BIC and miR-155 Are Highly Expressed in Hodgkin, Primary Mediastinal and Diffuse Large B Cell Lymphomas.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 970-970 ◽  
Author(s):  
Joost Kluiver ◽  
Sibrand Poppema ◽  
Debora de Jong ◽  
Tjasso Blokzijl ◽  
Geert Harms ◽  
...  

Abstract In a previous study we demonstrated high expression of the non-coding BIC gene in the vast majority of Hodgkin’s lymphomas (HLs). It is now known that BIC is a primary microRNA (pri-miRNA) that can be processed to miRNA-155. We analyzed HL and NHL cell lines and tissue samples to determine miR-155 expression levels. High levels of miR-155 could be demonstrated in all HL tissue samples and most cell lines. Most NHL subtypes were negative for BIC as determined by RNA-ISH. However, 8/8 cases of primary mediastinal B cell lymphoma (PMBL) showed BIC staining in 12% of the tumor cells. QRT-PCR analysis confirmed high relative BIC levels (35). 12/18 cases of DLBCL showed a variable BIC staining with on average 7% of positive cells. Interestingly, activated B cell-like (ABC-like) DLBCL showed on average 12% of tumor cells positive for BIC while germinal centre B cell-like (GCB-like) DLBCL only showed on average 4% of cells positive for BIC. Differences in BIC expression levels were confirmed by qRT-PCR analysis revealing average BIC levels of 22 for ABC-like and 12 for GCB-like DLBCL. Northern blot analysis showed expression of miR-155 in all DLBCL and PMBL derived cell lines and tissue samples analyzed. In summary, we demonstrate expression of pri-miRNA BIC and its derivative miR-155 in HL, PMBL and DLBCL. In DLBCL, BIC expression is most pronounced in the ABC-like phenotype, possibly as a result of NF-κB activation.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4525-4525
Author(s):  
Bernardo Martinez-Miguel ◽  
Melisa A. Martinez-Paniagua ◽  
Sara Huerta-Yepez ◽  
Rogelio Hernandez-Pando ◽  
Cesar R. Gonzalez-Bonilla ◽  
...  

Abstract The interaction between CD40, a member of the tumor necrosis factor super family, and its ligand CD154 is essential for the development of humoral and cellular immune responses. Selective inhibition or activation of this pathway forms the basis for the development of new therapeutics against immunologically-based diseases and malignancies. CD40 is expressed primarily on dendritic cells, macrophages and B cells. Engagement of CD40-CD154 induces activation and proliferation of B lymphocytes and triggers apoptosis of carcinoma and B lymphoma cells. Agonist CD40 antibodies mimic the signal of CD154-CD40 ligation on the surface of many tumors and mediate a direct cytotoxic effect in the absence of immune accessory molecules. CD40 expression is found on nearly all B cell malignancies. Engagement of CD40 in vivo inhibits B cell lymphoma xenografts in immune compromised mice. Several clinical trials have been reported targeting CD40 in cancer patients using recombinant CD154, mAbs and gene therapy, which were well tolerated and resulted in objective tumor responses. In addition to these therapies, CD54 mimetics have been considered with the objective to augment and potentiate the direct cytotoxic anti-tumor activity and for better accessibility to tumor sites. This approach was developed by us and we hypothesized that the genetic engineering of a fusion protein containing a CD154 peptide mimetic may be advantageous in that it may have a better affinity to CD40 on B cell malignancies and trigger cell death and the partner may be a carrier targeting other surface molecules expressed on the malignant cells. This hypothesis was tested by the development of a gene fusion of Salmonella typhi OmpC protein expressing the CD154 Trp140-Ser149 amino acid strand (Vega et al., Immunology2003; 110: 206–216). This OmpC-CD154p fusion protein binds CD40 and triggers the CD40 expressing B cells. In this study, we demonstrate that OmpC-CD154p treatment inhibits cell growth and proliferation of the B-NHL cell lines Raji and Ramos. In addition, significant apoptosis was achieved and the extent of apoptosis was a function of the concentration used and time of incubation. The anti-tumor effect was specific as treatment with OmpC alone had no effect. These findings establish the basis of the development of new fusion proteins with dual specificity (targeting the tumor cells directly or targeting the tumor cells and immune cells). The advantages of this approach over conventional CD40-targeted therapies as well as the mechanism of OmpC-CD154p-induced cell signaling and cell death will be presented.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2434-2434
Author(s):  
Soranobu Ninomiya ◽  
Leslie E Huye ◽  
Barbara Savoldo ◽  
Gianpietro Dotti ◽  
Helen E. Heslop ◽  
...  

Abstract Indoleamine 2,3-dioxygenase (IDO) is an intracellular enzyme that mediates the metabolism of tryptophan to kynurenines, which have an inhibitory activity on immune cells. IDO-positive tumors thus establish a microenvironment in which NK and T cells are inactivated, and high IDO expression by tumor cells and high serum kynurenine levels correlate with poor prognosis in diffuse large B-cell lymphoma patients. CD19-specific chimeric antigen receptor T cell (CART) therapy is a promising new approach against B-cell malignancies but not every tumor responds. To determine whether the presence of IDO in tumors affects CART activity, we first investigated the expression of IDO by the human B-cell lymphoma cell lines Raji, Daudi, BJAB and Jeko-1. We found that only Jeko-1 expresses IDO and produces kynurenine natively. IDO was not expressed by the other cell lines, even after exposure to IFNγ, a known IDO inducer. Based on these results, we chose Raji as a baseline IDO-negative cell line and made an IDO-positive Raji clone by retroviral transduction with human IDO cDNA (Raji-IDO); a clone transduced with an empty vector served as control (Raji-control). We injected SCID mice subcutaneously in opposite flanks with luciferase-transduced Raji-control and Raji-IDO cells. Seven days later, we injected human non-transduced T cells (NTs) or CARTs intravenously. In the NT group, tumors had similar growth on both sides. In contrast, in the CART group, Raji-control tumors had significantly slower growth than Raji-IDO tumors (3.1 ± 1.1×108 and 20 ± 7.3×108 bioluminescence units [BU] at day 7, respectively, P = 0.03). We also found that CARTs significantly inhibited Raji-control tumor growth (NT: 27 ± 6.8×108 vs CART: 3.1 ± 1.1×108 BU at day 7, P = 0.02), but did not affect Raji-IDO tumors (NT: 24 ± 5.4×108 vs CART: 20 ± 7.3×108 BU at day 7, P = 0.35). In another experiment, Raji-IDO cells were injected subcutaneously, and mice were treated with an IDO inhibitor (1-methyl-tryptophan, 1-MT), CARTs, or both. The combination treatment produced significantly better tumor control than either single therapy (1-MT: 45 ± 6.8×108 and CART: 22 ± 4.6×108 vs both: 8.2 ± 3×108 BU at day 7, P = 0.001 and 0.04, respectively). Thus, the IDO inhibitor protects CARTs against the deleterious effects of IDO-positive tumors. To investigate potential mechanisms for CART inhibition by IDO, we assessed the effect of kynurenine and found that even low concentrations (12.5 µM) inhibited CART proliferation in response to IL-2, IL-7, IL-15 or CD19-positive targets, although there was no effect on proliferation of B-cell lymphoma cell lines at this kynurenine concentration. CART apoptosis was also increased by kynurenine (8.6 ± 0.6%, 13.9 ± 2.2%, and 33.5 ± 10.6% annexin V-positive cells with 0, 12.5, or 25 µM kynurenine). In coculture of CARTs with wild-type Raji cells, the latter were eliminated by day 6 in the absence of kynurenine, but increased in numbers (in parallel with a decrease in CARTs) in its presence. Kynurenine also inhibited the release of IFNγ (13,143 ± 848 pg/mL vs 2,663 ± 1,873 pg/mL, P = 0.02) and IL-2 (718 ± 355 pg/mL and 199 ± 165 pg/mL, P = 0.03) by CARTs. Expression of granzyme B, PD-1 and CTLA-4 on CARTs was not significantly affected. Fludarabine has been reported to downregulate IDO expression in tumors and this drug is used in many lymphodepleting regimens that are administered before CART infusion in an effort to augment the efficacy of these therapies. However, the beneficial mechanism of lymphodepleting chemotherapy drugs is not fully understood. Therefore, we also measured the effect of fludarabine and mafosfamide (a cyclophosphamide analog) on IDO expression by Jeko-1 cells. We found that both drugs downregulate IDO expression by Jeko-1, even in the presence of IFNγ. In summary, expression of IDO by tumor cells inhibits CART activity, likely because kynurenine is produced and has negative effects on proliferation and cytokine secretion by CARTs. Fludarabine and cyclophosphamide downregulate IDO expression in tumors and this effect may contribute to the benefits of lymphodepletion before CART therapy. Direct IDO inhibitors may further improve clinical CART activity against IDO-positive tumors. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 33 (12) ◽  
pp. 2407-2421 ◽  
Author(s):  
Lucia Mundo ◽  
Leonardo Del Porro ◽  
Massimo Granai ◽  
Maria Chiara Siciliano ◽  
Virginia Mancini ◽  
...  

AbstractThe Epstein–Barr virus (EBV) is linked to various B-cell lymphomas, including Burkitt lymphoma (BL), classical Hodgkin lymphoma (cHL) and diffuse large B-cell lymphoma (DLBCL) at frequencies ranging, by routine techniques, from 5 to 10% of cases in DLBCL to >95% in endemic BL. Using higher-sensitivity methods, we recently detected EBV traces in a few EBV-negative BL cases, possibly suggesting a “hit-and-run” mechanism. Here, we used routine and higher-sensitivity methods (qPCR and ddPCR for conserved EBV genomic regions and miRNAs on microdissected tumor cells; EBNA1 mRNA In situ detection by RNAscope) to assess EBV infection in a larger lymphoma cohort [19 BL, 34 DLBCL, 44 cHL, 50 follicular lymphomas (FL), 10 T-lymphoblastic lymphomas (T-LL), 20 hairy cell leukemias (HCL), 10 mantle cell lymphomas (MCL)], as well as in several lymphoma cell lines (9 cHL and 6 BL). qPCR, ddPCR, and RNAscope consistently documented the presence of multiple EBV nucleic acids in rare tumor cells of several cases EBV-negative by conventional methods that all belonged to lymphoma entities clearly related to EBV (BL, 6/9 cases; cHL, 16/32 cases; DLBCL, 11/30 cases), in contrast to fewer cases (3/47 cases) of FL (where the role of EBV is more elusive) and no cases (0/40) of control lymphomas unrelated to EBV (HCL, T-LL, MCL). Similarly, we revealed traces of EBV infection in 4/5 BL and 6/7 HL cell lines otherwise conventionally classified as EBV negative. Interestingly, additional EBV-positive cases (1 DLBCL, 2 cHL) relapsed as EBV-negative by routine methods while showing EBNA1 expression in rare tumor cells by RNAscope. The relapse specimens were clonally identical to their onset biopsies, indicating that the lymphoma clone can largely loose the EBV genome over time but traces of EBV infection are still detectable by high-sensitivity methods. We suggest EBV may contribute to lymphoma pathogenesis more widely than currently acknowledged.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 631-631 ◽  
Author(s):  
Linfeng Chen ◽  
Yansheng Hao ◽  
Jing Ouyang ◽  
Bjoern Chapuy ◽  
Donna S. Neuberg ◽  
...  

Abstract B-cell receptor (BCR) signaling pathway components represent promising treatment targets in multiple B-cell tumors including diffuse large B-cell lymphoma (DLBCL). BCR signaling activates proximal pathway components such as the spleen tyrosine kinase (SYK) and downstream effectors including PI3K/AKT and NF-κB. In recent studies, we characterized distinct SYK/PI3K/AKT-dependent viability pathways in BCR DLBCL cell lines and primary tumors with high- or low-baseline NF-κB activity and defined additional SYK/PI3K/AKT-dependent processes essential for BCR DLBCLs with low- or high-baseline NF-κB (Chen et al Cancer Cell 2013; 23:826-838). For example, SYK/PI3K/AKT-dependent cholesterol biosynthesis was identified as a feed-forward mechanism of preserving the integrity of BCRs in lipid rafts in all BCR-dependent DLBCLs. In the current study, we sought to identify biomarkers of intact BCR signaling in DLBCLs with low or high baseline NF-κB. We found highly significant transcriptional upregulation of CXCR4 in all BCR-dependent DLBCLs treated with a chemical SYK inhibitor (R406). These results were of particular interest because BCR signaling inhibits stromal cell-derived factor (SDF)-1α chemotaxis and promotes intermobilization of the SDF-1α receptor, CXCR4. To expand on our findings, we analyzed CXCR4 transcript abundance in a large panel of cell lines including BCR-dependent/ low NF-κB (GC type) and BCR-dependent/ high NF-κB (ABC type) and BCR-independent/ OxPhos DLBCLs. CXCR4 transcript abundance was significantly and selectively upregulated in all BCR-dependent DLBCL lines assessed by qRT-PCR following chemical SYK inhibition. Consistent with the qRT-PCR results, SYK blockade also selectively upregulated cell surface CXCR4 expression in all BCR-dependent DLBCLs. Similar results were obtained following SYK knockdown with multiple independent shRNAs; CXCR4 transcript abundance was significantly and selectively upregulated in all BCR-dependent/ low NF-κB (GCB) DLBCLs and BCR-dependent/ high NF-κB (ABC) DLBCLs but was not altered in BCR-independent/ OxPhos tumors. Following SYK blockade, we also observed significant CXCR4 induction (by qRT-PCR) in multiple primary BCR-dependent DLBCLs (NF-κB low and NF-κB high) and unchanged CXCR4 levels in primary BCR-independent tumors. These data confirm that the observations regarding SYK blockade and CXCR4 induction in BCR-dependent DLBCL cell lines translated to primary tumors. Our recent studies indicate that all BCR-dependent DLBCLs exhibit SYK/PI3K/AKT signaling and modulation of FOXO1 regardless of baseline NF-κB levels. Given the recent identification of CXCR4 as a FOXO1 target, we asked whether modulation of CXCR4 in DLBCLs was via a PI3K/AKT pathway. In BCR-dependent DLBCLs with low or high baseline NF-κB, chemical PI3K inhibition (LY294002) phenocopied chemical SYK blockade (R406) with highly significant transcriptional upregulation of CXCR4. Moreover, in BCR-dependent DLBCL cell lines, enforced expression of a constitutively active form of AKT (myrAKT) abrogated CXCR4 upregulation resulting from SYK blockade. These data indicate that in BCR-dependent DLBCLs, CXCR4 is modulated by a SYK/PI3K/AKT-dependent pathway. To assess the functional significance of CXCR4 upregulation in BCR-dependent DLBCLs, we performed a transwell chemotaxis assay and evaluated migration toward a SDF-1α gradient. Following chemical SYK inhibition, all BCR-dependent DLBCLs exhibited significantly increased migration toward the CXCR4 ligand, SDF-1α, whereas the BCR-independent/ OxPhos DLBCLs were unaffected. The enhanced SDF-1α dependent migration was abrogated when the chemotaxis assay was performed in the presence of the specific CXCR4 inhibitor, AMD3100, confirming the specificity of the observed effect. Therefore, in BCR-dependent DLBCLs with low or high baseline NF-kB, SYK/PI3K inhibition is associated with significant and functionally relevant upregulation of CXCR4. For these reasons, CXCR4 may serve as a robust biomarker to assess the integrity of the BCR pathway and evaluate the efficacy of BCR inhibition in DLBCL. Disclosures: Off Label Use: R406 is not get approved, but is being evaluated in DLBCL and CLL.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5424-5424
Author(s):  
Nicolle H Rekers ◽  
Laura M Moesbergen ◽  
Nathalie J Hijmering ◽  
Wim Vos ◽  
Joost Oudejans ◽  
...  

Abstract Diffuse large B-cell lymphoma (DLBCL) remains eventually fatal in 30-40% of the patients, despite intensive chemotherapy (CHOP) in combination with rituximab. A subgroup of chemotherapy-refractory DLBCL is characterized by high expression levels of both pro- and anti-apoptotic genes, including MCL-1. Alternative splicing of the MCL-1 gene results in a Bcl-2-like anti-apoptotic MCL-1L protein and a BH3-only pro-apoptotic MCL-1S protein. In the present study, we investigated if a switch in alternative splicing of MCL-1 is involved in apoptosis-resistance in primary lymphoma cells of 20 DLBCL patients and 5 DLBCL cell lines. RT-MLPA analysis revealed that MCL-1L and MCL-1S are both expressed in all tested DLBCL samples and DLBCL cell lines, however expression levels varied strongly. An imbalance between the expression levels of MCL-1L and MCL-1S to an anti-apoptotic status was observed in DLBCL patient cells and DLBCL cell lines, especially in activated B-cell like (ABC)-DLBCL, compared to tonsillar germinal center B-cells. MCL-1 mRNA expression was confirmed at protein level using immunohistochemistry and western blot analysis. Co-immunoprecipitation demonstrated that MCL-1L inhibited apoptosis by binding of Bak in MCL-1L positive DLBCL cell lines. Knockdown of MCL-1L with siRNA analysis resulted in induction of apoptosis in both GCB- and ABC-DLBCL cell lines and also in increased sensitivity to the conventional chemotherapeutical drugs etoposide. Downregulation of MCL-1L using flavopiridol induced apoptotic cell death of MCL-1L-positive DLBCL cells with low Bcl-2 expression. In summary, a switch in alternative splicing of MCL-1 occurs in a subgroup of DLBCL leading to an increase in the level of anti-apoptotic MCL-1L that contributes to therapy-resistance. These preclinical data suggest that targeting of MCL-1L might be a therapeutic option for MCL-1L positive DLBCL. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2357-2357
Author(s):  
Britta Höhn ◽  
Harris S. Soifer ◽  
Stephen J. Forman ◽  
John J. Rossi

Abstract Non-Hodgkin’s lymphomas comprise a group of heterogeneous lymphoid malignancies that represent the fifth most common form of cancer in the United States. A hallmark of many types of B-cell lymphomas is the constitutive expression of oncogenes such as the transcription factors Bcl-6, STAT3 and c-Myc and the anti-apoptotic protein Bcl-2. Over expression of these genes causes uncontrolled proliferation and survival of malignant cells, making knockdown of these genes by RNA interference (RNAi) a rational strategy for therapeutic intervention. RNAi is a conserved endogenous mechanism in which small interfering RNAs (siRNAs) suppress target-specific gene expression by promoting mRNA degradation. We have designed potent Dicer-substrate siRNAs using different computer algorithms to predict accessible target sites in the mRNAs of B-cell lymphoma oncogene targets. The Dicer-substrate 27mers are designed asymmetrically, so that Dicer processing yields a predicted 21mer siRNA duplex for entry into RNA induced silencing complex (RISC). Dicer-substrate siRNAs show improved efficacy at lower concentrations compared with conventional 21mer siRNAs, suggesting the reduction of potential off-target effects. In addition, we have designed bifunctional siRNA duplexes that contain two fully target-complimentary antisense strands against two different target mRNAs, but that are only partially complementary to each other. In vitro cleavage assays indicate that our bifunctional siRNAs have sufficient complementarity to form stable duplexes and can be processed into smaller molecules by recombinant Dicer. When delivered to the Burkitt’s lymphoma cell line Raji by electroporation, the most effective siRNAs reduced target mRNA levels by ∼80% as determined by quantitative RT-PCR and immunoblot analysis. Silencing of transcription factors affected the expression of downstream target genes, indicating a relevant effect on growth on survival of lymphoma cells through oncogene down regulation by RNAi. One concern with RNAi-mediated therapy is the possible recognition of siRNA duplex by cell’s own response to double-stranded RNA (dsRNA) that could trigger an unwanted interferon response. To determine whether our Dicer-substrate siRNAs cause an interferon response, we monitored induction of the cellular dsRNA pathway by measuring gene expression of p56, OAS1 and interferon by quantitative RT-PCR after transfection of siRNAs in different cell lines. None of the analyzed siRNAs show a significant increase in the expression of interferon pathway related genes indicating that our selected siRNAs are powerful silencers of gene expression without inducing an interferon response. In future studies, these new identified siRNAs will be incorporated in nanoparticles or attached to antibodies / aptamers for cell-specific delivery to lymphoma cell lines to evaluate their potential alone or in combination with chemotherapeutic drugs in therapy for lymphoma.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 465-465
Author(s):  
Jianfei Qian ◽  
Sungyoul Hong ◽  
Liang Zhang ◽  
Yuhuan Zheng ◽  
Haiyan Li ◽  
...  

Abstract Abstract 465 Immunotherapy may complement the current treatments for lymphomas. The lack of suitable shared lymphoma-associated antigens limits its applicability. Therefore, identification and utilization of novel and more potent tumor-associated antigens, particularly those shared among patients, are urgently needed to improve the efficacy of immunotherapy in the diseases. Recent studies have shown that Dickkopf-1 (DKK1), a secreted protein and Wnt signaling pathway inhibitor, is highly expressed by myeloma and other tumor cells, and is absent from normal tissues and organs except placenta and prostate. In the present study we demonstrated that DKK1 is also overexpressed in mantle cell lymphoma (MCL) and diffuse large B-cell lymphoma (DLBCL). Using DKK1 peptide-pulsed dendritic cells (DCs), we successfully generated HLA-A*0201+ DKK1-specific CTL lines and clones in vitro. These CTLs effectively lysed DKK1+/HLA-A*0201+ lymphoma cell lines Jeko-1 and Granta 519 cells, but not DKK1-/HLA-A*0201+ BJAB, RL and Mino cells nor DKK1+/HLA-A*020- CA46 and Daudi cells. Furthermore, the T-cell clones efficiently killed DKK1+/HLA-A*0201+ primary B-cell lymphoma cells from patients but not lymphoma cells from DKK1–/HLA-A*0201+ patients. HLA-ABC or HLA-A*0201 blocking mAbs significantly inhibited T cell-mediated cytotoxicity against peptide-pulsed T2 cells (P < .01, compared with medium control). No inhibitory effect was observed with mAb against HLA-DR and isotype control IgG. The results indicate that the cytotoxicity was attributed to MHC class I and more specifically, HLA-A*0201-restricted CD8+ CTLs. The CTLs did not kill DKK1–/HLA-A*0201+ DCs, B cells, or PBMCs, These results suggest that the CTLs recognized DKK1 peptides that are naturally processed and presented in the context of HLA-A*0201 molecules on lymphoma cells. To determine the in vivo antitumor activity, NOD-SCID and SCID-hu mice were used for lymphoma cell lines and primary lymphoma cells, respectively. Mice were treated with DKK1-specific CTLs after tumor established in NOD-SCID and SCID-hu mice. Control mice were treated with naïve CD8+ T cells or PBS alone. Tumor burden was measured according to levels of circulating human B2M, and survival rates were determined. Low levels (< 50 ng/ml) of circulating human B2M were detected in group treated DKK1-specific CTLs, while high levels (≥ 150 ng/ml) of circulating human B2M were detected in control mice. In SCID-hu model, X-ray examination showed that established tumors were eradicated in 60% mice treated with DKK1-specific CTLs, while large tumor burdens were found in all control mice. In NOD-SCID model, 40% of mice survived with the treatment of DKK1-specific CTLs. TUNEL assay further confirmed that tumor cells were lysed by DKK1-specific CTLs not naïve CD8+ T cells. These results indicate that DKK1-specific CTLs are able to eradicate established, patient-derived primary B- cell lymphoma in the hosts and adoptive transfer of DKK1-specific CTLs may be used for B-cell lymphoma therapy. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3727-3727 ◽  
Author(s):  
Umeer Ashraf ◽  
Myron S. Czuczman ◽  
Cory Marvis ◽  
John Gibbs ◽  
Francisco J Hernandez-Ilizaliturri

Abstract Abstract 3727 Poster Board III-663 Deacetylases (DACs) are enzymes that remove the acetyl groups from target proteins, leading to regulation of gene transcription and other cellular processes. Entinostat (SNDX-275) is a novel and potent DAC inhibitor that is selective for class I DACs and is currently undergoing pre-clinical and clinical testing in Hodgkin lymphoma (HL). Potent synergistic anti-tumor activity has been observed by combining less potent DAC inhibitors with bortezomib in pre-clinical models. In our efforts to develop more therapeutic options for refractory/resistant B-cell lymphoma, we evaluated the effects of Eentinostat as a single agent and in combination with bortezomib against B-cell non-Hodgkin's lymphoma (NHL) cell lines and primary NHL cells. Studies were conducted in a panel of 12 NHL cell lines representing various subtypes of B-cell lymphoma (i.e. DLBCL/ABC, DLBCL/GCB, Burkitt's, transformed and MCL), which included: rituximab-[chemotherapy]-sensitive cell lines (RSCL, Raji, RL and DHL-4), rituximab-[chemotherapy]-resistant cell lines (RRCL, Raji-4RH, Raji-2R, RL-4RH, and DHL-4 4RH), and primary lymphoma cells isolated from patients with various subtypes of NHL and HL. Patient-derived tumor cells were isolated from fresh specimens by negative selection using magnetic beads. NHL cells and patient-derived primary cells were exposed to entinostat at different doses (0.01 to 100uM) either alone or in combination with CDDP (1 to 100μM), doxorubicin (4 to 16μM), vincristine (1 to 5μM), or bortezomib (1 to 10nM). Anti-tumor activity was measured after a 24 or 48 hr incubation. In cell lines, changes in mitochondrial potential and cell proliferation were determined by alamar blue reduction using a kinetic assay measuring activity at 4 hr intervals for 24 and 48 hrs. For patient-derived primary NHL cells, changes in ATP content (apoptosis) was determined using the cell titer glow assay. Entinostat was highly active in all the cell lines tested including rituximab-[chemotherapy]-resistant cell lines. The IC50 of Entinostat in the majority of the cells tested was 0.5 to 5uM at 48 hrs. Similar findings were observed in primary tumor cells derived from lymphoma patients. In addition, synergistic activity was observed by combining entinostat and bortezomib in both NHL cell lines, as well as in primary NHL/HL tumor specimens. A lesser degree of augmented anti-tumor activity was also observed when entinostat was combined with cisplatin or doxorubicin (but not vincristine). In summary, our data suggests that entinostat is a novel and potent DAC inhibitor with a wide therapeutic spectrum. Entinostat is capable of inducing cell death against various subtypes of B-cell lymphoma cell lines including RSCL, RRCL, as well as patient-derived primary tumor cells and augments the anti-tumor effects of bortezomib and other chemotherapeutic agents. Given the isoform selectivity of entinostat, the results indicate that HDAC1 and 2 may be the key targets of DAC inhibitors in HL and NHL cells. Ongoing studies are evaluating the mechanisms responsible for the synergistic effects of entinostat plus chemotherapy and will be updated at the annual meeting. Current findings strongly suggest that entinostat added to bortezomib and/or other chemo agents may become a novel and potent strategy in the treatment of aggressive and indolent NHL and HL in the future. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2715-2715
Author(s):  
Francisco J Hernandez-Ilizaliturri ◽  
Cory Marvis ◽  
Ilir Maraj ◽  
Mohammad M Chisti ◽  
John Gibbs ◽  
...  

Abstract Abstract 2715 Poster Board II-691 Deacetylases (DACs) are enzymes that remove the acetyl groups from target proteins [histones (class I) and non-histones (class II)], leading to regulation of gene transcription and other cellular processes. LBH589 is a novel and potent DAC class I and II inhibitor undergoing pre-clinical and clinical testing. In order to better characterize the role of DAC inhibitors in the treatment of refractory/resistant B-cell lymphomas we studied the anti-tumor effect that LBH589 had when used with chemotherapy agents and anti-CD20 monoclonal antibodies against a panel of rituximab-[chemotherapy]-sensitive cell lines (RSCL), rituximab-[chemotherapy]-resistant cell lines (RRCL), and primary lymphoma cells isolated from patients with treatment-naïve or refractory/relapsed B-cell lymphoma. Non-Hodgkin's lymphoma (NHL) cell lines were exposed to the following chemotherapy agents or monoclonal antibodies: CDDP, doxorubicin, vincristine, bortezomib versus rituximab or veltuzumab (or isotype control), alone or in combination with LBH589. In dose-sequence studies the treatment with LBH589 preceded or followed in vitro exposure to the chemotherapy agent or the monoclonal antibody by 24 hrs. Changes in mitochondrial potential were determined by alamar blue reduction using a kinetic assay. Patient-derived primary tumor cells (N=25) were exposed to either LBH589 (2-25uM), bortezomib (1 to 10nM) or both. Changes in ATP content were determined by cell titer glow assay. RNA was isolated from NHL cell lines exposed to LBH859 or bortezomib and changes in gene expression of the Bcl-2 family members were determined by qualitative polymerase chain reaction (PCR). LBH589 was active as a single agent against RSCL, RRCL or patient-derived primary tumor cells. In addition, Bcl-XL gene down-regulation was observed following exposure to LBH859. On the other hand, upregulation of Bak and downregulation of Mcl-1 were observed following proteasome inhibition. Synergistic activity was observed by combining LBH589 and chemotherapy agents, bortezomib or either of the two anti-CD20 mAbs studied. In tumor-derived primary cells from lymphoma patients, the combination of LBH589 and bortezomib resulted in significant anti-tumor activity in follicular, Hodgkin and diffuse large B-cell lymphoma. The sequence of administration impacted the degree of antitumor activity observed (ie in general, exposure of tumors cells initially to LBH589, followed by exposure to chemo/mAbs was associated with the greatest degree of anti-tumor activity). Our data suggests that LBH589 is active against various RSCL, RRCL and patient-derived primary tumor cells. Our findings strongly suggest that LBH589 added to anti-CD20 and/or chemotherapy results in a novel and potent treatment strategy against B-cell lymphoma. Research, in part, supported as part of a subproject on NIH PO1 grant CA103985-1 awarded to the Garden State Cancer Center, Belleville, NJ and NHI R-01 grant R01 CA136907-01A1 awarded to Roswell Park Cancer Institute Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4450-4450
Author(s):  
Satoru Kobayashi ◽  
Tomohiko Taki ◽  
Yoshiaki Chinen ◽  
Yasuhiko Tsutsumi ◽  
Muneo Ohshiro ◽  
...  

Abstract Abstract 4450 In B-cell malignancies, the genes implicated in B-cell differentiation, germinal center formation, apoptosis, and cell cycle regulation are juxtaposed to immunoglobulin loci through chromosomal translocations. In the present study, we have identified BTB and CNC homology 2 (BACH2) as a novel translocation partner gene of the immunoglobulin heavy chain (IGH) locus, resulting in chimeric Cδ-BACH2 gene in a patient with MYC-IGH-positive highly aggressive B-cell lymphoma/leukemia carrying der(14)t(8;14) and del(6)(q15). A 71-year-old male was diagnosed as having highly aggressive B-cell lymphoma/leukemia. SKY analysis revealed the representative karyotype of tumor cells as 45,X,-Y,der(3)t(3;X)(p21.2;q24),del(6)(q15),der(14)t(8;14)(q24;q32),del(16)(q22),der(20) t(3;20)(q21;p13). FISH and long-distance PCR analyses identified a fusion of MYC with Cγ. FISH analysis also detected a small IGH signal on del(6), and a VH on del(6)(q15). Genome copy number analysis using SNP-array detected an approximately 60Mb deletion in 6q15–25 region, and its centromeric breakpoint within BACH2 gene. The cDNA bubble PCR using BACH2 primers detected multiple PCR products, and sequence analysis has revealed that one of the products contained a sequence of the first exon of IGHCδ fused to 5’ untranslated region of BACH2 exon 2. Genomic fusion point of Cδ-BACH2, was within intron 1 of Cδ and intron 1 of BACH2. Cδ-BACH2 fusion transcript consisted of exon 1 of Cδ and exons 2 to 9 of BACH2, indicating that whole coding region of BACH2 was fused to Cδ. This suggested that Cδ-BACH2 fusion cause constitutive activation of BACH2. We next analyzed the expression levels of BACH2, MYC, PRDM1, and IRF4 genes in the patient using real-time PCR and compared them with those of several hematologic malignancy cell lines, including 14 non-Hodgkin's lymphoma (NHL), 10 multiple myeloma (MM), and 3 B-lineage acute lymphoblastic leukemia, and 3 EB-virus transformed B-cell lines from normal healthy volunteers. IGH-MYC-positive MM cell lines showed increased levels of MYC expression compared with the other cell lines. The MYC expression level in our patient was lower than those in IGH-MYC-positive MM cell lines; however, it was similar to 5 IGH-MYC-positive NHL cell lines [3 Burkitt lymphoma (BL) and 2 diffuse large B-cell lymphoma (DLBCL)]. On the other hand, BACH2 expression levels were higher in NHL cell lines, especially in BL cell lines, but not correlated with the IGH-MYC translocation status. Our IGH-BACH2-positive case also showed a higher level of BACH2 expression. The expression levels of IRF4 and PRDM1 were higher in MM cell lines than in other cell lines. PRDM1 is activated by MYC through IRF4, and the MYC is negatively regulated by activated PRDM1. In MM cell lines with IGH-MYC translocation, MYC was highly expressed regardless of high expression of PRDM1, indicating that MYC activated by IGH translocation could not be inhibited by PRDM1. Unexpectedly, the expression levels of PRDM1 and IRF4 were very low in NHL including our patient, suggesting that the regulation of MYC in NHL is different from that in MM. BACH2 is a B cell-specific transcription repressor, and is specifically required for class switch recombination, somatic hypermutation, and germinal center formation. One of the target genes of BACH2 is PRDM1 at 6q21-q22.1 that is required for plasma cell differentiation. In this patient, deletion of 6q15–25 was found, indicating loss of one PRDM1 allele. It was reported that PRDM1 is inactivated by chromosomal alterations in 24% of activated B cell–like DLBCL, suggesting that PRDM1 acts as a tumor suppressor gene, and its inactivation may contribute to lymphomagenesis by blocking post–GC differentiation. The combination of BACH2 and MYC in double IGH translocations is unique and consistent with previous reports demonstrating that each partner gene found in double or multiple IGH translocations is exclusively specific to certain types of B-cell lymphoma. These results suggest that the promoter of IGHCδ and/or Eμ enhancer of IGH activate the expression of BACH2, and that BACH2 may act as oncogene in some cases with B-cell lymphoma. Although the IGH-BACH2 translocation is rare in NHL, our data suggest that the BACH2 plays a critical role in B-cell lymphomagenesis through not only IGH translocation but also activation by some other mechanisms. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document