BCL6, p53 and Molecular Targeted Therapy in B-Cell Lymphomas.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1484-1484 ◽  
Author(s):  
Leandro C.A. Cerchietti ◽  
Jose M. Polo ◽  
Gustavo F. Da Silva ◽  
Steve M. Dowdy ◽  
Catoretti M. Giorgio ◽  
...  

Abstract The BCL6 transcriptional repressor is an oncogene often constitutively expressed in diffuse large B-cell lymphomas (DLBCL). The oncogenic mechanism of action of BCL6 presumably involves repression of its direct target genes. We recently developed a targeted therapy agent (called BPI - BCL6 peptide inhibitor) that specifically blocks transcriptional repression by BCL6, and which causes apoptosis in lymphoma cells in vitro and in vivo. We present here potent and stable derivatives of BPI able to specifically eradicate lymphoma cells after a single dose in vitro. Expression array studies of BCL6 target genes reactivated by BPI revealed that one such gene is the p53 tumor suppressor. p53 was also recently shown to be BCL6 target gene by Phan et. al., Nature 2004. We find that BCL6 represses p53 in DLBCL cells through recruitment of the SMRT and N-CoR corepressors, which explains how BPI, which blocks recruitment of these corepressors, reactivates p53. We next wished to determine the contribution of BCL6-mediated repression of p53 to lymphomagenesis, and how p53 modulation might affect BCL6 targeted therapy strategies for DLBCL. We found that BPI could induce p53 target gene expression in DLBCL cells with wild-type p53 and that small molecules or peptides that block p53 rescue apoptosis induced by BPI. In contrast, although BPI also induces p53 in DLCBL cells with mutant p53, there was no activation of p53 target genes and no rescue by p53 blocking molecules. However BPI causes apoptosis of DLBCL cells regardless of p53 status indicating the BCL6 mediates its oncogenic actions through both p53 dependent and independent pathways. p53 is usually wild-type in DLBCL and our analysis of >100 patients show that p53 protein is, surprisingly, still expressed in these tumors. These data suggest that p53 is not fully active in DLBCL cells, consistent with the fact that we found that BCL6 also directly represses upstream activators of p53 such as Chk1 and ATR. BCL6 blockade thus can fully restore activity of p53, both by increasing its expression levels and by enhancing its activation by upstream mediators. Accordingly, sequential administration of p53 activating molecules that enhance p53 activity, potently synergizes with BPI in killing lymphoma cells. BPI also synergizes with chemotherapy drugs that act in part through p53, such as doxorubicin. From these studies we conclude that i) BCL6 mediates lymphomagenesis by direct repression of p53 and upstream target gene pathways; ii) BCL6 positive lymphomas are dependent on BCL6 for their survival regardless of whether p53 is wild type or mutated; iii) Sequential targeting of BCL6 and p53 with BPI and a p53 activating molecule or doxorubicin is likely to be a highly effective therapeutic regimen for patients with DLBCL, especially for the majority who have wild-type p53; iv) The new BPI derivatives are sufficiently potent and stable to be tested in the clinical setting.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2617-2617
Author(s):  
Heiko Trautmann ◽  
Daniel T. Starczynowski ◽  
Christiane Pott ◽  
Lana Harder ◽  
Norbert Arnold ◽  
...  

Abstract REL/NF-κB transcription factors are implicated in the control of apoptosis and cell growth particular in hematopoetic lineages. The REL locus at chromosomal region 2p13–16 is frequently amplified in B-cell lymphomas including diffuse-large B-cell lymphoma (DLBCL) and may play a role in lymphomagenesis. Overexpression of wild-type REL can transform chicken lymphoid cells in culture, and several experimentally-generated mutations within the REL C-terminal transactivation domain (TAD) have been previously shown to enhance REL’s transforming ability. We analysed 83 B-cell lymphomas included in the ‘Deutsche Krebshilfe’ funded network „Molecular Mechanisms in Malignant Lymphoma“ for the presence of activating mutations in the coding region of REL. We performed a systematic dHPLC screening for mutation discovery and identified an identical point mutation in two human B-cell lymphomas (a t(14;18)-positive follicular lymphoma and a mediastinal B-cell lymphoma) that changes Ser525 to Pro within the REL TAD. In the mediastinal B-cell lymphoma, the mutation in REL was proven to be of germline origin. FISH showed an amplification of the REL locus in the tumor cells of this case. Quantitative allelic discrimination of S525P indicates that the mutant REL gene was over-represented in both cases. By in vitro experiments we could show that the S525P mutation enhances the in vitro transforming ability of REL in chicken spleen cells. In addition, REL-S525P differs from wild-type REL in its ability to activate certain κB site-containing reporter plasmids in transient transfection assays. In particular, REL-S525P has a reduced ability to activate the human manganese superoxide dismutase (MnSOD) promoter in A293 cells; however, the MnSOD protein is over-expressed in REL-S525P-transformed chicken spleen cells as compared to wild-type REL-transformed cells. Ser525 of REL falls within a sequence that is similar to other known phosphorylation sites of the IκB kinase, and REL-S525P shows a reduced ability to be phosphorylated by IKKα in vitro. The S525P mutation reduces IKKα- and TNFα-stimulated transactivation by REL, as measured in GAL4 reporter assays. Furthermore, REL-S525P-transformed chicken spleen cells are more resistant to TNFα-induced cell death than cells transformed by wild-type REL. These results represent the first identification of a tumor-derived activating mutation in the REL proto-oncogene, and they suggest that the S525P mutation contributes to the development of human B-cell lymphomas by altering REL’s ability to induce target gene expression by affecting an IKKα-regulated transactivation activity.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2004-2004
Author(s):  
Lan V Pham ◽  
Michael A. Thompson ◽  
Archito Tamayo ◽  
Changping Li ◽  
Garth Powis ◽  
...  

Abstract Abstract 2004 Diffuse large B cell lymphomas (DLBCL) are the most common non-Hodgkin (NHL-B) lymphoma. Current treatment is fairly successfully (∼70-80% remission with R-CHOP frontline chemotherapy), but relapse is common (∼50% after 2–3yrs) with poor salvage therapy options and short survival in relapsed/refractory (r/r) DLBCL. The greatest challenge in improving survival of DLBCL patients is overcoming chemo-resistance, which we currently have very little understanding of the basis. Elucidation of molecular pathways and tumor-encoded genes whose expression contribute to the intrinsic resistance and rapid cell growth of lymphoma cells could yield immediate clinical benefits and reveal new therapeutic targets for effective control and treatment of r/r lymphomas. Thioredoxin (Trx) family members play critical roles in the regulation of cellular redox homeostasis. Cancer cells exist in a stressed environment and rely on the Trxs for protection against stress-disregulated redox signaling. The most extensively studied member of the family is Trx-1 whose levels are increased in many human cancers, most likely in direct response to stress. Trx-1 contributes to many of the hallmarks of cancer including increased proliferation, resistance to cell death and increased angiogenesis. Trx-1 is a validated cancer drug target associated with aggressive tumor growth, resistance to standard therapy and decreased patient survival. In this study we showed that the majority of the DLBCL-derived cell lines express high basal levels of thioredoxin-1 (Trx-1) when compared to normal B cells by both Western blotting and real-time PCR. Tissue microarray (TMA) and immunohistochemistry analysis of 100 cases of DLBCL showed that 77% of the cases were positive for Trx-1 and 40% of the cases over-expressed Trx-1. Based on these observations, we hypothesize that aberrant expression of Trx-1 contributes to the development of drug resistance and cell growth and survival phenotype in DLBCL. Inhibition of Trx-1 by small interfering RNA (siRNA) inhibited DLBCL cell line growth in vitro. Down-regulation of Trx-1 also sensitized lymphoma cells to doxorubicin-induced cell growth inhibition. A doxorubicin-resistant human DLBCL cell line (McA), that was highly sensitive to doxorubicin, has been selected by continuously exposing cells to gradually increasing doses of doxorubicin. The resistant phenotype has been retained for over 6 months despite growth in drug-free medium. Western blot analysis detected increased expression of the Trx-1 in the resistant subline. These results indicate that Trx-1 plays a key role in growth and survival, as well as chemoresistance in the pathophysiology of DLBCL. Based on these data, we plan to use small molecule inhibitors in vitro as a proof of principle for possible future human phase I studies. Disclosures: Ford: Cell Point: Research Funding.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 117-117
Author(s):  
Lan V Pham ◽  
Jerry Bryant ◽  
Archito Tamayo ◽  
Richard Mendez ◽  
Edna Chum ◽  
...  

Abstract Abstract 117 Aggressive non-Hodgkin lymphomas (NHL), such as diffuse large B cell lymphoma (DLBCL) and mantle cell lymphoma (MCL), are very common in the US with increasing incidences. Although these lymphomas are now potentially curable, almost half the treated patients still develop relapsed/refractory (r/r) disease with poor survival outcomes, indicating an urgent need for better therapeutic approaches with improved efficacy, particularly in r/r lymphomas. The emerging area in lymphoma biology involving energy metabolism, has begun to identify likely potential molecular targets for novel therapeutics that can fundamentally change the conventional treatment of cancer. Glucose metabolism, besides its basic metabolic functions in cell physiology, has been shown to provide the major energy source fueling tumor cell growth and survival. We show here that aggressive B-cell lymphomas highly expressed glucose transporters types 1(Glut1) and 3 (Glut3) and hexokinase II (HKII) in both cell lines and primary cases. Since glucose metabolism is highly active and utilized in aggressive B-cell lymphomas, we explored exploiting this bi-functional pathway(s) as a targeted therapy. Previous studies have shown that 99mTc-ethylenedicysteine-N-Acetylglucosamine (ECG), a synthetic glucose analogue, accumulates in cancer cell nuclei and various tumors in animal models. Its' also been shown to differentiate tumor vs inflammation in animal models. Rhenium (Re-therapeutic metal) metal displays similar chemical properties to 99mTc (diagnostic metal), allowing the use of the same backbone (synthetic glucose analogue) for diagnostic and potential therapy. This analogue provides the capability to simultaneously monitor therapeutic activity during induction of tumor treatment therapy while assessing its efficacy. Re-ECG was synthesized by reacting Re-EC with glucosamine tetraacetate. After deprotection of tetraacetate, Re-ECG was produced. Thymidine incorporation assays indicated that Re-ECG inhibits cell proliferation of aggressive dividing lymphoma cells, but was less effective in slow growing lymphoma cells. Further analysis showed that Re-ECG uptake is also more prominent in highly proliferative lymphoma cells, where Re-ECG enters the nucleus and causes DNA damage that leads to lymphoma cell apoptosis. We also evaluated the biodistribution pattern of Re-ECG in a lymphoma SCID mouse model through imaging by single-photon emission computed tomography (SPECT), accelerator mass spectrometry (AMS) and liquid scintillation counting (LSC). The bio-distribution data revealed that radioactivity (14C-187Re-ECG) was retained well in tumor tissue 2 hours post-injection with little to no uptake in the plasma when compared to tumor. The compound was excreted over longer incubation time. Over all, we observed that the lymphoma tumor has an extended drug retention time when compared to other tissues. These results suggest that the metallic pharmaceutical agent 187Re-ECG is an excellent potential candidate for targeted therapy in aggressive r/r lymphomas. This application further defines the term theranostic for personalized medicine approaches utilizing bifunctional imaging/therapeutic agents. Disclosures: Bryant: Cell Point: Employment. Yang:UT MD Anderson Cancer Center: Patents & Royalties. Rollo:Cell Point: Employment. Ford:Cell Point: Research Funding.


1991 ◽  
Vol 88 (20) ◽  
pp. 8982-8986 ◽  
Author(s):  
G. Shaulsky ◽  
N. Goldfinger ◽  
A. Peled ◽  
V. Rotter

2006 ◽  
Vol 26 (12) ◽  
pp. 4448-4461 ◽  
Author(s):  
Monica Agromayor ◽  
Elzbieta Wloga ◽  
Benedetta Naglieri ◽  
John Abrashkin ◽  
Kapil Verma ◽  
...  

ABSTRACT Although many E2F target genes have been identified recently, very little is known about how any single E2F site controls the expression of an E2F target gene in vivo. To test the requirement for a single E2F site in vivo and to learn how E2F-mediated repression is regulated during development and tumorigenesis, we have constructed a novel series of wild-type and mutant Rb promoter-LacZ transgenic reporter lines that allow us to visualize the activity of a crucial E2F target in vivo, the retinoblastoma tumor suppressor gene (Rb). Two mutant Rb promoter-LacZ constructs were used to evaluate the importance of a single E2F site or a nearby activator (Sp1/Ets) site that is found mutated in low-penetrance retinoblastomas. The activity of the wild-type Rb promoter is dynamic, varying spatially and temporally within the developing nervous system. While loss of the activator site silences the Rb promoter, loss of the E2F site stimulates its activity in the neocortex, retina, and trigeminal ganglion. Surprisingly, E2F-mediated repression of Rb does not act globally or in a static manner but, instead, is a highly dynamic process in vivo. Using neocortical extracts, we detected GA-binding protein α (GABPα, an Ets family member) bound to the activator site and both E2F1 and E2F4 bound to the repressor site of the Rb promoter in vitro. Additionally, we detected binding of both E2F1 and E2F4 to the Rb promoter in vivo using chromatin immunoprecipitation analysis on embryonic day 13.5 brain. Unexpectedly, we detect no evidence for Rb promoter autoregulation in neuroendocrine tumors from Rb +/−; RbP-LacZ mice that undergo loss of heterozygosity at the Rb locus, in contrast to the situation in human retinoblastomas where high RB mRNA levels are found. In summary, this study provides the first demonstration that loss of an E2F site is critical for target gene repression in vivo and underscores the complexity of the Rb and E2F family network in vivo.


2015 ◽  
Vol 39 (1) ◽  
pp. 18-29 ◽  
Author(s):  
Hans G. Drexler ◽  
Stefan Ehrentraut ◽  
Stefan Nagel ◽  
Sonja Eberth ◽  
Roderick A.F. MacLeod

1999 ◽  
Vol 19 (1) ◽  
pp. 495-504 ◽  
Author(s):  
John Sok ◽  
Xiao-Zhong Wang ◽  
Nikoleta Batchvarova ◽  
Masahiko Kuroda ◽  
Heather Harding ◽  
...  

ABSTRACT CHOP (also called GADD153) is a stress-inducible nuclear protein that dimerizes with members of the C/EBP family of transcription factors and was initially identified as an inhibitor of C/EBP binding to classic C/EBP target genes. Subsequent experiments suggested a role for CHOP-C/EBP heterodimers in positively regulating gene expression; however, direct evidence that this is the case has so far not been uncovered. Here we describe the identification of a positively regulated direct CHOP-C/EBP target gene, that encoding murine carbonic anhydrase VI (CA-VI). The stress-inducible form of the gene is expressed from an internal promoter and encodes a novel intracellular form of what is normally a secreted protein. Stress-induced expression of CA-VI is both CHOP and C/EBPβ dependent in that it does not occur in cells deficient in either gene. A CHOP-responsive element was mapped to the inducibleCA-VI promoter, and in vitro footprinting revealed binding of CHOP-C/EBP heterodimers to that site. Rescue of CA-VIexpression in c/ebpβ−/− cells by exogenous C/EBPβ and a shorter, normally inhibitory isoform of the protein known as LIP suggests that the role of the C/EBP partner is limited to targeting the CHOP-containing heterodimer to the response element and points to a preeminent role for CHOP in CA-VI induction during stress.


1995 ◽  
Vol 15 (8) ◽  
pp. 4249-4259 ◽  
Author(s):  
A M Yahanda ◽  
J M Bruner ◽  
L A Donehower ◽  
R S Morrison

Loss or mutation of p53 is thought to be an early event in the malignant transformation of many human astrocytic tumors. To better understand the role of p53 in their growth and transformation, we developed a model employing cultured neonatal astrocytes derived from mice deficient in one (p53 +/-) or both (p53 -/-) p53 alleles, comparing them with wild-type (p53 +/+) cells. Studies of in vitro and in vivo growth and transformation were performed, and flow cytometry and karyotyping were used to correlate changes in growth with genomic instability. Early-passage (EP) p53 -/- astrocytes achieved higher saturation densities and had more rapid growth than EP p53 +/- and +/+ cells. The EP p53 -/- cells were not transformed, as they were unable to grow in serum-free medium or in nude mice. With continued passaging, p53 -/- cells exhibited a multistep progression to a transformed phenotype. Late-passage p53 -/- cells achieved saturation densities 50 times higher than those of p53 +/+ cells and formed large, well-vascularized tumors in nude mice. p53 +/- astrocytes exhibited early loss of the remaining wild-type p53 allele and then evolved in a manner phenotypically similar to p53 -/- astrocytes. In marked contrast, astrocytes retaining both wild-type p53 alleles never exhibited a transformed phenotype and usually senesced after 7 to 10 passages. Dramatic alterations in ploidy and karyotype occurred and were restricted to cells deficient in wild-type p53 following repeated passaging. The results of these studies suggest that loss of wild-type p53 function promotes genomic instability, accelerated growth, and malignant transformation in astrocytes.


Blood ◽  
2006 ◽  
Vol 109 (8) ◽  
pp. 3417-3423 ◽  
Author(s):  
Marina Bousquet ◽  
Cyril Broccardo ◽  
Cathy Quelen ◽  
Fabienne Meggetto ◽  
Emilienne Kuhlein ◽  
...  

Abstract We report a novel t(7;9)(q11;p13) translocation in 2 patients with B-cell acute lymphoblastic leukemia (B-ALL). By fluorescent in situ hybridization and 3′ rapid amplification of cDNA ends, we showed that the paired box domain of PAX5 was fused with the elastin (ELN) gene. After cloning the full-length cDNA of the chimeric gene, confocal microscopy of transfected NIH3T3 cells and Burkitt lymphoma cells (DG75) demonstrated that PAX5-ELN was localized in the nucleus. Chromatin immunoprecipitation clearly indicated that PAX5-ELN retained the capability to bind CD19 and BLK promoter sequences. To analyze the functions of the chimeric protein, HeLa cells were cotransfected with a luc-CD19 construct, pcDNA3-PAX5, and with increasing amounts of pcDNA3-PAX5-ELN. Thus, in vitro, PAX5-ELN was able to block CD19 transcription. Furthermore, real-time quantitative polymerase chain reaction (RQ-PCR) experiments showed that PAX5-ELN was able to affect the transcription of endogenous PAX5 target genes. Since PAX5 is essential for B-cell differentiation, this translocation may account for the blockage of leukemic cells at the pre–B-cell stage. The mechanism involved in this process appears to be, at least in part, through a dominant-negative effect of PAX5-ELN on the wild-type PAX5 in a setting ofPAX5 haploinsufficiency.


Sign in / Sign up

Export Citation Format

Share Document