINE | ID: mdl-39041287

ABSTRACT

Novel engineered IL-2 agonists strive to increase the therapeutic window of aldesleukin (human IL-2) by increasing selectivity toward effector over regulatory T cells and reducing dose-limiting toxicities. Here we describe ANV419, an IL-2/anti-IL2 antibody fusion protein designed for selective IL-2 receptor ßγ (IL-2 Rßγ) activation by sterically hindering IL-2 from binding to IL-2 Rα. The fusion protein has an IL-2 connected to the light chain complementarity-determining region (CDR) domain of a humanized antibody that binds to IL-2 at the same epitope as IL-2 Rα. Optimization of the selectivity and pharmacological properties led to the selection of ANV419. ANV419 preferentially expands CD8+ T cells and natural killer (NK) cells over Tregs and can be safely administered at doses that elicit strong pharmacodynamic effects and efficacy in mouse tumor models. Its anti-tumor efficacy was enhanced when combined with programmed cell death protein 1 (PD-1) or cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) checkpoint inhibitors. ANV419 also enhances the NK cell killing capacity and increases tumor growth inhibition when used alongside trastuzumab in a Her-2+ xenograft mouse model. In cynomolgus monkeys, the estimated half-life of ANV419 is 24 h, and doses that induced sustained expansion of effector cells were well tolerated without the severe toxicities typically observed with high-dose IL-2. These data support the clinical development of ANV419 in solid tumors and hematological malignancies as monotherapy and in combination with checkpoint inhibitors or agents that induce antibody-dependent cellular cytotoxicity. ANV419 is currently in Phase 1/2 clinical development and may provide cancer patients with a wider therapeutic window than aldesleukin.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-2 , Killer Cells, Natural , Recombinant Fusion Proteins , Animals , Killer Cells, Natural/immunology , Humans , Interleukin-2/immunology , CD8-Positive T-Lymphocytes/immunology , Mice , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Immunotherapy/methods , Macaca fascicularis , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Cell Line, Tumor , Female
2.
J Clin Invest ; 133(14)2023 07 17.
Article in English | MEDLINE | ID: mdl-37227784

ABSTRACT

Bulk analysis of renal allograft biopsies (rBx) identified RNA transcripts associated with acute cellular rejection (ACR); however, these lacked cellular context critical to mechanistic understanding of how rejection occurs despite immunosuppression (IS). We performed combined single-cell RNA transcriptomic and TCR-α/ß sequencing on rBx from patients with ACR under differing IS drugs: tacrolimus, iscalimab, and belatacept. We found distinct CD8+ T cell phenotypes (e.g., effector, memory, exhausted) depending upon IS type, particularly within expanded CD8+ T cell clonotypes (CD8EXP). Gene expression of CD8EXP identified therapeutic targets that were influenced by IS type. TCR analysis revealed a highly restricted number of CD8EXP, independent of HLA mismatch or IS type. Subcloning of TCR-α/ß cDNAs from CD8EXP into Jurkat 76 cells (TCR-/-) conferred alloreactivity by mixed lymphocyte reaction. Analysis of sequential rBx samples revealed persistence of CD8EXP that decreased, but were not eliminated, after successful antirejection therapy. In contrast, CD8EXP were maintained in treatment-refractory rejection. Finally, most rBx-derived CD8EXP were also observed in matching urine samples, providing precedent for using urine-derived CD8EXP as a surrogate for those found in the rejecting allograft. Overall, our data define the clonal CD8+ T cell response to ACR, paving the next steps for improving detection, assessment, and treatment of rejection.


Subject(s)
Kidney Transplantation , Transcriptome , Receptors, Antigen, T-Cell, alpha-beta/genetics , RNA , Allografts , Graft Rejection/genetics
3.
bioRxiv ; 2023 Feb 13.
Article in English | MEDLINE | ID: mdl-36798151

ABSTRACT

Bulk analysis of renal allograft biopsies (rBx) identified RNA transcripts associated with acute cellular rejection (ACR); however, these lacked cellular context critical to mechanistic understanding. We performed combined single cell RNA transcriptomic and TCRα/ß sequencing on rBx from patients with ACR under differing immunosuppression (IS): tacrolimus, iscalimab, and belatacept. TCR analysis revealed a highly restricted CD8 + T cell clonal expansion (CD8 EXP ), independent of HLA mismatch or IS type. Subcloning of TCRα/ß cDNAs from CD8 EXP into Jurkat76 cells (TCR -/- ) conferred alloreactivity by mixed lymphocyte reaction. scRNAseq analysis of CD8 EXP revealed effector, memory, and exhausted phenotypes that were influenced by IS type. Successful anti-rejection treatment decreased, but did not eliminate, CD8 EXP , while CD8 EXP were maintained during treatment-refractory rejection. Finally, most rBx-derived CD8 EXP were also observed in matching urine samples. Overall, our data define the clonal CD8 + T cell response to ACR, providing novel insights to improve detection, assessment, and treatment of rejection.

4.
Comput Struct Biotechnol J ; 20: 4717-4732, 2022.
Article in English | MEDLINE | ID: mdl-36147669

ABSTRACT

We developed a bioinformatics-led substrate discovery workflow to expand the known substrate repertoire of MALT1. Our approach, termed GO-2-Substrates, integrates protein function information, including GO terms from known substrates, with protein sequences to rank substrate candidates by similarity. We applied GO-2-Substrates to MALT1, a paracaspase and master regulator of NF-κB signalling in adaptive immune responses. With only 12 known substrates, the evolutionarily conserved paracaspase functions and phenotypes of Malt1 -/- mice strongly implicate the existence of undiscovered substrates. We tested the ranked predictions from GO-2-Substrates of new MALT1 human substrates by co-expression of candidates transfected with the oncogenic constitutively active cIAP2-MALT1 fusion protein or CARD11/BCL10/MALT1 active signalosome. We identified seven new MALT1 substrates by the co-transfection screen: TANK, TAB3, CASP10, ZC3H12D, ZC3H12B, CILK1 and ILDR2. Using catalytically inactive cIAP2-MALT1 (Cys464Ala), a MALT1 inhibitor, MLT-748, and noncleavable P1-Arg to Ala mutant versions of each substrate in dual transfections, we validated the seven new substrates in vitro. We confirmed the cleavage of endogenous TANK and the RNase ZC3H12D in B cells by Western blotting and mining TAILS N-terminomics datasets, where we also uncovered evidence for these and 12 other candidate substrates by endogenous MALT1. Thus, protein function information improves substrate predictions. The new substrates and other high-ranked MALT1 candidate substrates should open new biological frontiers for further validation and exploration of the function of MALT1 within and beyond NF-κB regulation.

5.
J Comp Eff Res ; 11(11): 789-803, 2022 08.
Article in English | MEDLINE | ID: mdl-35642553

ABSTRACT

Aim: This research compared patient and physician perceptions of quality of life (QoL) in C0-4 chronic venous disease (CVD). Methods: Qualitative standardized phone interviews were conducted with 100 patients and 60 specialists from Brazil, China, the Czech Republic, Italy and Russia. Results: In addition to the impact of physical symptoms on QoL, patient interviews revealed a high aesthetic and emotional burden of C0-4 CVD that contributes to social isolation and affects relationships. Physicians were aware of the physical impact but underestimated the other implications of CVD on their patients' QoL. Conclusion: Healthcare professional awareness of the overall impact of CVD on QoL needs improvement. All aspects of QoL should be assessed in order to manage CVD effectively.


Chronic venous disease (CVD) is a progressive condition that occurs when the functioning of the veins, which are blood vessels that move blood back to the heart, is compromised, leading to swelling and other physical changes in the legs. CVD can be debilitating to those who suffer from it, so the authors surveyed 100 people with CVD as well as 60 physicians who treat them to understand more about the impact of this disease. The authors found that CVD affects people not only physically but also aesthetically and emotionally, which impacts on relationships and leads to social isolation. Physicians are aware of the physical impact of CVD but often underestimate other burdens their patients might experience, so the authors suggest that physicians consult their patients on these aspects when treating them.


Subject(s)
Cardiovascular Diseases , Physicians , Chronic Disease , Humans , Lower Extremity , Quality of Life , Surveys and Questionnaires
6.
J Med Chem ; 63(23): 14594-14608, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33216547

ABSTRACT

The paracaspase MALT1 has gained increasing interest as a target for the treatment of subsets of lymphomas as well as autoimmune diseases, and there is a need for suitable compounds to explore the therapeutic potential of this target. Here, we report the optimization of the in vivo potency of pyrazolopyrimidines, a class of highly selective allosteric MALT1 inhibitors. High doses of the initial lead compound led to tumor stasis in an activated B-cell-like (ABC) diffuse large B-cell lymphoma (DLBCL) xenograft model, but this compound suffered from a short in vivo half-life and suboptimal potency in whole blood. Guided by metabolism studies, we identified compounds with reduced metabolic clearance and increased in vivo half-life. In the second optimization step, masking one of the hydrogen-bond donors of the central urea moiety through an intramolecular interaction led to improved potency in whole blood. This was associated with improved in vivo potency in a mechanistic model of B cell activation. The optimized compound led to tumor regression in a CARD11 mutant ABC-DLBCL lymphoma xenograft model.


Subject(s)
Blood/metabolism , Caspase Inhibitors/therapeutic use , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Urea/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Caspase Inhibitors/chemical synthesis , Caspase Inhibitors/metabolism , Caspase Inhibitors/pharmacokinetics , Cell Line, Tumor , Female , Half-Life , Humans , Mice, Inbred BALB C , Mice, SCID , Microsomes, Liver/metabolism , Neoplasms/drug therapy , Pyrazoles/chemical synthesis , Pyrazoles/metabolism , Pyrazoles/pharmacokinetics , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Pyrimidines/pharmacokinetics , Rats, Sprague-Dawley , Sheep , Urea/chemical synthesis , Urea/metabolism , Urea/pharmacokinetics , Xenograft Model Antitumor Assays
7.
J Med Chem ; 63(23): 14576-14593, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33252239

ABSTRACT

MALT1 plays a central role in immune cell activation by transducing NF-κB signaling, and its proteolytic activity represents a key node for therapeutic intervention. Two cycles of scaffold morphing of a high-throughput biochemical screening hit resulted in the discovery of MLT-231, which enabled the successful pharmacological validation of MALT1 allosteric inhibition in preclinical models of humoral immune responses and B-cell lymphomas. Herein, we report the structural activity relationships (SARs) and analysis of the physicochemical properties of a pyrazolopyrimidine-derived compound series. In human T-cells and B-cell lymphoma lines, MLT-231 potently and selectively inhibits the proteolytic activity of MALT1 in NF-κB-dependent assays. Both in vitro and in vivo profiling of MLT-231 support further optimization of this in vivo tool compound toward preclinical characterization.


Subject(s)
Caspase Inhibitors/therapeutic use , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Neoplasms/drug therapy , Urea/analogs & derivatives , Urea/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Caspase Inhibitors/chemical synthesis , Caspase Inhibitors/pharmacology , Drug Discovery , Female , Humans , Immunity, Humoral/drug effects , Male , Mice, Inbred BALB C , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Rats, Sprague-Dawley , Structure-Activity Relationship , T-Lymphocytes/drug effects , Urea/pharmacology , Xenograft Model Antitumor Assays
8.
J Comp Eff Res ; 9(17): 1205-1218, 2020 12.
Article in English | MEDLINE | ID: mdl-33079592

ABSTRACT

Aim: This study assessed the characteristics of individuals with chronic venous disease (CVD) and their treatment pathways. Materials & methods: A web-based survey enrolled representative populations of adults from Brazil, Czech Republic, France, Hungary, Italy, Romania, Russia and Spain, and identified those self-reporting CVD. Results: A total of 22% of respondents had signs/symptoms of CVD. Individuals with CVD were generally older, female and obese, and had more comorbidities than the general population. Common initial symptoms were tiredness, heaviness, pain, swelling in legs and night cramps. Participants waited ∼1 year before seeking treatment but most did not initially consult a physician; those who did tended to have more severe disease. Conclusion: One in five adults had CVD, but most did not seek a physician's help.


Subject(s)
Internet , Varicose Veins/epidemiology , Venous Insufficiency/epidemiology , Venous Insufficiency/therapy , Adolescent , Adult , Aged , Chronic Disease/epidemiology , Europe/epidemiology , Female , Humans , Male , Middle Aged , Prevalence , Risk Factors , Surveys and Questionnaires , Varicose Veins/diagnosis , Varicose Veins/therapy , Venous Insufficiency/diagnosis
9.
J Comp Eff Res ; 9(17): 1219-1232, 2020 12.
Article in English | MEDLINE | ID: mdl-33079605

ABSTRACT

Background: This international study assessed the characteristics and treatment of individuals with hemorrhoids. Materials & methods: Online survey among nationally representative populations of adults from Brazil, Czech Republic, France, Hungary, Italy, Romania, Russia and Spain, that identified participants who self-reported having hemorrhoidal disease. Results: Hemorrhoid prevalence was 11% (1725/16015); most respondents had low-severity disease (71%). Compared with the general population, participants with hemorrhoidal disease had more comorbidities (mean 3.1 vs 1.3) and included more women who had been pregnant (81 vs 68%). Common initial signs/symptoms were pain (60%), bleeding (47%) and discomfort (43%). Hemorrhoid respondents who consulted a physician were more likely to undergo interventions and take medications. Conclusion: The prevalence of hemorrhoidal disease in the adult population is 11%, mostly low-severity disease.


Subject(s)
Hemorrhoids/epidemiology , Hemorrhoids/therapy , Internet , Adolescent , Adult , Aged , Aged, 80 and over , Europe/epidemiology , Female , Hemorrhoids/diagnosis , Humans , Middle Aged , Prevalence , Surveys and Questionnaires , Young Adult
10.
Arthritis Rheumatol ; 72(6): 919-930, 2020 06.
Article in English | MEDLINE | ID: mdl-31943941

ABSTRACT

OBJECTIVE: Fcγ receptors (FcγR) play important roles in both protective and pathogenic immune responses. The assembly of the CBM signalosome encompassing caspase recruitment domain-containing protein 9, B cell CLL/lymphoma 10, and mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT-1) is required for optimal FcγR-induced canonical NF-κB activation and proinflammatory cytokine release. This study was undertaken to clarify the relevance of MALT-1 protease activity in FcγR-driven events and evaluate the therapeutic potential of selective MALT-1 protease inhibitors in FcγR-mediated diseases. METHODS: Using genetic and pharmacologic disruption of MALT-1 scaffolding and enzymatic activity, we assessed the relevance of MALT-1 function in murine and human primary myeloid cells upon stimulation with immune complexes (ICs) and in murine models of autoantibody-driven arthritis and immune thrombocytopenic purpura (ITP). RESULTS: MALT-1 protease function is essential for optimal FcγR-induced production of proinflammatory cytokines by various murine and human myeloid cells stimulated with ICs. In contrast, MALT-1 protease inhibition did not affect the Syk-dependent, FcγR-mediated production of reactive oxygen species or leukotriene B4 . Notably, pharmacologic MALT-1 protease inhibition in vivo reduced joint inflammation in the murine K/BxN serum-induced arthritis model (mean area under the curve for paw swelling of 45.42% versus 100% in control mice; P = 0.0007) but did not affect platelet depletion in a passive model of ITP. CONCLUSION: Our findings indicate a specific contribution of MALT-1 protease activity to FcγR-mediated events and suggest that MALT-1 protease inhibitors have therapeutic potential in a subset of FcγR-driven inflammatory disorders.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/immunology , Receptors, IgG/immunology , Animals , Antigen-Antibody Complex/metabolism , Blood Platelets/metabolism , Cytokines/immunology , Disease Models, Animal , Humans , Mice , Myeloid Cells/metabolism
11.
Bioorg Med Chem Lett ; 28(12): 2153-2158, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29759726

ABSTRACT

Starting from a weak screening hit, potent and selective inhibitors of the MALT1 protease function were elaborated. Advanced compounds displayed high potency in biochemical and cellular assays. Compounds showed activity in a mechanistic Jurkat T cell activation assay as well as in the B-cell lymphoma line OCI-Ly3, which suggests potential use of MALT1 inhibitors in the treatment of autoimmune diseases as well as B-cell lymphomas with a dysregulated NF-κB pathway. Initially, rat pharmacokinetic properties of this compound series were dominated by very high clearance which could be linked to amide cleavage. Using a rat hepatocyte assay a good in vitro-in vivo correlation could be established which led to the identification of compounds with improved PK properties.


Subject(s)
Antineoplastic Agents/pharmacology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Piperidines/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Hepatocytes/drug effects , Humans , Jurkat Cells , Microsomes/drug effects , Molecular Structure , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Piperidines/chemical synthesis , Piperidines/chemistry , Proteolysis/drug effects , Rats , Structure-Activity Relationship
12.
Immunol Cell Biol ; 96(1): 81-99, 2018 01.
Article in English | MEDLINE | ID: mdl-29359407

ABSTRACT

Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) is essential for immune responses triggered by antigen receptors but the contribution of its paracaspase activity is not fully understood. Here, we studied how MALT1 proteolytic function regulates T-cell activation and fate after engagement of the T-cell receptor pathway. We show that MLT-827, a potent and selective MALT1 paracaspase inhibitor, does not prevent the initial phase of T-cell activation, in contrast to the pan-protein kinase C inhibitor AEB071. However, MLT-827 strongly impacted cell expansion after activation. We demonstrate this is the consequence of profound inhibition of IL-2 production as well as reduced expression of the IL-2 receptor alpha subunit (CD25), resulting from defective canonical NF-κB activation and accelerated mRNA turnover mechanisms. Accordingly, MLT-827 revealed a unique transcriptional fingerprint of MALT1 protease activity, providing evidence for broad control of T-cell signaling pathways. Altogether, this first report with a potent and selective inhibitor elucidates how MALT1 paracaspase activity integrates several T-cell activation pathways and indirectly controls gamma-chain receptor dependent survival, to impact on T-cell expansion.


Subject(s)
Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , NF-kappa B/metabolism , T-Lymphocytes/immunology , Cell Proliferation , Cell Survival , Cells, Cultured , Gene Expression Regulation , Humans , Immunomodulation , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Proteolysis , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
13.
PLoS One ; 12(1): e0169026, 2017.
Article in English | MEDLINE | ID: mdl-28052131

ABSTRACT

The paracaspase MALT1 has arginine-directed proteolytic activity triggered by engagement of immune receptors. Recruitment of MALT1 into activation complexes is required for MALT1 proteolytic function. Here, co-expression of MALT1 in HEK293 cells, either with activated CARD11 and BCL10 or with TRAF6, was used to explore the mechanism of MALT1 activation at the molecular level. This work identified a prominent self-cleavage site of MALT1 isoform A (MALT1A) at R781 (R770 in MALT1B) and revealed that TRAF6 can activate MALT1 independently of the CBM. Intramolecular cleavage at R781/R770 removes a C-terminal TRAF6-binding site in both MALT1 isoforms, leaving MALT1B devoid of the two key interaction sites with TRAF6. A previously identified auto-proteolysis site of MALT1 at R149 leads to deletion of the death-domain, thereby abolishing interaction with BCL10. By using MALT1 isoforms and cleaved fragments thereof, as well as TRAF6 WT and mutant forms, this work shows that TRAF6 induces N-terminal auto-proteolytic cleavage of MALT1 at R149 and accelerates MALT1 protein turnover. The MALT1 fragment generated by N-terminal self-cleavage at R149 was labile and displayed enhanced signaling properties that required an intact K644 residue, previously shown to be a site for mono-ubiquitination of MALT1. Conversely, C-terminal self-cleavage at R781/R770 hampered the ability for self-cleavage at R149 and stabilized MALT1 by hindering interaction with TRAF6. C-terminal self-cleavage had limited impact on MALT1A but severely reduced MALT1B proteolytic and signaling functions. It also abrogated NF-κB activation by N-terminally cleaved MALT1A. Altogether, this study provides further insights into mechanisms that regulate the scaffolding and activation cycle of MALT1. It also emphasizes the reduced functional capacity of MALT1B as compared to MALT1A.


Subject(s)
Caspases/metabolism , Neoplasm Proteins/metabolism , Protein Isoforms/metabolism , T-Lymphocytes/metabolism , TNF Receptor-Associated Factor 6/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , B-Cell CLL-Lymphoma 10 Protein , Blotting, Western , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Caspases/genetics , Cell Line , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , HEK293 Cells , Humans , Immunoblotting , Jurkat Cells , Lymphocytes/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Mutagenesis , Neoplasm Proteins/genetics , Protein Isoforms/genetics , Signal Transduction/genetics , Signal Transduction/physiology , TNF Receptor-Associated Factor 6/genetics , Ubiquitination/genetics , Ubiquitination/physiology
14.
Sci Transl Med ; 8(367): 367ra166, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27903862

ABSTRACT

Interleukin-2 (IL-2) immunotherapy is an attractive approach in treating advanced cancer. However, by binding to its IL-2 receptor α (CD25) subunit, IL-2 exerts unwanted effects, including stimulation of immunosuppressive regulatory T cells (Tregs) and contribution to vascular leak syndrome. We used a rational approach to develop a monoclonal antibody to human IL-2, termed NARA1, which acts as a high-affinity CD25 mimic, thereby minimizing association of IL-2 with CD25. The structure of the IL-2-NARA1 complex revealed that NARA1 occupies the CD25 epitope of IL-2 and precisely overlaps with CD25. Association of NARA1 with IL-2 occurs with 10-fold higher affinity compared to CD25 and forms IL-2/NARA1 complexes, which, in vivo, preferentially stimulate CD8+ T cells while disfavoring CD25+ Tregs and improving the benefit-to-adverse effect ratio of IL-2. In two transplantable and one spontaneous metastatic melanoma model, IL-2/NARA1 complex immunotherapy resulted in efficient expansion of tumor-specific and polyclonal CD8+ T cells. These CD8+ T cells showed robust interferon-γ production and expressed low levels of exhaustion markers programmed cell death protein-1, lymphocyte activation gene-3, and T cell immunoglobulin and mucin domain-3. These effects resulted in potent anticancer immune responses and prolonged survival in the tumor models. Collectively, our data demonstrate that NARA1 acts as a CD25-mimobody that confers selectivity and increased potency to IL-2 and warrant further assessment of NARA1 as a therapeutic.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunotherapy/methods , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2/antagonists & inhibitors , Neoplasms/therapy , Animals , Binding Sites , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , Epitopes/chemistry , Gene Silencing , Humans , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Protein Conformation , Recombination, Genetic , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Regulatory/immunology
15.
Nat Commun ; 6: 8777, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26525107

ABSTRACT

Antigen receptor signalling activates the canonical NF-κB pathway via the CARD11/BCL10/MALT1 (CBM) signalosome involving key, yet ill-defined roles for linear ubiquitination. The paracaspase MALT1 cleaves and removes negative checkpoint proteins, amplifying lymphocyte responses in NF-κB activation and in B-cell lymphoma subtypes. To identify new human MALT1 substrates, we compare B cells from the only known living MALT1(mut/mut) patient with healthy MALT1(+/mut) family members using 10-plex Tandem Mass Tag TAILS N-terminal peptide proteomics. We identify HOIL1 of the linear ubiquitin chain assembly complex as a novel MALT1 substrate. We show linear ubiquitination at B-cell receptor microclusters and signalosomes. Late in the NF-κB activation cycle HOIL1 cleavage transiently reduces linear ubiquitination, including of NEMO and RIP1, dampening NF-κB activation and preventing reactivation. By regulating linear ubiquitination, MALT1 is both a positive and negative pleiotropic regulator of the human canonical NF-κB pathway-first promoting activation via the CBM--then triggering HOIL1-dependent negative-feedback termination, preventing reactivation.


Subject(s)
Caspases/genetics , Immunologic Deficiency Syndromes/genetics , Lymphocytes/immunology , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Ubiquitin-Protein Ligases/metabolism , Adolescent , Adult , Animals , Antigen-Presenting Cells , B-Lymphocytes/immunology , Caspases/immunology , Caspases/metabolism , Family , Female , Fluorescent Antibody Technique , GTPase-Activating Proteins/metabolism , Gene Knock-In Techniques , Humans , I-kappa B Kinase/metabolism , Immunoblotting , Immunologic Deficiency Syndromes/immunology , Immunoprecipitation , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Leukocytes, Mononuclear , Male , Mass Spectrometry , Mice , Microscopy, Confocal , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Mutation , NF-kappa B/immunology , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , Palatine Tonsil , Proteomics , RNA-Binding Proteins/metabolism , T-Lymphocytes/immunology , Tandem Mass Spectrometry , Transcription Factors , Ubiquitination/immunology
16.
J Immunol ; 194(8): 3723-34, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25762782

ABSTRACT

The paracaspase MALT1 plays an important role in immune receptor-driven signaling pathways leading to NF-κB activation. MALT1 promotes signaling by acting as a scaffold, recruiting downstream signaling proteins, as well as by proteolytic cleavage of multiple substrates. However, the relative contributions of these two different activities to T and B cell function are not well understood. To investigate how MALT1 proteolytic activity contributes to overall immune cell regulation, we generated MALT1 protease-deficient mice (Malt1(PD/PD)) and compared their phenotype with that of MALT1 knockout animals (Malt1(-/-)). Malt1(PD/PD) mice displayed defects in multiple cell types including marginal zone B cells, B1 B cells, IL-10-producing B cells, regulatory T cells, and mature T and B cells. In general, immune defects were more pronounced in Malt1(-/-) animals. Both mouse lines showed abrogated B cell responses upon immunization with T-dependent and T-independent Ags. In vitro, inactivation of MALT1 protease activity caused reduced stimulation-induced T cell proliferation, impaired IL-2 and TNF-α production, as well as defective Th17 differentiation. Consequently, Malt1(PD/PD) mice were protected in a Th17-dependent experimental autoimmune encephalomyelitis model. Surprisingly, Malt1(PD/PD) animals developed a multiorgan inflammatory pathology, characterized by Th1 and Th2/0 responses and enhanced IgG1 and IgE levels, which was delayed by wild-type regulatory T cell reconstitution. We therefore propose that the pathology characterizing Malt1(PD/PD) animals arises from an immune imbalance featuring pathogenic Th1- and Th2/0-skewed effector responses and reduced immunosuppressive compartments. These data uncover a previously unappreciated key function of MALT1 protease activity in immune homeostasis and underline its relevance in human health and disease.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Caspases/immunology , Cell Differentiation/immunology , Cell Proliferation , Encephalomyelitis, Autoimmune, Experimental/immunology , Neoplasm Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B-Lymphocytes, Regulatory/pathology , Caspases/genetics , Cell Differentiation/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Mice , Mice, Knockout , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Neoplasm Proteins/genetics , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology
17.
Cancer Biomark ; 15(1): 79-87, 2015.
Article in English | MEDLINE | ID: mdl-25524945

ABSTRACT

BACKGROUND: The JAK-STAT pathway is an important signaling pathway downstream of multiple cytokine and growth factor receptors. Dysregulated JAK-STAT signaling has been implicated in the pathogenesis of multiple human malignancies. OBJECTIVE: Given this pivotal role of JAK-STAT dysregulation, it is important to identify patients with an overactive JAK-STAT pathway for possible treatment with JAK inhibitors. METHODS: We developed a gene signature assay to detect overactive JAK-STAT signaling. The cancer cell line encyclopedia and associated gene-expression data were used to correlate the activation status of STAT5 with the induction of a set of STAT5 target genes. RESULTS: Four target genes were identified (PIM1, CISH, SOCS2, and ID1), the expression of which correlated significantly with pSTAT5 status in 40 hematologic tumor cell lines. In pSTAT5-positive models, the expression of the gene signature genes decreased following ruxolitinib treatment, which corresponded to pSTAT5 downmodulation. In pSTAT5-negative cell lines, neither pSTAT5 modulation nor a change in signature gene expression was observed following ruxolitinib treatment. CONCLUSIONS: The gene signature can potentially be used to stratify or enrich for patient populations with activated JAK-STAT5 signaling that might benefit from treatments targeting JAK-STAT signaling. Furthermore, the 4-gene signature is a predictor of the pharmacodynamic effects of ruxolitinib.


Subject(s)
Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Animals , Cell Line, Tumor , Female , Hematologic Neoplasms/drug therapy , Heterografts , Humans , Janus Kinases/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Nitriles , Pyrazoles/pharmacology , Pyrimidines , Signal Transduction/drug effects
18.
Cancer Discov ; 2(6): 512-523, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22684457

ABSTRACT

Janus kinase (JAK) inhibitors are being developed for the treatment of rheumatoid arthritis, psoriasis, myeloproliferative neoplasms, and leukemias. Most of these drugs target the ATP-binding pocket and stabilize the active conformation of the JAK kinases. This type I binding mode can lead to an increase in JAK activation loop phosphorylation, despite blockade of kinase function. Here we report that stabilizing the inactive state via type II inhibition acts in the opposite manner, leading to a loss of activation loop phosphorylation. We used X-ray crystallography to corroborate the binding mode and report for the first time the crystal structure of the JAK2 kinase domain in an inactive conformation. Importantly, JAK inhibitor-induced activation loop phosphorylation requires receptor interaction, as well as intact kinase and pseudokinase domains. Hence, depending on the respective conformation stabilized by a JAK inhibitor, hyperphosphorylation of the activation loop may or may not be elicited.


Subject(s)
Janus Kinases/antagonists & inhibitors , Janus Kinases/chemistry , Protein Kinase Inhibitors/pharmacology , Animals , Binding Sites , Cell Line, Tumor , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/chemistry , Mice , Phosphorylation/drug effects , Protein Binding , Protein Structure, Tertiary , STAT5 Transcription Factor/metabolism
19.
J Exp Med ; 209(2): 259-73, 2012 Feb 13.
Article in English | MEDLINE | ID: mdl-22271575

ABSTRACT

Enzymatic inhibitors of Janus kinase 2 (JAK2) are in clinical development for the treatment of myeloproliferative neoplasms (MPNs), B cell acute lymphoblastic leukemia (B-ALL) with rearrangements of the cytokine receptor subunit cytokine receptor-like factor 2 (CRLF2), and other tumors with constitutive JAK2 signaling. In this study, we identify G935R, Y931C, and E864K mutations within the JAK2 kinase domain that confer resistance across a panel of JAK inhibitors, whether present in cis with JAK2 V617F (observed in MPNs) or JAK2 R683G (observed in B-ALL). G935R, Y931C, and E864K do not reduce the sensitivity of JAK2-dependent cells to inhibitors of heat shock protein 90 (HSP90), which promote the degradation of both wild-type and mutant JAK2. HSP90 inhibitors were 100-1,000-fold more potent against CRLF2-rearranged B-ALL cells, which correlated with JAK2 degradation and more extensive blockade of JAK2/STAT5, MAP kinase, and AKT signaling. In addition, the HSP90 inhibitor AUY922 prolonged survival of mice xenografted with primary human CRLF2-rearranged B-ALL further than an enzymatic JAK2 inhibitor. Thus, HSP90 is a promising therapeutic target in JAK2-driven cancers, including those with genetic resistance to JAK enzymatic inhibitors.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Leukemia, B-Cell/enzymology , Myeloproliferative Disorders/enzymology , Resorcinols/pharmacology , Signal Transduction/physiology , Animals , Cell Line, Tumor , Cell Proliferation , DNA Primers/genetics , Female , Flow Cytometry , Gene Expression Profiling , HSP90 Heat-Shock Proteins/metabolism , Humans , Immunoblotting , Immunohistochemistry , Isoxazoles/therapeutic use , Janus Kinase 2/metabolism , Leukemia, B-Cell/drug therapy , Leukemia, B-Cell/genetics , Luciferases , Mice , Mice, Inbred BALB C , Mutagenesis , Mutation, Missense/genetics , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Phosphorylation , RNA, Small Interfering/genetics , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Resorcinols/therapeutic use , X-Ray Microtomography
20.
Mol Cancer Ther ; 9(7): 1945-55, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20587663

ABSTRACT

The recent discovery of an acquired activating point mutation in JAK2, substituting valine at amino acid position 617 for phenylalanine, has greatly improved our understanding of the molecular mechanism underlying chronic myeloproliferative neoplasms. Strikingly, the JAK2(V617F) mutation is found in nearly all patients suffering from polycythemia vera and in roughly every second patient suffering from essential thrombocythemia and primary myelofibrosis. Thus, JAK2 represents a promising target for the treatment of myeloproliferative neoplasms and considerable efforts are ongoing to discover and develop inhibitors of the kinase. Here, we report potent inhibition of JAK2(V617F) and JAK2 wild-type enzymes by a novel substituted quinoxaline, NVP-BSK805, which acts in an ATP-competitive manner. Within the JAK family, NVP-BSK805 displays more than 20-fold selectivity towards JAK2 in vitro, as well as excellent selectivity in broader kinase profiling. The compound blunts constitutive STAT5 phosphorylation in JAK2(V617F)-bearing cells, with concomitant suppression of cell proliferation and induction of apoptosis. In vivo, NVP-BSK805 exhibited good oral bioavailability and a long half-life. The inhibitor was efficacious in suppressing leukemic cell spreading and splenomegaly in a Ba/F3 JAK2(V617F) cell-driven mouse mechanistic model. Furthermore, NVP-BSK805 potently suppressed recombinant human erythropoietin-induced polycythemia and extramedullary erythropoiesis in mice and rats.


Subject(s)
Cell Proliferation/drug effects , Janus Kinase 2/antagonists & inhibitors , Polycythemia/prevention & control , Quinoxalines/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Erythropoiesis/drug effects , Humans , Janus Kinase 2/chemistry , Janus Kinase 2/genetics , K562 Cells , Mice , Mice, Inbred BALB C , Mice, SCID , Models, Molecular , Molecular Structure , Mutation , Phosphorylation/drug effects , Polycythemia/metabolism , Polycythemia/pathology , Protein Structure, Tertiary , Quinoxalines/chemistry , Rats , STAT5 Transcription Factor/metabolism , Splenomegaly/metabolism , Splenomegaly/pathology , Splenomegaly/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...