Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Nature ; 570(7759): 112-116, 2019 06.
Article in English | MEDLINE | ID: mdl-31092922

ABSTRACT

Solid tumours are infiltrated by effector T cells with the potential to control or reject them, as well as by regulatory T (Treg) cells that restrict the function of effector T cells and thereby promote tumour growth1. The anti-tumour activity of effector T cells can be therapeutically unleashed, and is now being exploited for the treatment of some forms of human cancer. However, weak tumour-associated inflammatory responses and the immune-suppressive function of Treg cells remain major hurdles to broader effectiveness of tumour immunotherapy2. Here we show that, after disruption of the CARMA1-BCL10-MALT1 (CBM) signalosome complex, most tumour-infiltrating Treg cells produce IFNγ, resulting in stunted tumour growth. Notably, genetic deletion of both or even just one allele of CARMA1 (also known as Card11) in only a fraction of Treg cells-which avoided systemic autoimmunity-was sufficient to produce this anti-tumour effect, showing that it is not the mere loss of suppressive function but the gain of effector activity by Treg cells that initiates tumour control. The production of IFNγ by Treg cells was accompanied by activation of macrophages and upregulation of class I molecules of the major histocompatibility complex on tumour cells. However, tumour cells also upregulated the expression of PD-L1, which indicates activation of adaptive immune resistance3. Consequently, blockade of PD-1 together with CARMA1 deletion caused rejection of tumours that otherwise do not respond to anti-PD-1 monotherapy. This effect was reproduced by pharmacological inhibition of the CBM protein MALT1. Our results demonstrate that partial disruption of the CBM complex and induction of IFNγ secretion in the preferentially self-reactive Treg cell pool does not cause systemic autoimmunity but is sufficient to prime the tumour environment for successful immune checkpoint therapy.


Subject(s)
B-Cell CLL-Lymphoma 10 Protein/antagonists & inhibitors , B7-H1 Antigen/antagonists & inhibitors , CARD Signaling Adaptor Proteins/antagonists & inhibitors , Immunotherapy/methods , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Multiprotein Complexes/antagonists & inhibitors , Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Female , Immune Tolerance , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Macrophages/immunology , Male , Mice , Neoplasms/immunology , Neoplasms/pathology
2.
Blood ; 137(6): 788-800, 2021 02 11.
Article in English | MEDLINE | ID: mdl-32785655

ABSTRACT

MALT1 inhibitors are promising therapeutic agents for B-cell lymphomas that are dependent on constitutive or aberrant signaling pathways. However, a potential limitation for signal transduction-targeted therapies is the occurrence of feedback mechanisms that enable escape from the full impact of such drugs. Here, we used a functional genomics screen in activated B-cell-like (ABC) diffuse large B-cell lymphoma (DLBCL) cells treated with a small molecule irreversible inhibitor of MALT1 to identify genes that might confer resistance or enhance the activity of MALT1 inhibition (MALT1i). We find that loss of B-cell receptor (BCR)- and phosphatidylinositol 3-kinase (PI3K)-activating proteins enhanced sensitivity, whereas loss of negative regulators of these pathways (eg, TRAF2, TNFAIP3) promoted resistance. These findings were validated by knockdown of individual genes and a combinatorial drug screen focused on BCR and PI3K pathway-targeting drugs. Among these, the most potent combinatorial effect was observed with PI3Kδ inhibitors against ABC-DLBCLs in vitro and in vivo, but that led to an adaptive increase in phosphorylated S6 and eventual disease progression. Along these lines, MALT1i promoted increased MTORC1 activity and phosphorylation of S6K1-T389 and S6-S235/6, an effect that was only partially blocked by PI3Kδ inhibition in vitro and in vivo. In contrast, simultaneous inhibition of MALT1 and MTORC1 prevented S6 phosphorylation, yielded potent activity against DLBCL cell lines and primary patient specimens, and resulted in more profound tumor regression and significantly improved survival of ABC-DLBCLs in vivo compared with PI3K inhibitors. These findings provide a basis for maximal therapeutic impact of MALT1 inhibitors in the clinic, by disrupting feedback mechanisms that might otherwise limit their efficacy.


Subject(s)
Antineoplastic Agents/therapeutic use , Feedback, Physiological/drug effects , Lymphoma, Large B-Cell, Diffuse/drug therapy , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Receptors, Antigen, B-Cell/immunology , Toll-Like Receptors/immunology , Animals , Antineoplastic Agents/pharmacology , Drug Design , Drug Resistance, Neoplasm , Drug Synergism , Female , Humans , Lymphoma, Large B-Cell, Diffuse/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred NOD , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/physiology , Neoplasm Proteins/physiology , Organoids/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , RNA, Small Interfering/genetics , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
3.
Nat Chem Biol ; 15(3): 304-313, 2019 03.
Article in English | MEDLINE | ID: mdl-30692685

ABSTRACT

MALT1 paracaspase is central for lymphocyte antigen-dependent responses including NF-κB activation. We discovered nanomolar, selective allosteric inhibitors of MALT1 that bind by displacing the side chain of Trp580, locking the protease in an inactive conformation. Interestingly, we had previously identified a patient homozygous for a MALT1 Trp580-to-serine mutation who suffered from combined immunodeficiency. We show that the loss of tryptophan weakened interactions between the paracaspase and C-terminal immunoglobulin MALT1 domains resulting in protein instability, reduced protein levels and functions. Upon binding of allosteric inhibitors of increasing potency, we found proportionate increased stabilization of MALT1-W580S to reach that of wild-type MALT1. With restored levels of stable MALT1 protein, the most potent of the allosteric inhibitors rescued NF-κB and JNK signaling in patient lymphocytes. Following compound washout, MALT1 substrate cleavage was partly recovered. Thus, a molecular corrector rescues an enzyme deficiency by substituting for the mutated residue, inspiring new potential precision therapies to increase mutant enzyme activity in other deficiencies.


Subject(s)
Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Gene Expression Regulation , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/therapy , Lymphocytes/metabolism , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Male , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/ultrastructure , NF-kappa B/metabolism , Neoplasm Proteins , Signal Transduction
4.
Bioorg Med Chem Lett ; 41: 127996, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33775836

ABSTRACT

We report the discovery of a novel series of 1,5-bisphenylpyrazoles as potent MALT1 inhibitors. Structure-activity relationship exploration of a hit compound led to a potent MALT1 inhibitor. Compound 33 showed strong activity against MALT1 (IC50: 0.49 µM), potent cellular activity (NF-κB inhibition and inhibition of IL2 production), and high selectivity against caspase-3, -8, and -9. The results of a kinetics study suggest that compound 33 is a non-competitive inhibitor of MALT1 protein.


Subject(s)
Cysteine Proteinase Inhibitors/pharmacology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Pyrazoles/pharmacology , Allosteric Regulation/drug effects , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Structure-Activity Relationship
5.
Genes Chromosomes Cancer ; 59(9): 517-524, 2020 09.
Article in English | MEDLINE | ID: mdl-32348592

ABSTRACT

MALT lymphomas with API2(BIRC3)-MALT1 translocation usually have an indolent clinical course and rarely transform into aggressive lymphoma, and there have been no lymphoma cell lines carrying API2-MALT1 translocation reported to date. We established a novel lymphoma cell line named BMA19, carrying the API2-MALT1 translocation from a patient with histologic transformation of intestinal MALT lymphoma. The cells were suggested to carry API2-MALT1 and MYC-IGH translocations by chromosomal analysis, and these translocations were confirmed by polymerase chain reaction analysis. The expression of MYC was shown to be enhanced as a result of the MYC-IGH translocation, and it is considered to have played a role in the histologic transformation of MALT lymphoma. Whole exome sequencing of BMA19 identified several nucleotide variations in genes reported to be mutated in previous studies of marginal zone lymphomas. The MALT1 inhibitor MI-2 specifically decreased cell growth, and the BMA19 cell line was suggested to be still dependent on the API2-MALT1 signal. Subtractive microarray analysis showed that one of the earliest events resulting from MALT1 inhibition is increased susceptibility to endoplasmic reticulum stress-induced apoptosis. The BMA19 cell line is considered to conserve the biological properties of MALT lymphoma and is expected to be a valuable tool for research into the pathogenesis of MALT lymphoma with an API2-MALT1 translocation.


Subject(s)
Cell Line Authentication/methods , Lymphoma/pathology , Oncogene Proteins, Fusion/genetics , Primary Cell Culture/methods , Aged, 80 and over , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation/drug effects , Endoplasmic Reticulum Stress , Humans , Lymphoma/genetics , Lymphoma/metabolism , Male , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Oncogene Proteins, Fusion/metabolism , Tumor Cells, Cultured
6.
J Cell Mol Med ; 24(13): 7550-7562, 2020 07.
Article in English | MEDLINE | ID: mdl-32452133

ABSTRACT

Glioblastoma multiforme (GBM) is the most common malignant tumour in the adult brain and hard to treat. Nuclear factor κB (NF-κB) signalling has a crucial role in the tumorigenesis of GBM. EGFR signalling is an important driver of NF-κB activation in GBM; however, the correlation between EGFR and the NF-κB pathway remains unclear. In this study, we investigated the role of mucosa-associated lymphoma antigen 1 (MALT1) in glioma progression and evaluated the anti-tumour activity and effectiveness of MI-2, a MALT1 inhibitor in a pre-clinical GBM model. We identified a paracaspase MALT1 that is involved in EGFR-induced NF-kB activation in GBM. MALT1 deficiency or inhibition significantly affected the proliferation, survival, migration and invasion of GBM cells both in vitro and in vivo. Moreover, MALT1 inhibition caused G1 cell cycle arrest by regulating multiple cell cycle-associated proteins. Mechanistically, MALTI inhibition blocks the degradation of IκBα and prevents the nuclear accumulation of the NF-κB p65 subunit in GBM cells. This study found that MALT1, a key signal transduction cascade, can mediate EGFR-induced NF-kB activation in GBM and may be potentially used as a novel therapeutic target for GBM.


Subject(s)
ErbB Receptors/metabolism , Glioblastoma/metabolism , Molecular Targeted Therapy , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , NF-kappa B/metabolism , Animals , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Epidermal Growth Factor/pharmacology , Glioblastoma/pathology , Humans , Mice, Knockout , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Neoplasm Invasiveness , Tumor Stem Cell Assay
7.
Int J Cancer ; 146(6): 1618-1630, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31291468

ABSTRACT

MALT1 is a key mediator of NF-κB signaling and a main driver of B-cell lymphomas. Remarkably, MALT1 is expressed in the majority of pancreatic ductal adenocarcinomas (PDACs) as well, but absent from normal exocrine pancreatic tissue. Following, MALT1 shows off to be a specific target in cancer cells of PDAC without affecting regular pancreatic cells. Therefore, we studied the impact of pharmacological MALT1 inhibition in pancreatic cancer and showed promising effects on tumor progression. Mepazine (Mep), a phenothiazine derivative, is a known potent MALT1 inhibitor. Newly, we described that biperiden (Bip) is a potent MALT1 inhibitor with even less pharmacological side effects. Thus, Bip is a promising drug leading to reduced proliferation and increased apoptosis in PDAC cells in vitro and in vivo. By compromising MALT1 activity, nuclear translocation of c-Rel is prevented. c-Rel is critical for NF-κB-dependent inhibition of apoptosis. Hence, off-label use of Bip or Mep represents a promising new therapeutic approach to PDAC treatment. Regularly, the Anticholinergicum Bip is used to treat neurological side effects of Phenothiazines, like extrapyramidal symptoms.


Subject(s)
Biperiden/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Phenothiazines/pharmacology , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Humans , Mice , Mice, Knockout , Models, Molecular , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/biosynthesis , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/chemistry , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , NF-kappa B/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-rel/metabolism , Random Allocation , Xenograft Model Antitumor Assays
8.
Eur J Haematol ; 105(4): 460-467, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32574386

ABSTRACT

OBJECTIVES: T-cell receptor (TCR) signaling-induced activation of NF-κB requires assembly of the CARD11-BCL10-MALT-1 complex and IκB kinase (IKK). Gain-of-function alterations in this component of the TCR/NF-κB pathway are associated with the development of HTLV-1-driven adult T-cell leukemia (ATL). We aimed to determine whether inhibition of MALT-1-mediated NF-κB activation could have anti-ATL activity. METHODS: RT-PCR, immunoblotting, and electrophoretic mobility shift assays were performed to assess expression levels of MALT-1 and the intracellular signaling cascades. Cell proliferation, cell cycle progression, and apoptotic events were examined using WST-8 assays, flow cytometry, and Hoechst 33342 staining. RESULTS: MALT-1 expression was upregulated in ATL-derived T-cell lines compared to that in normal PBMCs and uninfected or HTLV-1-transformed T-cell lines. Targeting MALT-1 with siRNA decreased cell proliferation. A MALT-1 inhibitor (MI-2) suppressed cleavage of the MALT-1-target protein, CYLD, and inhibited proliferation via G1 phase arrest. MI-2 induced apoptosis through caspase-3/8/9 activation and inhibited the phosphorylation of IKKα/ß and IκBα, resulting in the accumulation of IκBα and suppression of NF-κB-DNA binding. Additionally, MI-2 inhibited the expression of apoptosis- and cell cycle-related proteins regulated by NF-κB. CONCLUSIONS: MALT-1 plays an important regulatory role in NF-κB signaling during ATL-genesis, and targeting MALT-1 is a promising therapeutic strategy for this disease.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Leukemia-Lymphoma, Adult T-Cell/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Immunophenotyping , Leukemia-Lymphoma, Adult T-Cell/diagnosis , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/etiology , Molecular Targeted Therapy/methods , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Phosphorylation , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
9.
Int J Mol Sci ; 21(20)2020 Oct 20.
Article in English | MEDLINE | ID: mdl-33092214

ABSTRACT

Pulmonary fibrosis is a progressive disease characterized by lung remodeling due to excessive deposition of extracellular matrix. In this study, the bleomycin experimental model of pulmonary fibrosis was employed to investigate the anti-fibrotic and immunomodulatory activity of the inhibition of MALT1 protease activity. Mice received a single intra-tracheal administration of bleomycin (1 mg/kg) in the presence or absence of MI-2, a selective MALT1 inhibitor, (a dose of 30 mg/kg administered intra-peritoneally 1 h after bleomycin and daily until the end of the experiment). Seven days after bleomycin instillation mice were sacrificed and bronchoalveolar lavage fluid analysis, measurement of collagen content in the lung, histology, molecular analysis and immunohistochemistry were performed. To evaluate mortality and body weight gain a subset of mice was administered daily with MI-2 for 21 days. Mice that received MI-2 showed decreased weight loss and mortality, inflammatory cells infiltration, cytokines overexpression and tissue injury. Moreover, biochemical and immunohistochemical analysis displayed that MI-2 was able to modulate the excessive production of reactive oxygen species and the inflammatory mediator upregulation induced by bleomycin instillation. Additionally, MI-2 demonstrated anti-fibrotic activity by reducing transforming growth factor-ß (TGF-ß), α-smooth muscle actin (α-SMA) and receptor associated factor 6 (TRAF6) expression. The underlying mechanisms for the protective effect of MI-2 bleomycin induced pulmonary fibrosis may be attributed to its inhibition on NF-κB pathway. This is the first report showing the therapeutic role of MALT1 inhibition in a bleomycin model of pulmonary fibrosis, thus supporting further preclinical and clinical studies.


Subject(s)
Enzyme Inhibitors/pharmacology , Lung Injury/prevention & control , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Pulmonary Fibrosis/drug therapy , Animals , Bleomycin , Bronchoalveolar Lavage Fluid/chemistry , Collagen/metabolism , Cytokines/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung Injury/metabolism , Male , Mice , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , NF-kappa B/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Signal Transduction/drug effects , TNF Receptor-Associated Factor 6/metabolism , Transforming Growth Factor beta/metabolism
10.
J Virol ; 92(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30158289

ABSTRACT

Rabies virus is a neurovirulent RNA virus, which causes about 59,000 human deaths each year. Treatment for rabies does not exist due to incomplete understanding of the pathogenesis. MALT1 mediates activation of several immune cell types and is involved in the proliferation and survival of cancer cells. MALT1 acts as a scaffold protein for NF-κB signaling and a cysteine protease that cleaves substrates, leading to the expression of immunoregulatory genes. Here, we examined the impact of genetic or pharmacological MALT1 inhibition in mice on disease development after infection with the virulent rabies virus strain CVS-11. Morbidity and mortality were significantly delayed in Malt1-/- compared to Malt1+/+ mice, and this effect was associated with lower viral load, proinflammatory gene expression, and infiltration and activation of immune cells in the brain. Specific deletion of Malt1 in T cells also delayed disease development, while deletion in myeloid cells, neuronal cells, or NK cells had no effect. Disease development was also delayed in mice treated with the MALT1 protease inhibitor mepazine and in knock-in mice expressing a catalytically inactive MALT1 mutant protein, showing an important role of MALT1 proteolytic activity. The described protective effect of MALT1 inhibition against infection with a virulent rabies virus is the precise opposite of the sensitizing effect of MALT1 inhibition that we previously observed in the case of infection with an attenuated rabies virus strain. Together, these data demonstrate that the role of immunoregulatory responses in rabies pathogenicity is dependent on virus virulence and reveal the potential of MALT1 inhibition for therapeutic intervention.IMPORTANCE Rabies virus is a neurotropic RNA virus that causes encephalitis and still poses an enormous challenge to animal and public health. Efforts to establish reliable therapeutic strategies have been unsuccessful and are hampered by gaps in the understanding of virus pathogenicity. MALT1 is an intracellular protease that mediates the activation of several innate and adaptive immune cells in response to multiple receptors, and therapeutic MALT1 targeting is believed to be a valid approach for autoimmunity and MALT1-addicted cancers. Here, we study the impact of MALT1 deficiency on brain inflammation and disease development in response to infection of mice with the highly virulent CVS-11 rabies virus. We demonstrate that pharmacological or genetic MALT1 inhibition decreases neuroinflammation and extends the survival of CVS-11-infected mice, providing new insights in the biology of MALT1 and rabies virus infection.


Subject(s)
Brain/immunology , Inflammation/prevention & control , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/physiology , Rabies virus/immunology , Rabies/prevention & control , T-Lymphocytes/immunology , Animals , Brain/metabolism , Brain/virology , Cells, Cultured , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Rabies/immunology , Rabies/metabolism , T-Lymphocytes/pathology , T-Lymphocytes/virology
11.
Bioorg Med Chem Lett ; 29(23): 126743, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31678006

ABSTRACT

We describe a series of potent and highly selective small-molecule MALT1 inhibitors, optimized from a High-Throughput Screening hit. Advanced analogues such as compound 40 show high potency (IC50: 0.01 µM) in a biochemical assay measuring MALT1 enzymatic activity, as well as in cellular assays: Jurkat T cell activation (0.05 µM) and IL6/10 secretion (IC50: 0.10/0.06 µM) in the TMD8 B-cell lymphoma line. Compound 40 also inhibited cleavage of the MALT1 substrate RelB (IC50: 0.10 µM). Mechanistic enzymology results suggest that these compounds bind to the known allosteric site of the protease.


Subject(s)
Drug Discovery/methods , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Cell Line, Tumor , Humans
12.
Bioorg Med Chem Lett ; 29(14): 1694-1698, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31129051

ABSTRACT

Quinolines and thiazolopyridines were developed as allosteric inhibitors of MALT1, with good cellular potency and exquisite selectivity. Mouse pharmacokinetic (PK) profiling showed these to have low in vivo clearance, and moderate oral exposure. The thiazolopyridines were less lipophilic than the quinolines, and one thiazolopyridine example was active in our hIL10 mouse pharmacodynamic (PD) model upon oral dosing.


Subject(s)
Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Quinolines/therapeutic use , Animals , Disease Models, Animal , Humans , Quinolines/pharmacology
13.
Bioorg Med Chem Lett ; 29(11): 1336-1339, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30954428

ABSTRACT

Potent and selective substrate-based covalent inhibitors of MALT1 protease were developed from the tetrapeptide tool compound Z-VRPR-fmk. To improve cell permeability, we replaced one arginine residue. We further optimized a series of tripeptides and identified compounds that were potent in both a GloSensor reporter assay measuring cellular MALT1 protease activity, and an OCI-Ly3 cell proliferation assay. Example compounds showed good overall selectivity towards cysteine proteases, and one compound was selected for further profiling in ABL-DLBCL cells and xenograft efficacy models.


Subject(s)
Caspase Inhibitors/pharmacology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Peptides/pharmacology , Caspase Inhibitors/chemical synthesis , Caspase Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Peptides/chemical synthesis , Peptides/chemistry , Structure-Activity Relationship
14.
J Nat Prod ; 82(1): 154-162, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30600998

ABSTRACT

Bioassay-guided separation of an extract from a Dictyosporium sp. isolate led to the identification of six new compounds, 1-6, together with five known compounds, 7-11. The structures of the new compounds were primarily established by extensive 1D and 2D NMR experiments. The absolute configurations of compounds 3-6 were determined by comparison of their experimental electronic circular dichroism (ECD) spectra with DFT quantum mechanical calculated ECD spectra. Compounds 3-5 possess novel structural scaffolds, and biochemical studies revealed that oxepinochromenones 1 and 7 inhibited the activity of MALT1 protease.


Subject(s)
Enzyme Inhibitors/isolation & purification , Fungi/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology
15.
Infect Immun ; 86(12)2018 12.
Article in English | MEDLINE | ID: mdl-30249750

ABSTRACT

Disruption of the healthy intestinal microbiome and homeostasis of the intestinal immune system, which are closely interactive, are two key factors for ulcerative colitis. Here, we show that MI-2, a selective inhibitor of mucosa-associated lymphoid tissue lymphoma translocation-1 (MALT1), alleviated excessive inflammatory responses and was associated with restoration of healthy intestinal microbiome in mice suffering from dextran sulfate sodium (DSS)-induced colitis. We found that the diversity of intestinal microbiome of mice with DSS-induced colitis was significantly lower than that of healthy mice. However, MI-2 treatment in mice with DSS-induced colitis resulted in restored microbially diverse populations. To understand the possibility of the beneficial effect of the restored microbially diverse populations of MI-2-treated mice with DSS-induced colitis, we showed that inserting fecal microbiota from MI-2-treated mice with DSS-induced colitis and healthy control mice into mice with DSS-induced colitis could alleviate symptoms of colitis. The possibility of MI-2 treatment in DSS-induced colitis, associated with restoration of healthy microbially diverse populations in addition to reshaping host immune modulating capacity by reducing inflammatory cytokines (tumor necrosis factor alpha, interleukin-1ß [IL-1ß], IL-17α, and IL-22), may be considered therapeutic for ulcerative colitis.


Subject(s)
Acetanilides/pharmacology , Colitis/immunology , Colitis/therapy , Gastrointestinal Microbiome/drug effects , Intestines/immunology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Triazoles/pharmacology , Animals , Cells, Cultured , Colitis/chemically induced , Colitis/microbiology , Cytokines/immunology , Dextran Sulfate , Fecal Microbiota Transplantation , Feces/microbiology , Female , Humans , Inflammation , Intestines/drug effects , Intestines/microbiology , Macrophages , Mice , Mice, Inbred C57BL , Monocytes , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/immunology
16.
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
17.
Bioorg Med Chem ; 26(12): 3321-3344, 2018 07 23.
Article in English | MEDLINE | ID: mdl-29751989

ABSTRACT

Recent studies revealed that MALT1 is a promising therapeutic target for the treatment of ABC-DLBCL. Among several reported MALT1 inhibitors, MI-2 as an irreversible inhibitor represents a new class of ABC-DLBCL therapeutics. Due to its inherent potential cross-reactivity, further structure-activity relationship (SAR) study is imperative. In this work, five focused compound libraries based on the chemical structure of MI-2 are designed and synthesized. The systematic SARs revealed that the side chain of 2-methoxyethoxy has little impact on the activity and can be replaced by other functionalized groups, providing new MI-2 analogues with retained or enhanced potency. Compounds 81-83 with terminal hydroxyl group as side chain displayed enhanced activities against MALT1. Replacement of triazole core with pyrazole is also tolerant, while structural modifications on other sites are detrimental. These findings will facilitate further development of small-molecule MALT1 inhibitors.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Triazoles/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/pharmacology
18.
Blood Adv ; 8(15): 4003-4016, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38820414

ABSTRACT

ABSTRACT: Because multiple myeloma (MM) poses a formidable therapeutic challenge despite recent progress, exploring novel targets is crucial. Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) emerges as a promising paracaspase with druggable potential, especially unexplored in MM. Our study provided compelling evidence demonstrating a statistically significant elevation of MALT1 expression in human primary MM cells. Moreover, elevated MALT1 expression was associated with a poorer prognosis in MM. Genetic deletion of MALT1 reduced cell growth, colony formation, and tumor growth in vivo. Pharmacological inhibition with 1 µM of a small-molecular MALT1 inhibitor, Mi-2, effectively inhibited cell growth, inducing mitochondria-dependent apoptotic cell death. Mechanistically, MALT1 inhibition disrupted diverse signal transduction pathways, notably impeding nuclear factor κB (NF-κB). Significantly, the inhibition of MALT1 demonstrated a substantial suppression of NF-κB activation by elevating inhibitor of NF-κB, disrupting the nuclear localization of p65 and c-REL. This effect was observed in both the basal state and when stimulated by B-cell maturation antigen, highlighting the pivotal role of MALT1 inhibition in influencing MM cell survival. It was noteworthy that Mi-2 induces properties associated with immunogenic cell death (ICD), as evidenced by increased calreticulin, adenosine triphosphate release, and high-mobility group protein B1 upregulation, consequently triggering ICD-associated immune activation and enhancing CD8+ T-cell cytotoxicity in vitro. In conclusion, our research highlights MALT1 as a promising druggable target for therapeutic interventions in MM, providing insights into its molecular mechanisms in MM progression.


Subject(s)
B-Cell Maturation Antigen , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Multiple Myeloma , NF-kappa B , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , NF-kappa B/metabolism , Animals , Mice , B-Cell Maturation Antigen/metabolism , Cell Line, Tumor , Apoptosis/drug effects , Signal Transduction/drug effects , Cell Proliferation/drug effects , Cell Death/drug effects , Gene Expression Regulation, Neoplastic/drug effects
19.
Mol Cancer Ther ; 23(7): 949-960, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38507740

ABSTRACT

The activated B cell (ABC) subset of diffuse large B-cell lymphoma (DLBCL) is characterized by chronic B-cell receptor signaling and associated with poor outcomes when treated with standard therapy. In ABC-DLBCL, MALT1 is a core enzyme that is constitutively activated by stimulation of the B-cell receptor or gain-of-function mutations in upstream components of the signaling pathway, making it an attractive therapeutic target. We discovered a novel small-molecule inhibitor, ABBV-MALT1, that potently shuts down B-cell signaling selectively in ABC-DLBCL preclinical models leading to potent cell growth and xenograft inhibition. We also identified a rational combination partner for ABBV-MALT1 in the BCL2 inhibitor, venetoclax, which when combined significantly synergizes to elicit deep and durable responses in preclinical models. This work highlights the potential of ABBV-MALT1 monotherapy and combination with venetoclax as effective treatment options for patients with ABC-DLBCL.


Subject(s)
Drug Synergism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Proto-Oncogene Proteins c-bcl-2 , Xenograft Model Antitumor Assays , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Humans , Animals , Mice , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Cell Line, Tumor , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Cell Proliferation/drug effects , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Disease Models, Animal
20.
Front Immunol ; 13: 875320, 2022.
Article in English | MEDLINE | ID: mdl-35615349

ABSTRACT

MALT1 forms part of a central signaling node downstream of immunoreceptor tyrosine-based activation motif (ITAM)-containing receptors, across a broad range of immune cell subsets, and regulates NF-κB driven transcriptional responses via dual scaffolding-protease activity. Allosteric inhibition of MALT1 activity has demonstrated benefit in animal models of inflammation. However, development of MALT1 inhibitors to treat autoimmune and inflammatory diseases (A&ID) has been hindered by reports linking MALT1 inhibition and genetic loss-of-function to reductions in regulatory T-cell (Treg) numbers and development of auto-inflammatory syndromes. Using an allosteric MALT1 inhibitor, we investigated the consequence of pharmacological inhibition of MALT1 on proinflammatory cells compared to regulatory T-cells. Consistent with its known role in ITAM-driven responses, MALT1 inhibition suppressed proinflammatory cytokine production from activated human T-cells and monocyte-derived macrophages, and attenuated B-cell proliferation. Oral administration of a MALT1 inhibitor reduced disease severity and synovial cytokine production in a rat collagen-induced arthritis model. Interestingly, reduction in splenic Treg numbers was less pronounced in the context of inflammation compared with naïve animals. Additionally, in the context of the disease model, we observed an uncoupling of anti-inflammatory effects of MALT1 inhibition from Treg reduction, with lower systemic concentrations of inhibitor needed to reduce disease severity compared to that required to reduce Treg numbers. MALT1 inhibition did not affect suppressive function of human Tregs in vitro. These data indicate that anti-inflammatory efficacy can be achieved with MALT1 inhibition without impacting the number or function of Tregs, further supporting the potential of MALT1 inhibition in the treatment of autoimmune disease.


Subject(s)
Autoimmune Diseases , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , T-Lymphocytes, Regulatory , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Cytokines/genetics , Inflammation , Lymphocyte Activation , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors , NF-kappa B , Rats , T-Lymphocytes, Regulatory/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL