Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Mod Pathol ; 34(12): 2183-2191, 2021 12.
Article in English | MEDLINE | ID: mdl-34376807

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) is a catalytic component of the polycomb repressive complex 2 (PRC2) which reduces gene expression via trimethylation of a lysine residue of histone 3 (H3K27me3). Expression of EZH2 has not been assessed systematically in mantle cell lymphoma (MCL). Expression of EZH2 was assessed by immunohistochemistry in 166 patients with MCL. We also assessed other PRC2 components and H3K27me3. Fifty-seven (38%) of MCL patients were positive for EZH2 using 40% cutoff. EZH2 expression was associated with aggressive histologic variants (65% vs. 29%, p < 0.001), high Ki-67 proliferation rate (median, 72% vs. 19%, p < 0.001), and p53 overexpression (43% vs. 2%, p < 0.001). EZH2 expression did not correlate with expression of other PRC2 components (EED and SUZ12), H3K27me3, MHC-I, and MHC-II. Patients with EZH2 expression (EZH2+) had a poorer overall survival (OS) compared with patients without EZH2 expression (EZH2-) (median OS: 3.9 years versus 9.4 years, respectively, p < 0.001). EZH2 expression also predicted a poorer prognosis in MCL patients with classic histology (median OS, 4.6 years for EZH2+ and 9.6 years for EZH2-negative, respectively, p < 0.001) as well as aggressive histology (median OS, 3.7 years for EZH2+ and 7.9 years for EZH2-negative, respectively, p = 0.046). However, EZH2 expression did not independently correlate with overall survival in a multivariate analysis. Gene expression analysis and pathway enrichment analysis demonstrated a significant enrichment in cell cycle and mitotic transition pathways in MCL with EZH2 expression. EZH2 expression detected by immunohistochemistry is present in 38% of MCL cases and it is associated with high proliferation rate, p53 overexpression, aggressive histologic variants, and poorer OS. Based on gene expression profiling data, EZH2 expression could potentiate cell cycle machinery in MCL. These data suggest that assessment of EZH2 expression could be useful to stratify MCL patients into low- and high-risk groups.


Subject(s)
Biomarkers, Tumor/analysis , Enhancer of Zeste Homolog 2 Protein/analysis , Lymphoma, Mantle-Cell/enzymology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Histones/analysis , Humans , Immunohistochemistry , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/mortality , Lymphoma, Mantle-Cell/therapy , Male , Methylation , Middle Aged , Predictive Value of Tests , Risk Assessment , Risk Factors , Time Factors , Transcriptome , Treatment Outcome
2.
Cell Rep ; 34(11): 108870, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33730585

ABSTRACT

Ibrutinib, a bruton's tyrosine kinase (BTK) inhibitor, provokes robust clinical responses in aggressive mantle cell lymphoma (MCL), yet many patients relapse with lethal Ibrutinib-resistant (IR) disease. Here, using genomic, chemical proteomic, and drug screen profiling, we report that enhancer remodeling-mediated transcriptional activation and adaptive signaling changes drive the aggressive phenotypes of IR. Accordingly, IR MCL cells are vulnerable to inhibitors of the transcriptional machinery and especially so to inhibitors of cyclin-dependent kinase 9 (CDK9), the catalytic subunit of the positive transcription elongation factor b (P-TEFb) of RNA polymerase II (RNAPII). Further, CDK9 inhibition disables reprogrammed signaling circuits and prevents the emergence of IR in MCL. Finally, and importantly, we find that a robust and facile ex vivo image-based functional drug screening platform can predict clinical therapeutic responses of IR MCL and identify vulnerabilities that can be targeted to disable the evolution of IR.


Subject(s)
Adenine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/genetics , Piperidines/therapeutic use , Transcription, Genetic , Adenine/pharmacology , Adenine/therapeutic use , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/metabolism , Drug Resistance, Neoplasm/drug effects , Enhancer Elements, Genetic/genetics , Humans , Lymphoma, Mantle-Cell/enzymology , Lymphoma, Mantle-Cell/pathology , Male , Mice, Inbred NOD , Mice, SCID , Piperidines/pharmacology , Protein Kinases/metabolism , RNA Polymerase II/metabolism , Signal Transduction/drug effects , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Transcriptome/genetics , Treatment Outcome
3.
Oncogene ; 40(3): 551-563, 2021 01.
Article in English | MEDLINE | ID: mdl-33191405

ABSTRACT

Although somatic mutations of DNA repair genes are frequent in mantle cell lymphoma (MCL), our understanding of their germline defects is limited. In a Chinese family with maternal Lynch syndrome and paternal B cell non-Hodgkin lymphoma, one sibling developed both Lynch syndrome and MCL. Lynch syndrome is caused by heterozygous mutations in mismatch repair (MMR) genes. To understand the genetic predispositions in the family, we performed exome sequencing and analyses of affected individuals and their tumor samples. A novel germline indel, MLH1 Gly101fsX1, was identified as the cause of Lynch syndrome, and unstable microsatellite loci and mutational signatures as evidence of defective MMR were revealed in the MCL sample. Furthermore, we included additional 15 MCL patients with early onset, and found by exome sequencing that 11 patients carried heterozygous germline variants of 20 DNA repair genes, including MSH2 in MMR. In the MCL with MSH2 Arg359fsX16, unstable microsatellite loci and defective MMR signatures were also found. In addition, five patients also had heterozygous germline variants of genes involved in B cell functions. Thus, our study found germline variants of genes in single-strand break repair, double-strand break repair, and Fanconi anemia pathway in early onset MCL; and for the first time we identified germline defects of MMR in two MCLs.


Subject(s)
DNA Repair , Germ-Line Mutation , Lymphoma, Mantle-Cell/enzymology , MutL Protein Homolog 1/metabolism , MutS Homolog 2 Protein/metabolism , Mutation, Missense , Amino Acid Substitution , Humans , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/pathology , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/genetics
4.
Hematol Oncol Clin North Am ; 34(5): 903-921, 2020 10.
Article in English | MEDLINE | ID: mdl-32861286

ABSTRACT

The Bruton tyrosine kinase inhibitors (BTKi), acalabrutinib, ibrutinib, and zanubrutinib, are all approved in the United States for the treatment of relapsed mantle cell lymphoma (MCL). BTKi as a class have become the preferred therapy for most of the patients with relapsed MCL, and ongoing clinical trials are evaluating whether combining BTKi with other targeted agents may deepen response and further improve outcomes. Emerging evidence supports the efficacy of BTKi-containing combinations as frontline treatment, and clinical studies to define the role of this class of drugs for newly diagnosed patients with MCL are in progress.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase , Benzamides/therapeutic use , Lymphoma, Mantle-Cell , Neoplasm Proteins , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrazines/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/metabolism , Humans , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/enzymology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism
5.
Hematol Oncol Clin North Am ; 34(5): 923-939, 2020 10.
Article in English | MEDLINE | ID: mdl-32861287

ABSTRACT

In this review, we explore insights into the pathophysiology of Bruton tyrosine kinase inhibitor (BTKi) resistance in mantle cell lymphoma, and consider potential therapeutic targets. We review the possible clinical benefits of giving BTKis alongside other novel therapies, and evaluate clinical data for treatment strategies post BTKi progression that may help guide current practice. We conclude by considering future approaches, including the potential role of chimeric antigen receptor T-cell therapy.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase , Drug Resistance, Neoplasm , Immunotherapy, Adoptive , Lymphoma, Mantle-Cell , Protein Kinase Inhibitors/therapeutic use , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/genetics , Agammaglobulinaemia Tyrosine Kinase/metabolism , Humans , Lymphoma, Mantle-Cell/enzymology , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/therapy
6.
Cells ; 9(4)2020 04 21.
Article in English | MEDLINE | ID: mdl-32326225

ABSTRACT

Mantle cell lymphoma (MCL) is an aggressive subtype of non-Hodgkin's lymphoma. Despite being responsive to combination chemotherapy, median survival remains around 5 years due to high rates of relapse. Sphingolipid metabolism regulates MCL survival and proliferation and we found that sphingosine-1-phosphate (S1P) is upregulated in MCL cells. Therapeutic targeting of the S1P1 receptor or knockdown of sphingosine kinase 1 (SK1), the enzyme responsible for generating S1P, in human MCL cells results in a significant increase in Natural Killer T (NKT) cell activation. NKT cells recognize glycolipid antigens presented on CD1d and can reduce MCL tumor burden in vivo. Lipidomic studies identified cardiolipin, which has been reported to bind to CD1d molecules, as being upregulated in SK1 knockdown cells. We found that the pretreatment of antigen presenting cells with cardiolipin leads to increased cytokine production by NKT cell hybridomas. Furthermore, the ability of cardiolipin to activate NKT cells was dependent on the structure of its acyl chains. Collectively, these studies delineate novel pathways important for immune recognition of malignant cells and could lead to the development of new treatments for lymphoma.


Subject(s)
Lymphoma, Mantle-Cell/enzymology , Lymphoma, Mantle-Cell/immunology , Natural Killer T-Cells/immunology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Animals , Antigens, CD1d , Cardiolipins/chemistry , Cardiolipins/metabolism , Cell Line, Tumor , Humans , Lymphocyte Activation/immunology , Lysophospholipids/pharmacology , Mice, Inbred C57BL , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
7.
Blood ; 136(3): 299-312, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32325489

ABSTRACT

Coordination of a number of molecular mechanisms including transcription, alternative splicing, and class switch recombination are required to facilitate development, activation, and survival of B cells. Disruption of these pathways can result in malignant transformation. Recently, next-generation sequencing has identified a number of novel mutations in mantle cell lymphoma (MCL) patients including mutations in the ubiquitin E3 ligase UBR5. Approximately 18% of MCL patients were found to have mutations in UBR5, with the majority of mutations within the HECT domain of the protein that can accept and transfer ubiquitin molecules to the substrate. Determining if UBR5 controls the maturation of B cells is important to fully understand malignant transformation to MCL. To elucidate the role of UBR5 in B-cell maturation and activation, we generated a conditional mutant disrupting UBR5's C-terminal HECT domain. Loss of the UBR5 HECT domain leads to a block in maturation of B cells in the spleen and upregulation of proteins associated with messenger RNA splicing via the spliceosome. Our studies reveal a novel role of UBR5 in B-cell maturation by stabilization of spliceosome components during B-cell development and suggests UBR5 mutations play a role in MCL transformation.


Subject(s)
B-Lymphocytes/enzymology , Lymphoma, Mantle-Cell/enzymology , Mutation , Neoplasm Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Humans , Lymphoma, Mantle-Cell/genetics , Mice , Mice, Mutant Strains , Neoplasm Proteins/genetics , Protein Domains , Ubiquitin-Protein Ligases/genetics
8.
J Cell Mol Med ; 23(11): 7785-7795, 2019 11.
Article in English | MEDLINE | ID: mdl-31517438

ABSTRACT

The patients with mantle cell lymphoma (MCL) have translocation t(11;14) associated with cyclin D1 overexpression. We observed that iron (an essential cofactor of dioxygenases including prolyl hydroxylases [PHDs]) depletion by deferoxamine blocked MCL cells' proliferation, increased expression of DNA damage marker γH2AX, induced cell cycle arrest and decreased cyclin D1 level. Treatment of MCL cell lines with dimethyloxalylglycine, which blocks dioxygenases involving PHDs by competing with their substrate 2-oxoglutarate, leads to their decreased proliferation and the decrease of cyclin D1 level. We then postulated that loss of EGLN2/PHD1 in MCL cells may lead to down-regulation of cyclin D1 by blocking the degradation of FOXO3A, a cyclin D1 suppressor. However, the CRISPR/Cas9-based loss-of-function of EGLN2/PHD1 did not affect cyclin D1 expression and the loss of FOXO3A did not restore cyclin D1 levels after iron chelation. These data suggest that expression of cyclin D1 in MCL is not controlled by ENGL2/PHD1-FOXO3A pathway and that chelation- and 2-oxoglutarate competition-mediated down-regulation of cyclin D1 in MCL cells is driven by yet unknown mechanism involving iron- and 2-oxoglutarate-dependent dioxygenases other than PHD1. These data support further exploration of the use of iron chelation and 2-oxoglutarate-dependent dioxygenase inhibitors as a novel therapy of MCL.


Subject(s)
Cyclin D1/metabolism , Dioxygenases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Iron Chelating Agents/pharmacology , Ketoglutaric Acids/pharmacology , Lymphoma, Mantle-Cell/enzymology , Amino Acids, Dicarboxylic/pharmacology , Cell Hypoxia/drug effects , Cell Line, Tumor , DNA Damage , Deferoxamine/pharmacology , Dioxygenases/metabolism , Down-Regulation/drug effects , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Humans , Hydroxylation , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Iron Deficiencies , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Clin Adv Hematol Oncol ; 17(4): 223-233, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31188814

ABSTRACT

Mantle cell lymphoma (MCL) is a heterogeneous and uncommon non-Hodgkin lymphoma that affects predominantly older patients and often is associated with an aggressive clinical course. MCL relies upon B-cell receptor signaling through Bruton tyrosine kinase (BTK); therefore, the development of the BTK inhibitors ibrutinib and acalabrutinib represents a therapeutic breakthrough. In this review, we provide a summary of the efficacy and safety data from the landmark trials of single-agent ibrutinib and acalabrutinib that led to US Food and Drug Administration approval of these agents for patients with relapsed or refractory MCL. Toxicities of interest observed with ibrutinib include bleeding, atrial fibrillation, and increased risk for infection. The selectivity of acalabrutinib for BTK is greater than that of ibrutinib, which mitigates the risk for certain off-target toxicities, including atrial fibrillation; however, these toxicities, along with frequent headaches, still occur. Ongoing clinical trials are investigating both alternate BTK inhibitors and BTK inhibitors in combination with chemo-immunotherapy or other targeted agents in an effort to enhance the depth and duration of response. Trials to evaluate the use of these agents in the frontline setting are emerging and are likely to build upon the success of BTK inhibitors in patients with MCL.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Lymphoma, Mantle-Cell/drug therapy , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Adenine/analogs & derivatives , Antigens, CD20/drug effects , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/adverse effects , Benzamides/therapeutic use , Cardiovascular Diseases/chemically induced , Clinical Trials as Topic , Forecasting , Gastrointestinal Neoplasms/chemically induced , Hemorrhage/chemically induced , Humans , Immunologic Factors/administration & dosage , Lymphocytosis/chemically induced , Lymphoma, Mantle-Cell/enzymology , Molecular Targeted Therapy/adverse effects , Opportunistic Infections/chemically induced , Piperidines , Pyrazines/adverse effects , Pyrazines/therapeutic use , Pyrazoles/adverse effects , Pyrazoles/therapeutic use , Pyrimidines/adverse effects , Pyrimidines/therapeutic use , Salvage Therapy
10.
Med Sci Monit ; 25: 2599-2608, 2019 Apr 09.
Article in English | MEDLINE | ID: mdl-30964854

ABSTRACT

BACKGROUND Mantle cell lymphoma (MCL) is a high-grade B-cell lymphoma with poor prognosis. Fludarabine is used alone or in combination for relapsed and advanced-stage MCL. The expression of the signal transducer and activator of transcription 5B (STAT5B) gene is associated with tumorigenesis in solid tumors, but its role in MCL remains unknown. The aims of this study were to investigate the role of STAT5B in GRANTA-519 human mantle cell lymphoma cells and drug resistance. MATERIAL AND METHODS GRANTA-519 human mantle cell lymphoma cells were cultured with and without 10 µM fludarabine dephosphorylated 9-ß-D-arabinofuranosyl-2-fluoroadenine, (2-F-araA) or 10 µM 4-hydroperoxycyclophosphamide (4-HC). The MTT assay assessed cell proliferation. Flow cytometry was used to investigate the cell cycle in MCL cells treated with the specific inhibitor of the Akt pathway, LY294002, and assessed cell cycle and cell apoptosis. Western blot was used to detect the expression levels of p-Akt/Akt and STAT5B/p-STAT5B. The gene expression profiles of lymph node (LN)-derived MCL cells were compared with peripheral blood (PB)-derived lymphocytes using bioinformatics and hierarchical cluster analysis. Quantitative reverse transcription polymerase chain reaction (RT-qPCR) was performed to determine the expression of the marker of proliferation Ki-67 (MKI67) gene. RESULTS STAT5B was significantly upregulated in LN-derived MCL cells compared with PB lymphocytes. Increased expression of STAT5B was associated with increased MCL cell proliferation and reduced cell apoptosis and was associated with drug resistance and activation of Akt. CONCLUSIONS STAT5B promoted cell proliferation and drug resistance in human MCL cells by activating the Akt signaling pathway.


Subject(s)
Drug Resistance, Neoplasm/genetics , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/pathology , Proto-Oncogene Proteins c-akt/metabolism , STAT5 Transcription Factor/genetics , Signal Transduction , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Enzyme Activation , Gene Expression Regulation, Neoplastic , Humans , Lymph Nodes/pathology , Lymphocytes/metabolism , Lymphoma, Mantle-Cell/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , STAT5 Transcription Factor/metabolism
11.
Blood ; 133(9): 952-961, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30545835

ABSTRACT

The covalent Bruton tyrosine kinase (BTK) inhibitor ibrutinib is highly efficacious against multiple B-cell malignancies. However, it is not selective for BTK, and multiple mechanisms of resistance, including the C481S-BTK mutation, can compromise its efficacy. We hypothesized that small-molecule-induced BTK degradation may overcome some of the limitations of traditional enzymatic inhibitors. Here, we demonstrate that BTK degradation results in potent suppression of signaling and proliferation in cancer cells and that BTK degraders efficiently degrade C481S-BTK. Moreover, we discovered DD-03-171, an optimized lead compound that exhibits enhanced antiproliferative effects on mantle cell lymphoma (MCL) cells in vitro by degrading BTK, IKFZ1, and IKFZ3 as well as efficacy against patient-derived xenografts in vivo. Thus, "triple degradation" may be an effective therapeutic approach for treating MCL and overcoming ibrutinib resistance, thereby addressing a major unmet need in the treatment of MCL and other B-cell lymphomas.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Lymphoma, Mantle-Cell/drug therapy , Small Molecule Libraries/pharmacology , Adenine/analogs & derivatives , Animals , Humans , Ikaros Transcription Factor/metabolism , Lymphoma, Mantle-Cell/enzymology , Lymphoma, Mantle-Cell/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Piperidines , Proteolysis , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Br J Haematol ; 184(2): 215-222, 2019 01.
Article in English | MEDLINE | ID: mdl-30183069

ABSTRACT

Spleen tyrosine kinase (Syk) mediates B-cell receptor signalling in normal and malignant B cells. Entospletinib is an oral, selective Syk inhibitor. Entospletinib monotherapy was evaluated in a multicentre, phase 2 study of patients with relapsed or refractory indolent non-Hodgkin lymphoma or mantle cell lymphoma (MCL). Subjects received 800 mg entospletinib twice daily. Forty-one follicular lymphoma (FL), 17 lymphoplasmacytoid lymphoma/Waldenström macroglobulinaemia (LPL/WM), 17 marginal zone lymphoma (MZL) and 39 MCL patients were evaluated. The primary endpoint was a progression-free survival (PFS) rate (defined as not experiencing progression or death) at 16 weeks for patients with MCL and at 24 weeks for patients with FL, LPL/WM and MZL. The most common treatment-emergent adverse events were fatigue, nausea, diarrhoea, vomiting, headache and cough. Common laboratory abnormalities were anaemia, neutropenia and thrombocytopenia; aspartate transaminase, alanine transaminase, total bilirubin and serum creatinine were all increased. PFS at 16 weeks in the MCL cohort was 63·9% [95% confidence interval (CI) 45-77·8%]; PFS at 24 weeks in the FL, LPL/WM, MCL and MZL cohorts was 51·5% (95% CI 32·8-67·4%), 69·8% (95% CI 31·8-89·4%), 56·6% (95% CI 37·5-71·8%) and 46·2% (95% CI 18·5-70·2%), respectively. Entospletinib had limited single-agent activity with manageable toxicity in these patient populations.


Subject(s)
Indazoles/administration & dosage , Lymphoma, B-Cell, Marginal Zone/drug therapy , Lymphoma, Follicular/drug therapy , Lymphoma, Mantle-Cell/drug therapy , Pyrazines/administration & dosage , Waldenstrom Macroglobulinemia/drug therapy , Adult , Aged , Aged, 80 and over , Female , Humans , Indazoles/adverse effects , Lymphoma, B-Cell, Marginal Zone/enzymology , Lymphoma, B-Cell, Marginal Zone/pathology , Lymphoma, Follicular/enzymology , Lymphoma, Follicular/pathology , Lymphoma, Mantle-Cell/enzymology , Lymphoma, Mantle-Cell/pathology , Male , Middle Aged , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Pyrazines/adverse effects , Syk Kinase/antagonists & inhibitors , Syk Kinase/metabolism , Waldenstrom Macroglobulinemia/enzymology , Waldenstrom Macroglobulinemia/pathology
13.
Drugs ; 78(16): 1653-1663, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30390220

ABSTRACT

Bruton's tyrosine kinase (BTK) is crucial in B-cell development and survival. The role of BTK as a downstream kinase in the B-cell receptor (BCR) signaling pathway is well described. As a key player in the pathogenesis of B-cell malignancies, targeting of dysregulated BCR signaling has been explored by development of inhibitors of downstream mediators. Discovery of the biological function of BTK and the development of covalent inhibitors for clinical use, ibrutinib as the lead agent and acalabrutinib as the second clinically approved BTK inhibitor, have revolutionized the treatment options for B-cell malignancies. Currently, ibrutinib is approved for mantle cell lymphoma, chronic lymphocytic leukemia, lymphoplasmacytic lymphoma/Waldenström macroglobulinemia, small lymphocytic lymphoma, marginal zone lymphoma and chronic graft versus host disease, while acalabrutinib is approved for mantle cell lymphoma. Potential expansion of indications in other diseases is under investigation in several clinical trials, while combination of BTK inhibitors with either chemoimmunotherapy or other targeted agents is being systematically explored in B-cell malignancies.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , B-Lymphocytes/metabolism , Lymphoma, B-Cell , Protein Kinase Inhibitors/pharmacology , Adenine/analogs & derivatives , Antineoplastic Agents/therapeutic use , Benzamides/pharmacology , Drug Therapy, Combination , Humans , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/enzymology , Lymphoma, Mantle-Cell/enzymology , Molecular Targeted Therapy , Piperidines , Protein Kinase Inhibitors/therapeutic use , Pyrazines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology
15.
Expert Rev Hematol ; 11(6): 495-502, 2018 06.
Article in English | MEDLINE | ID: mdl-29737219

ABSTRACT

INTRODUCTION: Acalabrutinib, a selective Bruton tyrosine kinase (BTK) inhibitor, was granted accelerated approval by the FDA on 31 October 2017 for the treatment of patients with mantle cell lymphoma (MCL) who have received at least one prior therapy. Areas covered: This narrative review provides an overview of acalabrutinib, its use in clinical practice and potential future developments. Expert commentary: BTK inhibitors have demonstrated efficacy in patients with relapsed or refractory MCL. To prepare patients for therapy, all preexisting infections should be diagnosed and treated, and infection prophylaxis undertaken. Serious adverse reactions are rare with acalabrutinib; however, patients should be made aware of common adverse events such as headaches, which usually resolve within one month without medical treatment. Interaction with other drugs appears to be less of an issue with acalabrutinib than with ibrutinib; however, patients receiving acalabrutinib therapy must be advised not to take any additional medications without first consulting with their treating physician. A key unmet medical need is treatment options for patients in whom BTK inhibitors are discontinued, because of either intolerance or refractory disease. Patients not tolerating ibrutinib could be switched to acalabrutinib, which has improved selectivity and increased tolerability. First-line treatment with acalabrutinib is being investigated.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Benzamides/therapeutic use , Lymphoma, Mantle-Cell/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrazines/therapeutic use , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase/metabolism , Benzamides/adverse effects , Humans , Infection Control/methods , Infections/chemically induced , Infections/metabolism , Lymphoma, Mantle-Cell/enzymology , Lymphoma, Mantle-Cell/pathology , Piperidines , Protein Kinase Inhibitors/adverse effects , Pyrazines/adverse effects , Pyrazoles/adverse effects , Pyrazoles/therapeutic use , Pyrimidines/adverse effects , Pyrimidines/therapeutic use
16.
Best Pract Res Clin Haematol ; 31(1): 105-113, 2018 03.
Article in English | MEDLINE | ID: mdl-29452660

ABSTRACT

Mantle cell lymphoma is an aggressive Non-Hodgkin's lymphoma that is considered incurable with standard therapies. Most patients treated with frontline immunochemotherapy relapse within a few years and do not usually respond to salvage chemotherapy. Persistent activation of the B-cell receptor pathway is critical to the pathogenesis of mantle cell lymphoma. Inhibition of Bruton's tyrosine kinase, an essential B-cell receptor pathway component with ibrutinib has shown clinical activity and has changed how MCL is treated in the relapsed/refractory setting. However, resistance to ibrutinib is common and response is limited. Novel agents targeting the B-cell receptor pathway along with therapies outside of the pathway will be reviewed in this article. Ongoing and future studies will better define how these agents should be utilized in the ever-changing treatment landscape of mantle cell lymphoma.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Lymphoma, Mantle-Cell , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase/metabolism , Humans , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/enzymology , Lymphoma, Mantle-Cell/mortality , Lymphoma, Mantle-Cell/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Piperidines , Receptors, Antigen, B-Cell/metabolism , Recurrence
17.
J Cancer Res Clin Oncol ; 144(4): 697-706, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29392403

ABSTRACT

PURPOSE: We researched into the effect and mechanism of AC0010, a novel BTK inhibitor, in MCL, and compared its efficacy and safety with Ibrutinib to develop a preclinical study for the future therapy of MCL. METHODS: MTS assay was used to detect the growth inhibition caused by AC0010 and Ibrutinib, respectively, in MCL cell lines (Jeko-1 and JVM-2), primary MCL cells, and normal peripheral lymphocytes. Apoptosis of Jeko-1 and JVM-2 after exposure into AC0010 and Ibrutinib was conducted by flow cytometry; the expression of apoptosis-related proteins was checked by Western blot. q-PCR and Western blot were applied to examine the expression of BTK and p-BTK at mRNA and protein level as well as the BTK-ralated signaling pathways. MCL xenograft was developed to testify the efficacy and safety of AC0010 in vivo. RESULTS: In contrast with Ibrutinib, AC0010 proved to be more toxic to MCL cells in vitro (p < 0.01) with no augment in cytotoxicity to normal peripheral lymphocytes, and it can induce obvious apoptosis in MCL cell lines (p < 0.01) through caspase family and Bcl-2 family. AC0010 at 300 mg/kg can prolong the survival rate in MCL xenograft (p < 0.01). The phosphorylation of BTK is inhibited by AC0010 following simultaneously inhibition of BCR-BTK and PI3K/AKT signaling pathway in MCL cells. CONCLUSION: AC0010 is a novel BTK inhibitor of great efficacy and safety in MCL.


Subject(s)
Lymphoma, Mantle-Cell/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase , Animals , Female , Humans , Lymphoma, Mantle-Cell/enzymology , Mice , Mice, SCID , Piperidines , Protein Kinase Inhibitors/adverse effects , Pyrazoles/adverse effects , Pyrazoles/pharmacology , Pyrimidines/adverse effects , Pyrimidines/pharmacology , Xenograft Model Antitumor Assays
19.
Exp Hematol ; 59: 72-81.e2, 2018 03.
Article in English | MEDLINE | ID: mdl-29287939

ABSTRACT

Mantle cell lymphoma (MCL) is a relatively rare subtype of B-cell non-Hodgkin lymphoma (NHL) that has a poor prognosis despite recent advances in immunochemotherapy and molecular targeted therapeutics against NHL. Therefore, the development of a new therapeutic strategy for MCL is urgently needed. In this study, we show for the first time that 3-phosphoinositide-dependent protein kinase 1 (PDPK1), an oncogenic serine-threonine protein kinase, is commonly expressed in its phosphorylated active form in patient-derived tumor cells of various types of B-cell NHL cells, including diffuse large B-cell lymphoma, follicular lymphoma, and MCL. Blockade of PDPK1 activity by small-molecule inhibitors specific for PDPK1 (BX-912 and GSK2334470) or by RNA interference exerted antiproliferative effects in all four MCL-derived cell lines examined and these growth-inhibitory effects were mediated by both induction of apoptosis and G2/M cell cycle blockade. In addition, blockade of PDPK1 led to inactivation of its downstream effector kinase RSK2, but not AKT, suggesting the importance of the PDPK1/RSK2 signaling pathway in the proliferation and survival of MCL cells. Finally, when combined with anticancer agents, including genotoxic agents, a proteasome inhibitor, and a BH3 mimetic in vitro, the PDPK1 inhibitor BX-912 showed additive growth-inhibitory effects against MCL-derived cell lines in most settings. In particular, the combination of BX-912 and ABT-263, a BH3 mimetic, resulted in the enhancement of the induction of apoptosis. In conclusion, our results suggest that PDPK1 is a potential novel therapeutic target in MCL and indicate that clinical development of PDPK1-targeted therapy for MCL is desirable.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/antagonists & inhibitors , Aniline Compounds/pharmacology , Indazoles/pharmacology , Lymphoma, Mantle-Cell/drug therapy , Pyrimidines/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Cell Line, Tumor , Humans , Lymphoma, Mantle-Cell/enzymology , Lymphoma, Mantle-Cell/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL