Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Haematologica ; 104(3): 576-586, 2019 03.
Article in English | MEDLINE | ID: mdl-30262568

ABSTRACT

Mutations in genes of the RAS-BRAF-MAPK-ERK pathway have not been fully explored in patients with chronic lymphocytic leukemia. We, therefore, analyzed the clinical and biological characteristics of chronic lymphocytic leukemia patients with mutations in this pathway and investigated the in vitro response of primary cells to BRAF and ERK inhibitors. Putative damaging mutations were found in 25 of 452 patients (5.5%). Among these, BRAF was mutated in nine patients (2.0%), genes upstream of BRAF (KITLG, KIT, PTPN11, GNB1, KRAS and NRAS) were mutated in 12 patients (2.6%), and genes downstream of BRAF (MAPK2K1, MAPK2K2, and MAPK1) were mutated in five patients (1.1%). The most frequent mutations were missense, subclonal and mutually exclusive. Patients with these mutations more frequently had increased lactate dehydrogenase levels, high expression of ZAP-70, CD49d, CD38, trisomy 12 and unmutated immunoglobulin heavy-chain variable region genes and had a worse 5-year time to first treatment (hazard ratio 1.8, P=0.025). Gene expression analysis showed upregulation of genes of the MAPK pathway in the group carrying RAS-BRAF-MAPK-ERK pathway mutations. The BRAF inhibitors vemurafenib and dabrafenib were not able to inhibit phosphorylation of ERK, the downstream effector of the pathway, in primary cells. In contrast, ulixertinib, a pan-ERK inhibitor, decreased phospho-ERK levels. In conclusion, although larger series of patients are needed to corroborate these findings, our results suggest that the RAS-BRAF-MAPK-ERK pathway is one of the core cellular processes affected by novel mutations in chronic lymphocytic leukemia, is associated with adverse clinical features and could be pharmacologically inhibited.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , MAP Kinase Signaling System , Mutation , Proto-Oncogene Proteins B-raf/metabolism , ras Proteins/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Cell Line, Tumor , Cell Proliferation/drug effects , Computational Biology/methods , Female , Gene Expression Profiling , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Transcriptome , Young Adult
2.
Br J Haematol ; 177(4): 557-561, 2017 05.
Article in English | MEDLINE | ID: mdl-28295185

ABSTRACT

Imbalances in the composition of BCL2 family proteins contribute to tumourigenesis and therapy resistance of mantle cell lymphoma (MCL), making these proteins attractive therapy targets. We studied the efficiency of dual targeting the NOXA/MCL1 axis by combining fatty acid synthase inhibitors (NOXA stabilization) with the CDK inhibitor Dinaciclib (MCL1 reduction). This combination synergistically induced apoptosis in cell lines and primary MCL cells and led to almost complete inhibition of tumour progression in a mouse model. Apoptosis was NOXA-dependent and correlated with the NOXA/MCL1 ratio, highlighting the importance of the NOXA/MCL1 balance for effective cell death induction in MCL.


Subject(s)
Lymphoma, Mantle-Cell/drug therapy , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Animals , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Cyclic N-Oxides , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Female , Humans , Indolizines , Lactones/pharmacology , Mice, SCID , Orlistat , Pyridinium Compounds/pharmacology
3.
Clin Cancer Res ; 23(11): 2831-2841, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-27903679

ABSTRACT

Purpose: Acalabrutinib (ACP-196) is a novel, potent, and highly selective Bruton tyrosine kinase (BTK) inhibitor, which binds covalently to Cys481 in the ATP-binding pocket of BTK. We sought to evaluate the antitumor effects of acalabrutinib treatment in two established mouse models of chronic lymphocytic leukemia (CLL).Experimental Design: Two distinct mouse models were used, the TCL1 adoptive transfer model where leukemic cells from Eµ-TCL1 transgenic mice are transplanted into C57BL/6 mice, and the human NSG primary CLL xenograft model. Mice received either vehicle or acalabrutinib formulated into the drinking water.Results: Utilizing biochemical assays, we demonstrate that acalabrutinib is a highly selective BTK inhibitor as compared with ibrutinib. In the human CLL NSG xenograft model, treatment with acalabrutinib demonstrated on-target effects, including decreased phosphorylation of PLCγ2, ERK, and significant inhibition of CLL cell proliferation. Furthermore, tumor burden in the spleen of the mice treated with acalabrutinib was significantly decreased compared with vehicle-treated mice. Similarly, in the TCL1 adoptive transfer model, decreased phosphorylation of BTK, PLCγ2, and S6 was observed. Most notably, treatment with acalabrutinib resulted in a significant increase in survival compared with mice receiving vehicle.Conclusions: Treatment with acalabrutinib potently inhibits BTK in vivo, leading to on-target decreases in the activation of key signaling molecules (including BTK, PLCγ2, S6, and ERK). In two complementary mouse models of CLL, acalabrutinib significantly reduced tumor burden and increased survival compared with vehicle treatment. Overall, acalabrutinib showed increased BTK selectivity compared with ibrutinib while demonstrating significant antitumor efficacy in vivo on par with ibrutinib. Clin Cancer Res; 23(11); 2831-41. ©2016 AACR.


Subject(s)
Benzamides/administration & dosage , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Pyrazines/administration & dosage , Adenine/analogs & derivatives , Adoptive Transfer/methods , Agammaglobulinaemia Tyrosine Kinase , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis/drug effects , Disease Models, Animal , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mice , Mice, Transgenic , Piperidines , Protein Kinase Inhibitors/administration & dosage , Protein-Tyrosine Kinases/genetics , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Xenograft Model Antitumor Assays
4.
Oncotarget ; 7(5): 5507-20, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26701728

ABSTRACT

Clinical responses to bendamustine in chronic lymphocytic leukemia (CLL) are highly heterogeneous and no specific markers to predict sensitivity to this drug have been reported. In order to identify biomarkers of response, we analyzed the in vitro activity of bendamustine and the gene expression profile in primary CLL cells. We observed that mRNA expression of CD69 (CD69) and ITGAM (CD11b) constitute the most powerful predictor of response to bendamustine. When we interrogated the predictive value of the corresponding cell surface proteins, the expression of the activation marker CD69 was the most reliable predictor of sensitivity to bendamustine. Importantly, a multivariate analysis revealed that the predictive value of CD69 expression was independent from other clinico-biological CLL features. We also showed that when CLL cells were co-cultured with distinct subtypes of stromal cells, an upregulation of CD69 was accompanied by a reduced sensitivity to bendamustine. In agreement with this, tumor cells derived from lymphoid tumor niches harbored higher CD69 expression and were less sensitive to bendamustine than their peripheral blood counterparts. Furthermore, pretreatment of CD69 high CLL cases with the B-cell receptor (BCR) pathway inhibitors ibrutinib and idelalisib decreased CD69 levels and enhanced bendamustine cytotoxic effect. Collectively, our findings indicate that CD69 could be a predictor of bendamustine response in CLL patients and the combination of clinically-tested BCR signaling inhibitors with bendamustine may represent a promising strategy for bendamustine low responsive CLL cases.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Apoptosis/drug effects , Bendamustine Hydrochloride/pharmacology , Lectins, C-Type/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Purines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinazolinones/pharmacology , Adenine/analogs & derivatives , Adult , Aged , Aged, 80 and over , Antigens, CD/genetics , Antigens, Differentiation, T-Lymphocyte/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Cell Proliferation/drug effects , Female , Follow-Up Studies , Humans , Lectins, C-Type/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Neoplasm Staging , Piperidines , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Transcriptome , Tumor Cells, Cultured
5.
Oncotarget ; 6(25): 21159-72, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26110568

ABSTRACT

Acadesine is a nucleoside analogue with known activity against B-cell malignancies. Herein, we showed that in mantle cell lymphoma (MCL) cells acadesine induced caspase-dependent apoptosis through turning on the mitochondrial apoptotic machinery. At the molecular level, the compound triggered the activation of the AMPK pathway, consequently modulating known downstream targets, such as mTOR and the cell motility-related vasodilator-stimulated phosphoprotein (VASP). VASP phosphorylation by acadesine was concomitant with a blockade of CXCL12-induced migration. The inhibition of the mTOR cascade by acadesine, committed MCL cells to enter in apoptosis by a translational downregulation of the antiapoptotic Mcl-1 protein. In contrast, Bcl-2 protein levels were unaffected by acadesine and MCL samples expressing high levels of Bcl-2 tended to have a reduced response to the drug. Targeting Bcl-2 with the selective BH3-mimetic agent ABT-199 sensitized Bcl-2high MCL cells to acadesine. This effect was validated in vivo, where the combination of both agents displayed a more marked inhibition of tumor outgrowth than each drug alone. These findings support the notions that antiapoptotic proteins of the Bcl-2 family regulate MCL cell sensitivity to acadesine and that the combination of this agent with Bcl-2 inhibitors might be an interesting therapeutic option to treat MCL patients.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Cell Adhesion Molecules/metabolism , Lymphoma, Mantle-Cell/metabolism , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Ribonucleosides/administration & dosage , Sulfonamides/administration & dosage , Actins/chemistry , Aminoimidazole Carboxamide/administration & dosage , Animals , Apoptosis , Cell Line, Tumor/drug effects , Cell Movement , Chemokine CXCL12/metabolism , Chemotaxis , Down-Regulation , Drug Resistance, Neoplasm , Drug Synergism , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, SCID , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasm Transplantation
6.
Oncotarget ; 6(26): 22734-49, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26068951

ABSTRACT

Mutations or deregulated expression of the components of the spliceosome can influence the splicing pattern of several genes and contribute to the development of tumors. In this context, we report that the spliceosome modulator sudemycin induces selective cytotoxicity in primary chronic lymphocytic leukemia (CLL) cells when compared with healthy lymphocytes and tumor cells from other B-lymphoid malignancies, with a slight bias for CLL cases with mutations in spliceosome-RNA processing machinery. Consistently, sudemycin exhibits considerable antitumor activity in NOD/SCID/IL2Rγ-/- (NSG) mice engrafted with primary cells from CLL patients. The antileukemic effect of sudemycin involves the splicing modulation of several target genes important for tumor survival, both in SF3B1-mutated and -unmutated cases. Thus, the apoptosis induced by this compound is related to the alternative splicing switch of MCL1 toward its proapoptotic isoform. Sudemycin also functionally disturbs NF-κB pathway in parallel with the induction of a spliced RELA variant that loses its DNA binding domain. Importantly, we show an enhanced antitumor effect of sudemycin in combination with ibrutinib that might be related to the modulation of the alternative splicing of the inhibitor of Btk (IBTK). In conclusion, we provide first evidence that the spliceosome is a relevant therapeutic target in CLL, supporting the use of splicing modulators alone or in combination with ibrutinib as a promising approach for the treatment of CLL patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cyclohexylamines/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Spiro Compounds/pharmacology , Adenine/analogs & derivatives , Adult , Aged , Animals , Cyclohexylamines/administration & dosage , Drug Synergism , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Middle Aged , Mutation , Piperidines , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , RNA Splicing , Spiro Compounds/administration & dosage , Spliceosomes/drug effects , Spliceosomes/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Oncotarget ; 5(16): 6788-800, 2014 Aug 30.
Article in English | MEDLINE | ID: mdl-25216518

ABSTRACT

Phosphatidylinositol-3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway activation contributes to mantle cell lymphoma (MCL) pathogenesis and drug resistance. Antitumor activity has been observed with mTOR inhibitors. However, they have shown limited clinical efficacy in relation to drug activation of feedback loops. Selective PI3K inhibition or dual PI3K/mTOR catalytic inhibition are different therapeutic approaches developed to achieve effective pathway blockage. Here, we have performed a comparative analysis of the mTOR inhibitor everolimus, the pan-PI3K inhibitor NVP-BKM120 and the dual PI3K/mTOR inhibitor NVP-BEZ235 in primary MCL cells. We found NVP-BEZ235 to be more powerful than everolimus or NVP-BKM120 in PI3K/Akt/mTOR signaling inhibition, indicating that targeting the PI3K/Akt/mTOR pathway at multiple levels is likely to be a more effective strategy for the treatment of MCL than single inhibition of these kinases. Among the three drugs, NVP-BEZ235 induced the highest change in gene expression profile. Functional validation demonstrated that NVP-BEZ235 inhibited angiogenesis, migration and tumor invasiveness in MCL cells. NVP-BEZ235 was the only drug able to block IL4 and IL6/STAT3 signaling which compromise the therapeutic effect of chemotherapy in MCL. Our findings support the use of the dual PI3K/mTOR inhibitor NVP-BEZ235 as a promising approach to interfere with the microenvironment-related processes in MCL.


Subject(s)
Lymphoma, Mantle-Cell/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Aminopyridines/pharmacology , Cell Line, Tumor , Cell Proliferation , Chemokine CXCL12/metabolism , Everolimus , Human Umbilical Vein Endothelial Cells , Humans , Imidazoles/pharmacology , Interleukin-4/antagonists & inhibitors , Interleukin-4/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Lymphoma, Mantle-Cell/metabolism , Morpholines/pharmacology , Quinolines/pharmacology , Signal Transduction/drug effects , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Transcriptome , Tumor Microenvironment/drug effects
8.
Oncotarget ; 5(3): 726-39, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24519895

ABSTRACT

Mantle cell lymphoma (MCL) is considered one of the most challenging lymphoma, with limited responses to current therapies. Acadesine, a nucleoside analogue has shown antitumoral effects in different preclinical cancer models as well as in a recent phase I/II clinical trial conducted in patients with chronic lymphocytic leukemia. Here we observed that acadesine exerted a selective antitumoral activity in the majority of MCL cell lines and primary MCL samples, independently of adverse cytogenetic factors. Moreover, acadesine was highly synergistic, both in vitro and in vivo, with the anti-CD20 monoclonal antibody rituximab, commonly used in combination therapy for MCL. Gene expression profiling analysis in harvested tumors suggested that acadesine modulates immune response, actin cytoskeleton organization and metal binding, pointing out a substantial impact on metabolic processes by the nucleoside analog. Rituximab also induced changes on metal binding and immune responses.The combination of both drugs enhanced the gene signature corresponding to each single agent, showing an enrichment of genes involved in inflammation, metabolic stress, apoptosis and proliferation. These effects could be important as aberrant apoptotic and proinflammatory pathways play a significant role in the pathogenesis of MCL. In summary, our results suggest that acadesine exerts a cytotoxic effect in MCL in combination with rituximab, by decreasing the proliferative and survival signatures of the disease, thus supporting the clinical examination of this strategy in MCL patients.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Lymphoma, Mantle-Cell/drug therapy , Ribonucleosides/pharmacology , Aminoimidazole Carboxamide/administration & dosage , Aminoimidazole Carboxamide/pharmacology , Animals , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibodies, Monoclonal, Murine-Derived/immunology , Antigens, CD20/immunology , Apoptosis/drug effects , Cell Line, Tumor , Cohort Studies , Disease Models, Animal , Drug Synergism , Female , Humans , Mice , Mice, SCID , Random Allocation , Ribonucleosides/administration & dosage , Rituximab , Xenograft Model Antitumor Assays
9.
Haematologica ; 98(11): 1739-47, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23850807

ABSTRACT

Phosphatidylinositol-3-kinase pathway is constitutively activated in chronic lymphocytic leukemia mainly due to microenvironment signals, including stromal cell interaction and CXCR4 and B-cell receptor activation. Because of the importance of phosphatidylinositol-3-kinase signaling in chronic lymphocytic leukemia, we investigated the activity of the NVP-BKM120, an orally available pan class I phosphatidylinositol-3-kinase inhibitor. Sensitivity to NVP-BKM120 was analyzed in chronic lymphocytic leukemia primary samples in the context of B-cell receptor and microenvironment stimulation. NVP-BKM120 promoted mitochondrial apoptosis in most primary cells independently of common prognostic markers. NVP-BKM120 activity induced the blockage of phosphatidylinositol-3-kinase signaling, decreased Akt and FoxO3a phosphorylation leading to concomitant Mcl-1 downregulation and Bim induction. Accordingly, selective knockdown of BIM rescued cells from NVP-BKM120-induced apoptosis, while the kinase inhibitor synergistically enhanced the apoptosis induced by the BH3-mimetic ABT-263. We also found NVP-BKM120 to inhibit B-cell receptor- and stroma-dependent Akt pathway activation, thus sensitizing chronic lymphocytic leukemia cells to bendamustine and fludarabine. Furthermore, NVP-BKM120 down-regulated secretion of chemokines after B-cell receptor stimulation and inhibited cell chemotaxis and actin polymerization upon CXCR4 triggering by CXCL12. Our findings establish that NVP-BKM120 effectively inhibits the phosphatidylinositol-3-kinase signaling pathway and disturbs the protective effect of the tumor microenvironment with the subsequent apoptosis induction through the Akt/FoxO3a/Bim axis. We provide here a strong rationale for undertaking clinical trials of NVP-BKM120 in chronic lymphocytic leukemia patients alone or in combination therapies.


Subject(s)
Aminopyridines/pharmacology , Apoptosis Regulatory Proteins/physiology , Forkhead Transcription Factors/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Membrane Proteins/physiology , Morpholines/pharmacology , Oncogene Protein v-akt/physiology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins/physiology , Tumor Microenvironment/physiology , Aged , Aged, 80 and over , Aminopyridines/therapeutic use , Bcl-2-Like Protein 11 , Cell Movement/drug effects , Cell Movement/physiology , Coculture Techniques , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Forkhead Box Protein O3 , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Middle Aged , Morpholines/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Tumor Microenvironment/drug effects
10.
Clin Cancer Res ; 19(3): 586-97, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23231952

ABSTRACT

PURPOSE: We evaluated the antitumoral properties of the multikinase inhibitor sorafenib in mantle cell lymphoma (MCL), an aggressive B lymphoma for which current therapies have shown limited efficacy. EXPERIMENTAL DESIGN: Sensitivity to sorafenib was analyzed in MCL cell lines and primary samples in the context of BCR and microenvironment simulation. Sorafenib signaling was characterized by quantitative PCR, Western blotting, immunofluorescence, and protein immunoprecipitation. Migration analysis included flow cytometric counting, actin polymerization assays, and siRNA-mediated knockdown of focal adhesion kinase (FAK). In vivo antitumor effect of sorafenib and bortezomib was analyzed in an MCL xenograft mouse model. RESULTS: Sorafenib rapidly dephosphorylates the BCR-associated kinases, Syk and Lyn, as well as FAK, an Src target involved in focal adhesion. In this line, sorafenib displays strong synergy with the Syk inhibitor, R406. Sorafenib also blocks Mcl-1 and cyclin D1 translation, which promotes an imbalance between pro- and antiapoptotic proteins and facilitates Bax release from cyclin D1, leading to the induction of mitochondrial apoptosis and caspase-dependent and -independent mechanisms. Moreover, sorafenib inhibits MCL cell migration and CXCL12-induced actin polymerization. FAK knockdown partially prevents this inhibitory effect, indicating that FAK is a relevant target of sorafenib. Furthermore, sorafenib enhances the antitumoral activity of bortezomib in an MCL xenograft mouse model as well as overcomes stroma-mediated bortezomib resistance in MCL cells. CONCLUSION: We show for the first time that sorafenib interferes with BCR signaling, protein translation and modulates the microenvironment prosurvival signals in MCL, suggesting that sorafenib, alone or in combination with bortezomib, may represent a promising approach to treat patients with MCL.


Subject(s)
Cell Movement/drug effects , Drug Resistance, Neoplasm , Lymphoma, Mantle-Cell/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Protein Biosynthesis/drug effects , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/drug effects , Actins/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Cell Line, Tumor , Chemokine CXCL12/metabolism , Cyclin D1/genetics , Disease Models, Animal , Female , Humans , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/pathology , Mice , Myeloid Cell Leukemia Sequence 1 Protein , Niacinamide/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Multimerization , Proto-Oncogene Proteins c-bcl-2/genetics , Sorafenib , Stromal Cells , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...