BCL6 was initially discovered as an oncogene in B-cell lymphomas, where it drives the malignant phenotype by repressing proliferation and DNA damage checkpoints and blocking B-cell terminal differentiation. BCL6 mediates its effects by binding to hundreds of target genes and then repressing these genes by recruiting several different chromatin-modifying corepressor complexes. Structural characterization of BCL6–corepressor complexes suggested that BCL6 might be a druggable target. Accordingly, a number of compounds have been designed to bind to BCL6 and block corepressor recruitment. These compounds, based on peptide or small-molecule scaffolds, can potently block BCL6 repression of target genes and kill lymphoma cells. In the case of diffuse large B-cell lymphomas (DLBCL), BCL6 inhibitors are equally effective in suppressing both the germinal center B-cell (GCB)- and the more aggressive activated B-cell (ABC)-DLBCL subtypes, both of which require BCL6 to maintain their survival. In addition, BCL6 is implicated in an expanding scope of hematologic and solid tumors. These include, but are not limited to, B-acute lymphoblastic leukemia, chronic myeloid leukemia, breast cancer, and non–small cell lung cancer. BCL6 inhibitors have been shown to exert potent effects against these tumor types. Moreover, mechanism-based combinations of BCL6 inhibitors with other agents have yielded synergistic and often quite dramatic activity. Hence, there is a compelling case to accelerate the development of BCL6-targeted therapies for translation to the clinical setting. Clin Cancer Res; 23(4); 885–93. ©2016 AACR.

BCL6 (B-cell lymphoma 6) is emerging as a key oncoprotein and therapeutic target. BCL6 was first identified as a locus affected by chromosomal translocations in diffuse large B-cell lymphomas (DLBCL; ref. 1). However, it is now known to be broadly expressed in many lymphomas regardless of genetic lesions. Its role in lymphomagenesis stems from its function in the humoral immune system, where upregulation of BCL6 is required for the formation of germinal centers (GC) during the humoral immune response (2–4). GCs are transient structures that form in response to antigen stimulation. Within GCs, B cells tolerate massive proliferation and the mutagenic effect of the DNA-editing enzyme AICDA to undergo immunoglobulin affinity maturation (5). All of this is orchestrated by and dependent on BCL6, a powerful transcriptional repressor that silences hundreds of genes. Some of these target genes control DNA damage sensing (i.e., ATR, CHEK1, TP53, ARF, etc.) and proliferation checkpoints (CDKN1A, CDKN1B, CDKN2A, CDKN2B, PTEN, etc.; ref. 6). BCL6 also represses genes required for exit from the GC reaction and plasma cell differentiation (e.g., IRF4, PRDM1; ref. 6). This ensures that GC B cells have sufficient time to acquire somatic hypermutation of their immunoglobulin genes. It is thus easy to visualize how deregulated suppression of these target genes could result in malignant transformation of B cells. Indeed constitutive expression of BCL6 in GC B cells drives the development of DLBCL in mice (7–9).

BCL6 also represses numerous oncogenes in GC B cells, including MYC, BCL2, BMI1, CCND1, and various others (10, 11). Through this function, BCL6 may mitigate its own pro-oncogenic checkpoint repression effect and, thus, reduce the potential for malignant transformation of GC B cells. This effect is abrogated in the presence of BCL2 or MYC translocations, which drive expression of these oncogenes through aberrant regulatory elements. The presence of both MYC and/or BCL2 together with BCL6 (regardless of translocations) is clearly deleterious. It provides B cells with simultaneous suppression of checkpoints through BCL6, along with the progrowth and survival effects of MYC and BCL6. Not surprisingly, the combination of MYC and/or BCL2 with BCL6 in DLBCL has been linked to unfavorable clinical outcomes (12).

In the normal immune response, BCL6 function is terminated by the disruption of BCL6 transcriptional complexes through CD40-induced ERK signaling and downregulation of BCL6 mRNA by IRF4 and PRDM1 (13–15). Termination of BCL6 function is required for B cells to exit the GC reaction. Yet in DLBCLs, a variety of mechanisms contribute to aberrant persistence of BCL6 expression. These include fusion of the BCL6 coding region to heterologous promoters via chromosomal translocations and somatic mutation of binding sites for repressors of BCL6 expression, such as IRF4, and BCL6 itself (15, 16). Somatic mutations of the BCL6 ubiquitin ligase FBXO11 can enhance the half-life of BCL6 protein in DLBCL (17). Induction of Hsp90 activation, which occurs almost universally in DLBCL, forms a positive feedback loop whereby (i) HSP90 maintains BCL6 mRNA and protein stability; (ii) HSP90 enhances BCL6 repressor function by directly forming a complex on chromatin; and (iii) BCL6 repression of EP300 prevents acetylation and inactivation of HSP90, thus further enhancing BCL6 protein expression (18, 19). BCL6 expression can also be aberrantly maintained by hypermethylation of regulatory CpGs contained in the BCL6 first intron (20). The powerful tumorigenic activity of BCL6 and the myriad ways that lymphoma cells maintain its activity have fueled interest in the development of BCL6 inhibitors.

To understand BCL6 as a therapeutic target, it is first necessary to consider how it mediates its biological actions. BCL6 has a trimodular structure consisting of an N-terminal BTB/POZ domain that mediates transcriptional repression, an unstructured middle region containing a second repression domain (RD2), and a series of six C2H2 zinc fingers at the C-terminus that bind to DNA and other proteins (6). The biochemical and biological functions, together with the partner proteins of each of the BCL6 domains, are summarized in Fig. 1. One approach to targeting BCL6 is to completely abrogate its functions, for example, using molecules that could block its zinc fingers from binding to DNA or by destroying or downregulating the entire protein using approaches such as RNAi, antisense molecules, or small molecules that target proteins for proteolytic destruction (e.g., degronomids). Yet in the case of BCL6, such an effect is not desirable. This is because BCL6 knockout mice, in addition to failure to form GCs, also manifest a severe and lethal systemic inflammatory disease driven by T cells and macrophages (21). BCL6-deficient mice die within a few weeks of birth and exhibit massive tissue infiltration of inflammatory cells in lung, heart, and other tissues (2–4). Loss of BCL6 in macrophages causes accelerated atherosclerosis in mice (22).

Figure 1.

The biological functions of BCL6 are mediated through specific protein domains. The figure shows a cartoon representation of the BCL6 domain structure, indicating for each one the biochemical function, partner proteins, and biological functions. HDAC, histone deacetylase.

Figure 1.

The biological functions of BCL6 are mediated through specific protein domains. The figure shows a cartoon representation of the BCL6 domain structure, indicating for each one the biochemical function, partner proteins, and biological functions. HDAC, histone deacetylase.

Close modal

Fortunately, it is possible to avoid these adverse effects by targeting specific sites on BCL6 that drive its cancer functions but leave its anti-inflammatory functions intact. Structure–function studies show that the BCL6 BTB domain mediates repression by recruiting the corepressor proteins SMRT, NCOR, and BCOR to an extended groove motif that forms along the BTB dimer interface (Figs. 1 and 2A and B; refs. 23, 24). BCL6 can bind two corepressors at a time because there are grooves on either side of the dimer. The corepressors bind through an 18 amino acid BCL6-binding domain (BBD) that is highly conserved between SMRT and NCOR, but which is completely different in the case of BCOR (23, 24). Importantly, the BCL6 lateral groove residues that make contact with the BBD are unique to BCL6 and not other BTB proteins (23), which could facilitate the development of specific inhibitors that do not affect other BTB domains. Point mutation of the lateral groove abolishes the repressor function of the BCL6 BTB domain (23).

Figure 2.

The BTB domain of BCL6. The images represent the BCL6 BTB domain alone or in a complex with the SMRT-BBD or compound 79-6, as determined by X-ray crystallography. The BTB domain forms an obligate homodimer, with the two monomers shown in red and blue in each panel. A, Ribbon view, with arrows pointing to the charged pocket motif and the hydrophobic surface, indicating examples of specific residues that participate in these features. B, Space fill representation of the BCL6 BTB domain in a complex with the SMRT BBD, which is the polypeptide chain shown in purple. The lateral groove is indicated in the boxed area.

Figure 2.

The BTB domain of BCL6. The images represent the BCL6 BTB domain alone or in a complex with the SMRT-BBD or compound 79-6, as determined by X-ray crystallography. The BTB domain forms an obligate homodimer, with the two monomers shown in red and blue in each panel. A, Ribbon view, with arrows pointing to the charged pocket motif and the hydrophobic surface, indicating examples of specific residues that participate in these features. B, Space fill representation of the BCL6 BTB domain in a complex with the SMRT BBD, which is the polypeptide chain shown in purple. The lateral groove is indicated in the boxed area.

Close modal

The repressive mechanism of action of the BCL6 BTB domain is quite intricate. NCOR and SMRT have related structures and form similar chromatin-modifying complexes that deacetylate histones (25, 26), whereas BCOR is completely different and forms a variant Polycomb PRC1 complex with multiple distinct effects on chromatin (27). On one hand, BCL6 preferentially represses gene promoters by forming a complex with BCOR (28). However, the BCOR complex can only stably assemble at promoters that are marked with histone 3 lysine 27 trimethylation (H3K27me3) by the Polycomb PRC2 protein EZH2 (29). H3K27me3 forms a binding site for CBX8, one of the components of the BCOR complex. Hence, BCL6-mediated promoter repression in GC and lymphoma cells requires “combinatorial tethering” of the BCOR complex through the simultaneous actions of BCL6 and EZH2 (29). On the other hand, BCL6 also represses gene enhancers, but this effect is linked to preferential interaction with SMRT and NCOR complexes, which include HDAC3 (28). BCL6 enhancer repression is driven through this indirect recruitment of HDAC3 to remove the activating H3K27acetyl mark. Notably, the genes that BCL6 represses by forming promoter repression complexes are mostly different from those that it represses by forming enhancer complexes (28).

Remarkably, mice expressing a mutated form of BCL6, where the BTB domain cannot bind to SMRT, NCOR, and BCOR, live normal, healthy lives without any evidence of inflammatory syndrome. These mice are nonetheless unable to form GCs, specifically due to failure of GC B cells to proliferate and survive (30). This is because BCL6 largely represses DNA damage and proliferation checkpoint genes through the BTB domain, which may explain (as discussed below) why this domain is also important in other tumor types. Indeed, mutation of the BTB domain abrogated the ability of the lymphoma oncogene EZH2Y641 to drive preneoplastic lymphoproliferation in vivo (29). Hence, the BCL6 BTB domain is required for GC formation and lymphomagenesis, but not anti-inflammatory effects. Instead, the anti-inflammatory effect is due to direct competition of the BCL6 zinc fingers with STAT proteins for binding to inflammatory chemokines in macrophages (30). Finally, the minimal BCL6 RD2 domain consists of approximately 40 amino acids and is required for interactions with MTA3 and HDAC2 (31, 32). Mice engineered to express RD2-mutant BCL6 also live normal, healthy lives and have no inflammation (32). They do lack GCs, although in this case, the mechanism involves partial loss of function of T-follicular helper (TFH) cells and disruption of B-cell homing to the GC (32). This extraordinary cell-context biochemical specialization of BCL6 makes it possible to target specific functions relevant to tumor cells and, at the same time, avoid disrupting other critical functions.

The most straightforward approach to targeting BCL6 is disrupting the interaction between the BCL6 BTB domain lateral groove and its corepressors because (i) this protein surface mediates BCL6 oncogenic effects; (ii) the protein interaction is structurally characterized; (iii) it involves a unique interface not conserved in other proteins; and (iv) targeting the BTB lateral groove will not induce deleterious inflammatory effects caused by complete loss of BCL6. Targeting the BTB dimer interface would likely be unsuitable, as BTB domains aggregate and are degraded if they cannot dimerize, which could unleash the inflammatory effects of BCL6 deficiency. The BTB domain has other features, such as a charged pocked motif and a hydrophobic surface for oligomerization, which have not been functionally characterized (Fig. 2A; ref. 23).

Protein–protein interactions have long been thought to be “undruggable” (33) given that many protein interactions involve large surfaces that may be difficult to disrupt with small molecules. However, given sufficient knowledge of the protein interface, it is proving possible to design drugs that exploit vulnerable “sweet spots” (34). The first BCL6 inhibitor contained the 17 amino acids from SMRT-BBD, along with a protein transduction domain for cell penetration (35). This BCL6 peptide inhibitor (BPI) disrupted BCL6 repression complexes, induced expression of BCL6 target genes, killed DLBCL cells in vitro, and phenocopied the BCL6 BTB domain mutant phenotype in vivo (i.e., loss of GC formation without inflammation; Fig. 3). Although BPI was active at low micromolar concentrations, it was also readily degraded by proteases (35). The bulk of intermolecular contacts between the BBD and BCL6 are confined to a 9 amino acid sequence, which was used as the warhead for a second-generation peptide using D-amino acids resistant to proteases (36). The peptide sequence was inverted to maintain proper stereochemistry and included a fusogenic motif for superior uptake within cells. The resulting retro-inverso (RI)-BPI peptide retained specificity for BCL6, but not other BTB proteins; exhibited superior stability; and did not induce immunogenicity (36). RI-BPI killed BCL6-dependent DLBCLs at an average concentration of 16 μmol/L, similar to BPI (Fig. 3; ref. 36). Pharmacokinetic studies in mice revealed peak RI-BPI intratumor concentrations after 30 minutes and persistence in the nuclei of lymphoma cells for >24 hours (36). RI-BPI was nontoxic to mice, even after 1 year of continuous administration (36). These peptides thus appear suitable for use in humans. An alternative peptidomimetic strategy employed aptamer screening to identify Apt48, which binds to the BCL6 BTB domain in a different manner than BPI (Fig. 3; ref. 37). Apt48 promoted reexpression of BCL6 target genes and cell-cycle arrest in lymphoma cells, suggesting interference with BCL6 function.

Figure 3.

Current BCL6 inhibitors. The figure depicts the structure and known activities of published BCL6 inhibitors. ChIP, chromatin immunoprecipitation; EMSA, electrophoretic mobility shift assay; GI50, growth inhibition 50%; ITC, isothermal titration calorimetry; MST, microscale thermophoresis; NA, not applicable; NMR, nuclear magnetic resonance; qPCR quantitative PCR; SPR, surface plasmon resonance.

Figure 3.

Current BCL6 inhibitors. The figure depicts the structure and known activities of published BCL6 inhibitors. ChIP, chromatin immunoprecipitation; EMSA, electrophoretic mobility shift assay; GI50, growth inhibition 50%; ITC, isothermal titration calorimetry; MST, microscale thermophoresis; NA, not applicable; NMR, nuclear magnetic resonance; qPCR quantitative PCR; SPR, surface plasmon resonance.

Close modal

The BCL6 BTB lateral groove–BBD complex was used for in silico screening of small-molecule libraries. These studies yielded a family of compounds with reproducible and specific BCL6 inhibition. A representative compound called “79-6” was shown to bind an aromatic pocket situated in the BTB domain lateral groove (38). 79-6 induced disruption of BCL6 transcriptional complexes, reactivation of BCL6 target genes, and selective killing of BCL6-dependent DLBCL cells (Fig. 3; ref. 38). Even though this small molecule was active in animal models, its binding affinity of approximately 138 μmol/L was lower than the endogenous BBD (∼20 μmol/L; ref. 38). A novel in silico–based fragment-based screening method called “SILCS” (39, 40) was subsequently used to design the most potent BCL6 inhibitor to date, FX1 (41). FX1 is the first reported inhibitor with higher affinity for the BCL6 BTB domain than the endogenous corepressor proteins (Kd ∼ 4 μmol/L), demonstrating the feasibility of blocking large protein interactions with small molecules (Fig. 3; ref. 41). SILCS modeling combined with NMR analysis revealed a more favorable orientation of the compound within the lateral groove aromatic pocket. FX1 outperformed all previous BCL6 inhibitors in cell-based pharmacodynamic assays while maintaining specificity for BCL6. The molecule exhibited favorable pharmacokinetics in vivo and lack of toxicity similar to previous generations of BCL6 inhibitors (41). Other small molecules with potential BCL6-binding activity include the natural product resveratrol (42), which can induce antiproliferative and anti-inflammatory activities in different types of cancer cells through various mechanisms (Fig. 3; ref. 43). Ansamycin antibiotics may also bind to BCL6. The structure of one of the members of this family, rifabutin, with the BCL6 BTB domain showed binding to the lateral groove similar to 79-6, although with a lower binding affinity of approximately 1 mmol/L (Fig. 3; ref. 44).

High BCL6 expression is usually associated with germinal center B (GCB)-type DLBCLs (45), which are the most obvious target disease for BCL6 inhibitors. Accordingly, daily administration of RI-BPI at a dose of 7.5 mg/kg/day reduced the growth of established BCL6-dependent GCB-DLBCL xenografts by 60% to 80% but did not affect BCL6-independent DLBCL xenografts (36). Activity was concentration dependent, with doses of 50 mg/kg/day leading to complete regression of GCB-DLBCLs (6). In spite of its weak binding, the small molecule 79-6 concentrates to high levels within lymphoma cells, which enhances its activity (38). Administration of 50 mg/kg/day 79-6 to SCID mice induced a 65% to 70% reduction in the size of established BCL6-dependent DLBCL xenografts but had no effect on BCL6-independent DLBCLs (38). In contrast, the potent small molecule FX1 induced complete regression of GCB-DLBCL xenografts at 25 mg/kg but did not suppress BCL6-independent lymphomas (41). In all of these studies, the various inhibitors induced BCL6 target gene reexpression, proliferation arrest, and apoptosis in DLBCL tumor xenografts. More importantly, all three compounds were active in killing primary human DLBCL specimens cultured ex vivo, underlining the potential for BCL6 inhibitors to have activity clinical trials (36, 38, 41).

From the predictive biomarker perspective, it would be useful to classify patients as having BCL6-dependent or BCL6-independent lymphomas. The simple expression of BCL6 may not be sufficient to indicate BCL6 dependence, as a few GCB-DLBCL cell lines express BCL6 but are not affected by genetic or pharmacologic BCL6 inhibition (36, 38, 41, 46). A recent study overexpressing BCL6 in hematopoietic cells in mice showed development of lymphomas that were BCL6 independent. These data suggest that BCL6 might sometimes act in a hit-and-run manner (47). Expression of BCL6 is, therefore, necessary but perhaps not in all cases sufficient to predict response to BCL6 inhibitors. Gene expression profiles might help to identify BCL6-dependent lymphomas. Indeed, one study identified a putative BCL6 functional signature by analyzing transcriptional profiles in primary DLBCL patient specimens. When applied to DLBCL cell lines, this signature accurately predicted which ones would be BCL6 dependent and which ones are BCL6 independent based on their response to BCL6 inhibitor (46). Perhaps functionally relevant biomarkers such as these could aid in patient selection for BCL6 inhibitor trials.

The relevance of BCL6 is not limited to GCB-DLBCLs. In fact, BCL6 is expressed in most activated B-cell (ABC)-type DLBCLs, albeit at lower levels (41, 48). Indeed, BCL6 translocation and amplification is largely restricted to ABC-DLBCLs, providing genetic evidence for the importance of BCL6 in these tumors (47). It is worth noting that BCL6 transcript or protein abundance is not associated with the degree of biological dependency on BCL6. Along these lines, BCL6 shRNA knockdown of BCL6 was just as deleterious to ABC-DLBCL cells as to GCB-DLBCL cells (41). ABC-DLBCL cells respond to similar doses of BCL6 inhibitor, such as FX1, which also suppressed the growth of ABC-DLBCL xenografts in vivo. Most importantly, primary human ABC- and GCB-DLBCL cells respond equally well to FX1 ex vivo (41). Given that ABC-DLBCLs manifest inferior outcome, it is warranted to develop BCL6-targeted therapy for these patients. Follicular lymphomas manifest features of GCB cells and, thus, generally express BCL6. We have found that primary low-grade follicular lymphoma cells are killed by the BCL6 inhibitor RI-BPI (E. Valls and A.M. Melnick; unpublished data). Burkitt lymphomas arise from GC B cells, and BL cell lines are potently killed by BCL6 inhibitors in vitro and in vivo (35). Although BCL6 is not usually expressed in multiple myeloma, withdrawal of microenvironment survival signals could induce BCL6 expression (49), raising the possibility that these B-cell neoplasms could be sensitive to BCL6 inhibitors as part of combination therapy regimens.

BCL6 is implicated in an expanding list of tumors. Table 1 summarizes the spectrum of BCL6-dependent tumors, the way in which BCL6 was inhibited, and the phenotype associated with the inhibition. For example, BCL6 is a direct transcriptional target of MLL fusion proteins in 11q23-rearranged B-acute lymphoblastic leukemias (B-ALL; ref. 50). Binding of MLL fusion protein to the BCL6 promoter was associated with hypomethylation and induction of BCL6 expression. Treatment with RI-BPI reduced colony formation capacity and induced apoptosis in MLL fusion cell lines and primary patient samples (50). Exposure to ABL tyrosine kinase inhibitors (TKI) strongly induces BCL6 expression in BCR-ABL+ B-ALLs. In this context, BCL6 functioned as a survival feedback mechanism that enables leukemia cells to resist TKI treatment (51). Combined treatment of primary human BCR-ABL+ B-ALL with TKI and RI-BPI yielded massively synergistic antileukemia effects in vitro and in vivo (51). Chronic myeloid leukemias (CML) are also driven by the BCR–ABL fusion protein. Similar to B-ALLs, exposure to TKI induced BCL6 in CML cells (52). CD34+ CML leukemic stem cells (LSC) upregulate BCL6 in response to TKI in a FOXO-dependent manner. It has been appreciated that CML LSCs are less responsive to TKI than bulk leukemia cells, and, in many patients, the disease is not eradicated by TKI therapy alone (52). It is notable that CML LSCs require BCL6 for their ability to form colonies, and initiate leukemia in mice. Treatment of patient-derived CML cells with RI-BPI selectively induced cell-cycle arrest of CD34+ LSCs, causing depletion of CD34+ CD38 LSCs, whereas CD34 subpopulations remained intact (52). Hence, BCL6-targeted therapy could potentially eradicate CML LSCs and reduce the need for long-term TKI therapy.

Table 1.

Therapeutic targeting of BCL6 in lymphomas and other tumors

MalignancyDescriptionBCL6 RNABCL6 inhibitionPhenotypeIn vivo inhibitionReference
Lymphoma 
 GCB-DLBCL Germinal center B-cell diffuse large B-cell lymphoma Yes siBCL6/shBCL6/BPI/RI-BPI/79-6/FX1 Decreased proliferation and apoptosis Yes 21, 29, 31, 34 
 ABC-DLBCL Activated B-cell diffuse large B-cell lymphoma Yes siBCL6/shBCL6/BPI/RI-BPI/79-6/FX1 Decreased proliferation and apoptosis Yes 21, 29, 31, 34 
 FL Follicular lymphoma Yes RI-BPI Apoptosis NA 51 
Ph+ B-ALL B-cell acute lymphoblastic leukemia with Philadelphia chromosome Yes shBCL6/RI-BPI Reduced colony formation and delayed progression Yes 44 
MLLr B-ALL MLL-rearranged B-cell acute lymphoblastic leukemia Yes siBCL6/RI-BPI Apoptosis NA 43 
CML Chronic myeloid leukemia Yes RI-BPI/dnBCL6 Selectively eradicates CD34+ CD38 leukemia-forming colonies Yes 45 
Breast cancer Breast cancer Yes siBCL6/79-6 Reduced EMT and invasion Yes 46, 47, 48 
NSCLC Non–small cell lung cancer Yes FX1/shBCL6 Inhibits proliferation Yes 49 
MalignancyDescriptionBCL6 RNABCL6 inhibitionPhenotypeIn vivo inhibitionReference
Lymphoma 
 GCB-DLBCL Germinal center B-cell diffuse large B-cell lymphoma Yes siBCL6/shBCL6/BPI/RI-BPI/79-6/FX1 Decreased proliferation and apoptosis Yes 21, 29, 31, 34 
 ABC-DLBCL Activated B-cell diffuse large B-cell lymphoma Yes siBCL6/shBCL6/BPI/RI-BPI/79-6/FX1 Decreased proliferation and apoptosis Yes 21, 29, 31, 34 
 FL Follicular lymphoma Yes RI-BPI Apoptosis NA 51 
Ph+ B-ALL B-cell acute lymphoblastic leukemia with Philadelphia chromosome Yes shBCL6/RI-BPI Reduced colony formation and delayed progression Yes 44 
MLLr B-ALL MLL-rearranged B-cell acute lymphoblastic leukemia Yes siBCL6/RI-BPI Apoptosis NA 43 
CML Chronic myeloid leukemia Yes RI-BPI/dnBCL6 Selectively eradicates CD34+ CD38 leukemia-forming colonies Yes 45 
Breast cancer Breast cancer Yes siBCL6/79-6 Reduced EMT and invasion Yes 46, 47, 48 
NSCLC Non–small cell lung cancer Yes FX1/shBCL6 Inhibits proliferation Yes 49 

NOTE: The spectrum of tumors that are dependent on BCL6 and are suppressed by BCL6 inhibitors.

Abbreviations: EMT, epithelial–mesenchymal transition; NA, not applicable.

Angioimmunoblastic T-cell lymphomas (AITL) manifest the phenotypic hallmarks of TFH cells. BCL6 is required for the formation of TFH cells (53). AITLs strongly express BCL6, and, hence, it has been proposed that BCL6 might be a therapeutic target in this disease (53). Up to 50% of breast tumors and many breast cancer cell lines contain amplification of the BCL6 locus (54–56). Functional studies suggest that BCL6 contributes to the development of breast cancer (55–58). BCL6 is important for tumor maintenance as treatment of breast cancer cell lines with RI-BPI or 79-6 reduced cell viability (54). BCL6 may have cell context-specific functions in breast cancer cells, as its target genes are only partially overlapping with those in B cells (54). BCL6 was shown to be required for survival and proliferation of non–small cell lung cancer (NSCLC) cells, in part due to repression of genes involved in the DNA damage response. Inhibition of BCL6 by FX1 kills NSCLC cells in vitro and in vivo and exerts synergistic effect with cisplatin in xenograft models (59).

Given the complexity and heterogeneity of tumor cells, it is unlikely that single agents will cure disease. Hence, rational combination of BCL6 inhibitors with other drugs is the best way to achieve maximal antitumor effect. Blocking the BCL6 BTB domain is not expected to induce major biological side effects, so it should be straightforward to combine BCL6 inhibitors with other therapeutic agents (36, 38, 41). For example, the fact that BCL6 and EZH2 cooperate and require each other to repress transcription led to the notion of combining EZH2 and BCL6 inhibitors (29). As might be expected, this combination yielded enhanced reactivation of BCL6/EZH2 target genes, with corresponding cooperative effects against DLBCL cells in vitro and in vivo as well as against primary human DLBCL specimens ex vivo (29). The result was observed in EZH2-mutant and wild-type DLBCL cells and is summarized in Fig. 4A (29).

Figure 4.

Strategies for rational combinatorial therapy against BCL6 in lymphomas. A, Targeting the BCL6–EZH2 combinatorial tethering mechanism. BCL6 and EZH2 cooperate to recruit the BCOR corepressor complex. BCL6 directly binds to BCOR, and EZH2 deposits the H3K27me3 mark that is bound by the CBX8 subunit of the BCOR complex. Targeting BCL6 and EZH2 together results in more thorough disabling of transcriptional repression in lymphomas and greater therapeutic effect. B, Targeting the HSP90–BCL6 positive feedback loop through which BCL6 targets, including TP53, are suppressed at the transcriptional and posttranslational levels. Disruption of this axis can be partially achieved by targeting BCL6, HSP90, or HDACs, but more complete suppression of this axis by hitting at least two of these components is highly synergistic. C, Targeting the BCL6-BCL2 oncogene switching mechanism. Targeting BCL6 results in derepression of BCL2, enabling lymphoma cells to survive by switching to a dependency on BCL2. Targeting both together prevents lymphoma cells from utilizing this escape mechanism and also is useful in cases with BCL2 translocation where BCL2 has become independent of regulation through BCL6. HDAC, histone deacetylase.

Figure 4.

Strategies for rational combinatorial therapy against BCL6 in lymphomas. A, Targeting the BCL6–EZH2 combinatorial tethering mechanism. BCL6 and EZH2 cooperate to recruit the BCOR corepressor complex. BCL6 directly binds to BCOR, and EZH2 deposits the H3K27me3 mark that is bound by the CBX8 subunit of the BCOR complex. Targeting BCL6 and EZH2 together results in more thorough disabling of transcriptional repression in lymphomas and greater therapeutic effect. B, Targeting the HSP90–BCL6 positive feedback loop through which BCL6 targets, including TP53, are suppressed at the transcriptional and posttranslational levels. Disruption of this axis can be partially achieved by targeting BCL6, HSP90, or HDACs, but more complete suppression of this axis by hitting at least two of these components is highly synergistic. C, Targeting the BCL6-BCL2 oncogene switching mechanism. Targeting BCL6 results in derepression of BCL2, enabling lymphoma cells to survive by switching to a dependency on BCL2. Targeting both together prevents lymphoma cells from utilizing this escape mechanism and also is useful in cases with BCL2 translocation where BCL2 has become independent of regulation through BCL6. HDAC, histone deacetylase.

Close modal

The positive feedback loop between BCL6, Hsp90, and EP300 mentioned earlier was the basis to combine BCL6 inhibitor with drugs against either Hsp90 or histone deacetylase (HDAC) inhibitors (18). This is because inhibition of Hsp90 impairs BCL6 repressor functions and reduces BCL6 mRNA and protein levels (Fig. 4B). This approach yields enhanced derepression of BCL6 target genes, such as EP300, resulting in reduced Hsp90 activity through EP300-mediated acetylation, increased TP53 expression and function, and restored DNA damage and proliferation checkpoints (18). Accordingly, the combination of RI-BPI with Hsp90 or HDAC inhibitor was highly synergistic in vitro and yielded more potent antilymphoma effects in vivo (18). In a similar vein, enhanced checkpoint reactivation through combination of BCL6 inhibitor and TP53 activator also yielded enhanced antitumor effects (60). Indeed, as BCL6 reactivates DNA damage checkpoints, it is not surprising that combinations of chemotherapy drugs plus BCL6 inhibitor yield enhanced activity against lymphomas and leukemias (41). The feedback mechanism where upregulation of BCL6 can protect BCR-ABL+ neoplasms from TKIs supports the testing of BCL6 inhibitor combination therapy in B-ALL and CML (51, 52).

Other combination therapies could target feedback mechanisms that enable cells to escape killing by BCL6 inhibitors. For example, BCL6 directly binds and represses BCL2 and BCL2L1 (BCL-XL; Fig. 4C; refs. 10, 11). Inhibition of BCL6 induces the expression of BCL2 and BCL-XL, which can help lymphoma cells survive exposure to BCL6 inhibitors, a phenomenon that has been called “oncogene switching.” The combination of BCL6 inhibitors with BH3 mimetics overcomes this effect and yields synergistic activity (61). BCL6 represses multiple NFκB-related genes, which are induced after inhibiting BCL6. The effects of NFκB are complex, but proof-of-principle experiments combining proteasome inhibitors with BCL6 inhibitors showed highly synergistic actions. BCL6 also represses STAT3 (62, 63), an oncogenic driver in tumors including breast cancer, which could be induced by BCL6 inhibitors. Suppression of STAT activation through Jak2 or STATs inhibition caused additive loss of breast cancer cell viability, perhaps in part by suppressing additional STAT protumorigenic effects (54, 63). As the functions of BCL6 in various tumor types are better understood, it is likely that other such mechanism-based opportunities will be developed.

In summary, BCL6 is a suitable therapeutic target that plays a broad and critical role in many cancers. Targeting the BCL6 BTB domain is an effective means of disrupting its functions without inducing significant off-target effects against normal tissues. Mechanism-based combination therapy of BCL6 inhibitors yields dramatic antitumor effects. Hence, we propose that development of clinical grade BCL6 compounds is a mission worth vigorously pursuing.

A.D. MacKerell Jr is a cofounder and chief scientific officer of SilcsBio LLC and is a consultant/advisory board member for BioVia. A.M. Melnick reports receiving other commercial research support from Eli Lilly, GlaxoSmithKline, Janssen, and Roche and is a consultant/advisory board member for Boehringer Ingelheim, Eli Lilly, Epizyme, and Roche. M.G. Cardenas, F. Xue, A. D. MacKerell Jr, and A. M. Melnick are listed as coinventors on a patent on BCL6 inhibitors owned by Weill Cornell Medicine. No potential conflicts of interest were disclosed by the other authors.

This work was supported by the Leukemia & Lymphoma Society Therapy Acceleration Program.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Ye
BH
,
Lista
F
,
Lo Coco
F
,
Knowles
DM
,
Offit
K
,
Chaganti
RS
, et al
Alterations of a zinc finger-encoding gene, BCL-6, in diffuse large-cell lymphoma
.
Science
1993
;
262
:
747
50
.
2.
Ye
BH
,
Cattoretti
G
,
Shen
Q
,
Zhang
J
,
Hawe
N
,
de Waard
R
, et al
The BCL-6 proto-oncogene controls germinal-centre formation and Th2-type inflammation
.
Nat Genet
1997
;
16
:
161
70
.
3.
Dent
AL
,
Shaffer
AL
,
Yu
X
,
Allman
D
,
Staudt
LM
. 
Control of inflammation, cytokine expression, and germinal center formation by BCL-6
.
Science
1997
;
276
:
589
92
.
4.
Fukuda
T
,
Yoshida
T
,
Okada
S
,
Hatano
M
,
Miki
T
,
Ishibashi
K
, et al
Disruption of the Bcl6 gene results in an impaired germinal center formation
.
J Exp Med
1997
;
186
:
439
48
.
5.
Klein
U
,
Dalla-Favera
R
. 
Germinal centres: role in B-cell physiology and malignancy
.
Nat Rev Immunol
2008
;
8
:
22
33
.
6.
Hatzi
K
,
Melnick
A
. 
Breaking bad in the germinal center: how deregulation of BCL6 contributes to lymphomagenesis
.
Trends Mol Med
2014
;
20
:
343
52
.
7.
Cattoretti
G
,
Pasqualucci
L
,
Ballon
G
,
Tam
W
,
Nandula
SV
,
Shen
Q
, et al
Deregulated BCL6 expression recapitulates the pathogenesis of human diffuse large B cell lymphomas in mice
.
Cancer Cell
2005
;
7
:
445
55
.
8.
Baron
BW
,
Anastasi
J
,
Montag
A
,
Huo
D
,
Baron
RM
,
Karrison
T
, et al
The human BCL6 transgene promotes the development of lymphomas in the mouse
.
Proc Natl Acad Sci U S A
2004
;
101
:
14198
203
.
9.
Kusam
S
,
Vasanwala
FH
,
Dent
AL
. 
Transcriptional repressor BCL-6 immortalizes germinal center-like B cells in the absence of p53 function
.
Oncogene
2004
;
23
:
839
44
.
10.
Ci
W
,
Polo
JM
,
Cerchietti
L
,
Shaknovich
R
,
Wang
L
,
Yang
SN
, et al
The BCL6 transcriptional program features repression of multiple oncogenes in primary B cells and is deregulated in DLBCL
.
Blood
2009
;
113
:
5536
48
.
11.
Saito
M
,
Novak
U
,
Piovan
E
,
Basso
K
,
Sumazin
P
,
Schneider
C
, et al
BCL6 suppression of BCL2 via Miz1 and its disruption in diffuse large B cell lymphoma
.
Proc Natl Acad Sci U S A
2009
;
106
:
11294
9
.
12.
Burotto
M
,
Berkovits
A
,
Dunleavy
K
. 
Double hit lymphoma: from biology to therapeutic implications
.
Expert Rev Hematol
2016
;
9
:
669
78
.
13.
Polo
JM
,
Ci
W
,
Licht
JD
,
Melnick
A
. 
Reversible disruption of BCL6 repression complexes by CD40 signaling in normal and malignant B cells
.
Blood
2008
;
112
:
644
51
.
14.
Ranuncolo
SM
,
Polo
JM
,
Dierov
J
,
Singer
M
,
Kuo
T
,
Greally
J
, et al
Bcl-6 mediates the germinal center B cell phenotype and lymphomagenesis through transcriptional repression of the DNA-damage sensor ATR
.
Nat Immunol
2007
;
8
:
705
14
.
15.
Saito
M
,
Gao
J
,
Basso
K
,
Kitagawa
Y
,
Smith
PM
,
Bhagat
G
, et al
A signaling pathway mediating downregulation of BCL6 in germinal center B cells is blocked by BCL6 gene alterations in B cell lymphoma
.
Cancer Cell
2007
;
12
:
280
92
.
16.
Pasqualucci
L
,
Migliazza
A
,
Basso
K
,
Houldsworth
J
,
Chaganti
RS
,
Dalla-Favera
R
. 
Mutations of the BCL6 proto-oncogene disrupt its negative autoregulation in diffuse large B-cell lymphoma
.
Blood
2003
;
101
:
2914
23
.
17.
Duan
S
,
Cermak
L
,
Pagan
JK
,
Rossi
M
,
Martinengo
C
,
di Celle
PF
, et al
FBXO11 targets BCL6 for degradation and is inactivated in diffuse large B-cell lymphomas
.
Nature
2012
;
481
:
90
3
.
18.
Cerchietti
LC
,
Hatzi
K
,
Caldas-Lopes
E
,
Yang
SN
,
Figueroa
ME
,
Morin
RD
, et al
BCL6 repression of EP300 in human diffuse large B cell lymphoma cells provides a basis for rational combinatorial therapy
.
J Clin Invest
2010
;
120
:
4569
82
.
19.
Cerchietti
LC
,
Lopes
EC
,
Yang
SN
,
Hatzi
K
,
Bunting
KL
,
Tsikitas
LA
, et al
A purine scaffold Hsp90 inhibitor destabilizes BCL-6 and has specific antitumor activity in BCL-6-dependent B cell lymphomas
.
Nat Med
2009
;
15
:
1369
76
.
20.
Lai
AY
,
Fatemi
M
,
Dhasarathy
A
,
Malone
C
,
Sobol
SE
,
Geigerman
C
, et al
DNA methylation prevents CTCF-mediated silencing of the oncogene BCL6 in B cell lymphomas
.
J Exp Med
2010
;
207
:
1939
50
.
21.
Toney
LM
,
Cattoretti
G
,
Graf
JA
,
Merghoub
T
,
Pandolfi
PP
,
Dalla-Favera
R
, et al
BCL-6 regulates chemokine gene transcription in macrophages
.
Nat Immunol
2000
;
1
:
214
20
.
22.
Barish
GD
,
Yu
RT
,
Karunasiri
MS
,
Becerra
D
,
Kim
J
,
Tseng
TW
, et al
The Bcl6-SMRT/NCoR cistrome represses inflammation to attenuate atherosclerosis
.
Cell Metab
2012
;
15
:
554
62
.
23.
Ahmad
KF
,
Melnick
A
,
Lax
S
,
Bouchard
D
,
Liu
J
,
Kiang
CL
, et al
Mechanism of SMRT corepressor recruitment by the BCL6 BTB domain
.
Mol Cell
2003
;
12
:
1551
64
.
24.
Ghetu
AF
,
Corcoran
CM
,
Cerchietti
L
,
Bardwell
VJ
,
Melnick
A
,
Prive
GG
. 
Structure of a BCOR corepressor peptide in complex with the BCL6 BTB domain dimer
.
Mol Cell
2008
;
29
:
384
91
.
25.
Guenther
MG
,
Lane
WS
,
Fischle
W
,
Verdin
E
,
Lazar
MA
,
Shiekhattar
R
. 
A core SMRT corepressor complex containing HDAC3 and TBL1, a WD40-repeat protein linked to deafness
.
Genes Dev
2000
;
14
:
1048
57
.
26.
Li
J
,
Wang
J
,
Wang
J
,
Nawaz
Z
,
Liu
JM
,
Qin
J
, et al
Both corepressor proteins SMRT and N-CoR exist in large protein complexes containing HDAC3
.
EMBO J
2000
;
19
:
4342
50
.
27.
Gearhart
MD
,
Corcoran
CM
,
Wamstad
JA
,
Bardwell
VJ
. 
Polycomb group and SCF ubiquitin ligases are found in a novel BCOR complex that is recruited to BCL6 targets
.
Mol Cell Biol
2006
;
26
:
6880
9
.
28.
Hatzi
K
,
Jiang
Y
,
Huang
C
,
Garrett-Bakelman
F
,
Gearhart
MD
,
Giannopoulou
EG
, et al
A hybrid mechanism of action for BCL6 in B cells defined by formation of functionally distinct complexes at enhancers and promoters
.
Cell Rep
2013
;
4
:
578
88
.
29.
Beguelin
W
,
Teater
M
,
Gearhart
MD
,
Calvo Fernandez
MT
,
Goldstein
RL
,
Cardenas
MG
, et al
EZH2 and BCL6 Cooperate to Assemble CBX8-BCOR complex to repress bivalent promoters, mediate germinal center formation and lymphomagenesis
.
Cancer Cell
2016
;
30
:
197
213
.
30.
Huang
C
,
Hatzi
K
,
Melnick
A
. 
Lineage-specific functions of Bcl-6 in immunity and inflammation are mediated by distinct biochemical mechanisms
.
Nat Immunol
2013
;
14
:
380
8
.
31.
Fujita
N
,
Jaye
DL
,
Geigerman
C
,
Akyildiz
A
,
Mooney
MR
,
Boss
JM
, et al
MTA3 and the Mi-2/NuRD complex regulate cell fate during B lymphocyte differentiation
.
Cell
2004
;
119
:
75
86
.
32.
Huang
C
,
Gonzalez
DG
,
Cote
CM
,
Jiang
Y
,
Hatzi
K
,
Teater
M
, et al
The BCL6 RD2 domain governs commitment of activated B cells to form germinal centers
.
Cell Rep
2014
;
8
:
1497
508
.
33.
Arkin
MR
,
Whitty
A
. 
The road less traveled: modulating signal transduction enzymes by inhibiting their protein-protein interactions
.
Curr Opin Chem Biol
2009
;
13
:
284
90
.
34.
Wells
JA
,
McClendon
CL
. 
Reaching for high-hanging fruit in drug discovery at protein-protein interfaces
.
Nature
2007
;
450
:
1001
9
.
35.
Polo
JM
,
Dell'Oso
T
,
Ranuncolo
SM
,
Cerchietti
L
,
Beck
D
,
Da Silva
GF
, et al
Specific peptide interference reveals BCL6 transcriptional and oncogenic mechanisms in B-cell lymphoma cells
.
Nat Med
2004
;
10
:
1329
35
.
36.
Cerchietti
LC
,
Yang
SN
,
Shaknovich
R
,
Hatzi
K
,
Polo
JM
,
Chadburn
A
, et al
A peptomimetic inhibitor of BCL6 with potent antilymphoma effects in vitro and in vivo
.
Blood
2009
;
113
:
3397
405
.
37.
Chattopadhyay
A
,
Tate
SA
,
Beswick
RW
,
Wagner
SD
,
Ko Ferrigno
P
. 
A peptide aptamer to antagonize BCL-6 function
.
Oncogene
2006
;
25
:
2223
33
.
38.
Cerchietti
LC
,
Ghetu
AF
,
Zhu
X
,
Da Silva
GF
,
Zhong
S
,
Matthews
M
, et al
A small-molecule inhibitor of BCL6 kills DLBCL cells in vitro and in vivo
.
Cancer Cell
2010
;
17
:
400
11
.
39.
Guvench
O
,
MacKerell
AD
 Jr
. 
Computational fragment-based binding site identification by ligand competitive saturation
.
PLoS Comput Biol
2009
;
5
:
e1000435
.
40.
Raman
EP
,
Yu
W
,
Guvench
O
,
Mackerell
AD
. 
Reproducing crystal binding modes of ligand functional groups using Site-Identification by Ligand Competitive Saturation (SILCS) simulations
.
J Chem Inf Model
2011
;
51
:
877
96
.
41.
Cardenas
MG
,
Yu
W
,
Beguelin
W
,
Teater
MR
,
Geng
H
,
Goldstein
RL
, et al
Rationally designed BCL6 inhibitors target activated B cell diffuse large B cell lymphoma
.
J Clin Invest
2016
;
126
:
3351
62
.
42.
Faber
AC
,
Chiles
TC
. 
Resveratrol induces apoptosis in transformed follicular lymphoma OCI-LY8 cells: evidence for a novel mechanism involving inhibition of BCL6 signaling
.
Int J Oncol
2006
;
29
:
1561
6
.
43.
Hsieh
TC
,
Wu
JM
. 
Resveratrol: Biological and pharmaceutical properties as anticancer molecule
.
Biofactors
2010
;
36
:
360
9
.
44.
Evans
SE
,
Goult
BT
,
Fairall
L
,
Jamieson
AG
,
Ko Ferrigno
P
,
Ford
R
, et al
The ansamycin antibiotic, rifamycin SV, inhibits BCL6 transcriptional repression and forms a complex with the BCL6-BTB/POZ domain
.
PLoS One
2014
;
9
:
e90889
.
45.
Alizadeh
AA
,
Eisen
MB
,
Davis
RE
,
Ma
C
,
Lossos
IS
,
Rosenwald
A
, et al
Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling
.
Nature
2000
;
403
:
503
11
.
46.
Polo
JM
,
Juszczynski
P
,
Monti
S
,
Cerchietti
L
,
Ye
K
,
Greally
JM
, et al
Transcriptional signature with differential expression of BCL6 target genes accurately identifies BCL6-dependent diffuse large B cell lymphomas
.
Proc Natl Acad Sci U S A
2007
;
104
:
3207
12
.
47.
Green
MR
,
Vicente-Duenas
C
,
Romero-Camarero
I
,
Long Liu
C
,
Dai
B
,
Gonzalez-Herrero
I
, et al
Transient expression of Bcl6 is sufficient for oncogenic function and induction of mature B-cell lymphoma
.
Nat Commun
2014
;
5
:
3904
.
48.
Iqbal
J
,
Greiner
TC
,
Patel
K
,
Dave
BJ
,
Smith
L
,
Ji
J
, et al
Distinctive patterns of BCL6 molecular alterations and their functional consequences in different subgroups of diffuse large B-cell lymphoma
.
Leukemia
2007
;
21
:
2332
43
.
49.
Hideshima
T
,
Mitsiades
C
,
Ikeda
H
,
Chauhan
D
,
Raje
N
,
Gorgun
G
, et al
A proto-oncogene BCL6 is up-regulated in the bone marrow microenvironment in multiple myeloma cells
.
Blood
2010
;
115
:
3772
5
.
50.
Geng
H
,
Brennan
S
,
Milne
TA
,
Chen
WY
,
Li
Y
,
Hurtz
C
, et al
Integrative epigenomic analysis identifies biomarkers and therapeutic targets in adult B-acute lymphoblastic leukemia
.
Cancer Discov
2012
;
2
:
1004
23
.
51.
Duy
C
,
Hurtz
C
,
Shojaee
S
,
Cerchietti
L
,
Geng
H
,
Swaminathan
S
, et al
BCL6 enables Ph+ acute lymphoblastic leukaemia cells to survive BCR-ABL1 kinase inhibition
.
Nature
2011
;
473
:
384
8
.
52.
Hurtz
C
,
Hatzi
K
,
Cerchietti
L
,
Braig
M
,
Park
E
,
Kim
YM
, et al
BCL6-mediated repression of p53 is critical for leukemia stem cell survival in chronic myeloid leukemia
.
J Exp Med
2011
;
208
:
2163
74
.
53.
Cortes
JR
,
Palomero
T
. 
The curious origins of angioimmunoblastic T-cell lymphoma
.
Curr Opin Hematol
2016
;
23
:
434
43
.
54.
Walker
SR
,
Liu
S
,
Xiang
M
,
Nicolais
M
,
Hatzi
K
,
Giannopoulou
E
, et al
The transcriptional modulator BCL6 as a molecular target for breast cancer therapy
.
Oncogene
2015
;
34
:
1073
82
.
55.
Bos
R
,
van Diest
PJ
,
van der Groep
P
,
Greijer
AE
,
Hermsen
MA
,
Heijnen
I
, et al
Protein expression of B-cell lymphoma gene 6 (BCL-6) in invasive breast cancer is associated with cyclin D1 and hypoxia-inducible factor-1alpha (HIF-1alpha)
.
Oncogene
2003
;
22
:
8948
51
.
56.
Logarajah
S
,
Hunter
P
,
Kraman
M
,
Steele
D
,
Lakhani
S
,
Bobrow
L
, et al
BCL-6 is expressed in breast cancer and prevents mammary epithelial differentiation
.
Oncogene
2003
;
22
:
5572
8
.
57.
Yu
JM
,
Sun
W
,
Hua
F
,
Xie
J
,
Lin
H
,
Zhou
DD
, et al
BCL6 induces EMT by promoting the ZEB1-mediated transcription repression of E-cadherin in breast cancer cells
.
Cancer Lett
2015
;
365
:
190
200
.
58.
Wu
Q
,
Liu
X
,
Yan
H
,
He
YH
,
Ye
S
,
Cheng
XW
, et al
B-cell lymphoma 6 protein stimulates oncogenicity of human breast cancer cells
.
BMC Cancer
2014
;
14
:
418
.
59.
Marullo
R
,
Ahn
H
,
Cardenas
M
,
Melnick
A
,
Xue
F
,
Cerchietti
L
, et al
The transcription factor BCL6 is a rational target in non-small cell lung cancer (NSCLC) [abstract]
.
In
:
Proceedings of the 107th Annual Meeting of the American Association for Cancer Research
; 
2016 Apr
16
20
;
New Orleans, LA. Philadelphia (PA)
:
AACR
; 
2016
.
Abstract nr 1271
.
60.
Cerchietti
LC
,
Polo
JM
,
Da Silva
GF
,
Farinha
P
,
Shaknovich
R
,
Gascoyne
RD
, et al
Sequential transcription factor targeting for diffuse large B-cell lymphomas
.
Cancer Res
2008
;
68
:
3361
9
.
61.
Dupont
T
,
Yang
SN
,
Patel
J
,
Hatzi
K
,
Malik
A
,
Tam
W
, et al
Selective targeting of BCL6 induces oncogene addiction switching to BCL2 in B-cell lymphoma
.
Oncotarget
2016
;
7
:
3520
32
.
62.
Ding
BB
,
Yu
JJ
,
Yu
RY
,
Mendez
LM
,
Shaknovich
R
,
Zhang
Y
, et al
Constitutively activated STAT3 promotes cell proliferation and survival in the activated B-cell subtype of diffuse large B-cell lymphomas
.
Blood
2008
;
111
:
1515
23
.
63.
Walker
SR
,
Nelson
EA
,
Yeh
JE
,
Pinello
L
,
Yuan
GC
,
Frank
DA
. 
STAT5 outcompetes STAT3 to regulate the expression of the oncogenic transcriptional modulator BCL6
.
Mol Cell Biol
2013
;
33
:
2879
90
.