添加链接
link管理
链接快照平台
  • 输入网页链接,自动生成快照
  • 标签化管理网页链接
The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely. As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsement of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer BCL6 was highly expressed in no responsive ABC-DLBCL patients after Ibrutinib treatment. b ROC curve for discriminating no response or response through BCL6 expression. c C481S BTK HBL-1 cells were treated with 0–8 uM Ibrutinib or combination with 17.5 uM FX1. d MTT-based assessment of the IC 50 value of Ibrutinib compared with Ibrutinib combining FX1. Values were calculated mean ± SD ( n = 3, and ** P < 0.01)

Discussion

Even though patients of DLBCL respond sensitively to first-line treatment, approximately 40% of patients still could not benefit from it [ 4 ]. Ibrutinib is an inhibitor of BTK, showed obvious efficacy in rel/ref DLBCL, especially ABC subtype [ 11 ]. A phase I study of Ibrutinib combined rituximab, ifosfamide, carboplatin, and etoposide (R-ICE) for patients with rel/ref DLBCL reported the high rate of overall response of 90%, including 11 patients achieved complete remission (CR) and 7 patients partial remission (PR) [ 19 ]. And another phase III study indicated Ibrutinib and rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) improved event-free survival (EFS), progression-free survival (PFS), overall survival (OS) in ABC DLBCL patients [ 20 ]. Increasing clinical trials investigate Ibrutinib curative effect on DLBCL. We knew that Ibrutinib was approved to be used for several B-cell malignancies in FDA in 2013 [ 10 ]. However, the emergence of Ibrutinib resistance has limited its efficacy [ 21 , 22 ], so it’s important to study the mechanism of Ibrutinib resistance in DLBCL.

In this study, three microarray expression profile datasets were analyzed to further study mechanism of Ibrutinib resistance. First, a total of 237 common DEGs between Ibrutinib sensitive and resistant cell lines were identified through two expression profiles, including 100 upregulated and 137 downregulated genes. Second, GO analysis was performed to study functional roles, including biological process, molecular functions and cellular component. The upregulated DEGs were correlated with transcription and downregulated DEGs were signal transduction and negative regulation of apoptotic process. Third, signal pathway enrichment analysis showed that upregulated DEGs were mainly enriched in cell cycle and downregulated DEGs were mainly enriched in cytokines-based pathways. What is more, PPI network was conducted to illustrate interactions among the DEGs at the protein level, of which, a total of 7 genes- BCL6 , IL10 , IL2RB , IRF4 , CD80 , PMDR1 , GZMB - were selected as hub genes with the threshold of MCC score > 10,000. And through predicting miRNA associated with hub genes, we found that miR-30 family may be related to Ibrutinib resistance in ABC-DLBCL. From the cBioportal database, we found that BCL6 showed the highest level of genetic alterations among above hub genes. Using another expression profile, we found that BCL6 highly expressed in no responsive ABC-DLBCL patients, and the AUC of ROC curve was 0.67.

BCL6 , a transcription repressor, plays an important role of initiation and maintenance of germinal center reactions [ 23 , 24 ], which has been identified as one of predictors of outcome in several cancers, such as DLBCL and B-cell acute lymphoblastic leukemia (B-ALL) [ 25 , 26 ]. It was reported that BCL6 is associated with tyrosine kinase inhibitors (TKI) resistance in Philadelphia chromosome positive (Ph+) ALL and chronic myeloid leukemia (CML) cells [ 27 , 28 ]. And another study showed overexpression of BCL6 inhibited the sensitivity of methotrexate in children with B-ALL by promoting ZEB1 expression [ 29 ]. What is more, Julie et al. found association between BCL6 overexpression and etoposide resistance in DLBCL cell lines [ 30 ]. Cardenas et al. reported that BCL6 expresses in most ABC-DLBCL at a low level [ 31 ]. It was interesting that BCL6 was upregulated in Ibrutinib-resistant ABC-DLBCL cell lines in our study. And BCL6 had the highest MCC score in Ibrutinib-resistant PPI network and linked with another hub genes. So using another gene profile validated that BCL6 highly expressed in no responsive ABC-DLBCL patients after Ibrutinib treatment. And in vitro experiment was carried out to validate if BCL6 inhibitor can enhance the sensitivity of Ibrutinib in C481S BTK HBL-1 cells. FX1, a BCL6 inhibitor, destroyed the formation of BCL6 repression complex and suppressed ABC-DLBCL cell lines with IC 50 of 35 uM [ 31 ]. FX1 used in our study was lower than its IC 50 , which can increase sensitivity of Ibrutinb in C481S BTK HBL-1 cells. Thus, our finding that BCL6 may be involved in drug resistance is consistent with previous studies. What is more, BCL6 maybe the potential target to improve Ibrutinib sensitivity in C481S BTK HBL-1 cells.

BCL6 inhibits expression of various target genes via binding gene promoters. BCL6 not only destroys interactions between T and B cells by CD80 and PD-L1 but also inhibits B cell differentiation across decreasing expression of PRDM1 and IRF4 [ 32 37 ]. Above studies are consistent with the upregulation of BCL6 , and downregulation of other hub genes in our present study. PRDM1 / BLIMP1 encodes a transcriptional repressor, which is necessary for differentiation of B cells into plasma cells [ 38 ]. Studies reported that PRDM1 acts as a tumor suppressor gene in ABC-DLBCL in vivo mouse models [ 39 , 40 ]. As previously described, PRDM1 is frequently inactivated by genetic alterations, including genetic deletions or mutations or transcriptional repression in ABC-DLBCL [ 38 , 41 ]. Parekh et al. found that different genetic alterations within PRDM1 had adverse prognostic factors [ 42 ]. And inactivation of PRDM1 can upregulate expression of C-MYC and downregulate expression of p53 pathway molecule in ABC-DLBCL [ 42 , 43 ]. These results suggest that inactivation of PRDM1 is closely linked to development of ABC-DLBCL.

IRF4 was essential for regulating gene transcription and mitochondrial homeostasis in plasma cells [ 44 ]. IRF4 activates or is repressed by BCL6 , and co-expresses with PRPM1 affecting plasma cells development [ 45 47 ]. However, most studies found that it does not express PRDM1 protein though the presence of IRF4 in ABC-DLBCL, suggesting other regulatory mechanisms between them [ 40 ]. Abnormal expression of IRF4 is linked to several blood malignancies. For example, expression of IRF4 is related to poor survival outcomes in peripheral T-cell lymphoma and chronic lymphocytic leukemia (CLL) [ 48 , 49 ]. What is more, studies showed IRF4 dysregulation is associated with resistance to immunomodulatory compounds in Waldenström’s macroglobulinemia and myeloma [ 50 , 51 ]. A previous study has reported that Ibrutinib downregulates IRF4 and consequently synergizes with lenalidomide in killing ABC DLBCL [ 52 ]. Another study indicated mutation of IRF4 may explain the rel/ref phenotype of ABC-DLBCL [ 5 ]. Therefore, the role of IRF4 in ABC-DLBCL need further explore.

Lin et al. identified that upregulation of miR-30 family can directly downregulate BCL6 in B-lymphocytes and lymphoma cells [ 53 ]. Current studies found miR-30 family played a significant role in various tumors. Zhang et al. proved miR-30d could inhibit autophagy thereby promoting cell apoptosis [ 54 ]. The higher expression of miRNA-30c had better outcome with tamoxifen treatment in breast cancer [ 55 ]. Another investigation found that overexpression of miR-30b and miR-30c have better outcome after TKIs treatment in non-small cell lung cancer [ 56 ]. Interestingly, miR-30 family is considered as oncogenic miRNA, too. For instance, Gaziel-Sovran et al. reported that miR-30b and miR-30d promoted invasion of melanoma cells leading to generated IL10 and reduced immune cells activation and recruitment [ 57 ]. Taken together, miR-30 family has complex functions in various cancers. However, the role of miR-30 family in Ibrutinib resistance of ABC-DLBCL has not been reported. Our study showed that miR-30 family may mediate Ibrutinib resistance in ABC-DLBCL, which is worthy of further exploration.

Conclusion

In summary, the present study has analyzed DEGs based on two microarray expression ( {"type":"entrez-geo","attrs":{"text":"GSE138126","term_id":"138126"}} GSE138126 and {"type":"entrez-geo","attrs":{"text":"GSE93985","term_id":"93985"}} GSE93985 ). BCL6 was identified as the core gene for Ibrutinib resistance in ABC-DLBCL. Using another expression profile ( {"type":"entrez-geo","attrs":{"text":"GSE93984","term_id":"93984"}} GSE93984 ) showed that BCL6 highly expressed in no responsive ABC-DLBCL patients after Ibrutinib treatment. Further study found that BCL6 inhibitor may increase the sensitivity of C481S BTK HBL-1 cells to Ibrutinib therapy. And miRNA target prediction results showed that miR-30 family were involved in Ibrutinib resistance in ABC-DLBCL. And miR-30 family can directly downregulate BCL6 which was reported before [ 53 ]. The BCL6 maybe a potential target overcoming Ibrutinib resistance in ABC-DLBCL.

Acknowledgements

We thank Prof. Zhu for providing the C481S BTK HBL-1 cells.

Author contributions

JL, YH, and YZ contributed equally to this work and are co-first authors. JH and JL conceived and designed the study; YZ and YH found and analyzed the data, YC and JW carried experiments, LW and PJ wrote the paper, JH reviewed the paper.

Funding

This work was supported by National Natural Science Foundation of China (81870135, 81470326, U2005204, 82000142), the Construction Project of the Fujian Medical Center of Hematology (Min201704), the Cooperation Project of University and Industry in Fujian Province (2017Y4005), Startup found of scientific research, Fujian medical university (2019QH1022, 2019QH2012, 2019QH2016).

Availability of data and materials

{"type":"entrez-geo","attrs":{"text":"GSE138126","term_id":"138126"}} GSE138126 , {"type":"entrez-geo","attrs":{"text":"GSE93985","term_id":"93985"}} GSE93985 , and GSE9394 were downloaded from the Gene Expression Omnibus (GEO) database.

Compliance with ethical standards

Conflict of interest

There is no conflict of interest to disclose.

Consent to participate

All authors agree to submit articles for publication.

Consent for publication

All authors agree with publication.

Ethical approval

The studies involving human participants were reviewed and approved by all data are from public database on the internet.

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Jiazheng Li, Yan Huang and Yun Zhang contributed equally as co-first authors.

References

1. Bachy E, Salles G. Treatment approach to newly diagnosed diffuse large B-cell lymphoma. Semin Hematol. 2015; 52 (2):107–118. doi: 10.1053/j.seminhematol.2015.01.001. [ PubMed ] [ CrossRef ] [ Google Scholar ]
2. Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016; 127 (20):2375–2390. doi: 10.1182/blood-2016-01-643569. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
3. Lenz G, Wright G, Dave SS, Xiao W, Powell J, Zhao H. Stromal gene signatures in large-B-Cell Lymphomas. N Engl J Med. 2008; 359 (22):2313–2323. doi: 10.1056/NEJMoa0802885. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
4. Li S, Young KH, Medeiros LJ. Diffuse large B-cell lymphoma. Pathology. 2018; 50 (1):74–87. doi: 10.1016/j.pathol.2017.09.006. [ PubMed ] [ CrossRef ] [ Google Scholar ]
5. Mareschal S, Dubois S, Viailly PJ, Bertrand P, Bohers E, Maingonnat C, et al. Whole exome sequencing of relapsed/refractory patients expands the repertoire of somatic mutations in diffuse large B-cell lymphoma. Genes Chromosomes Cancer. 2016; 55 (3):251–267. doi: 10.1002/gcc.22328. [ PubMed ] [ CrossRef ] [ Google Scholar ]
6. Morin RD, Assouline S, Alcaide M, Mohajeri A, Johnston RL, Chong L, et al. Genetic landscapes of relapsed and refractory diffuse large B-cell lymphomas. Clin Cancer Res. 2016; 22 (9):2290–2300. doi: 10.1158/1078-0432.ccr-15-2123. [ PubMed ] [ CrossRef ] [ Google Scholar ]
7. Jørgensen LK, Poulsen MØ, Laursen MB, Marques SC, Johnsen HE, Bøgsted M, et al. MicroRNAs as novel biomarkers in diffuse large B-cell lymphoma—a systematic review. Dan Med J. 2015; 62 (5):A5048. [ PubMed ] [ Google Scholar ]
8. Garzon R, Calin GA, Croce CM. MicroRNAs in cancer. Annu Rev Med. 2009; 60 :167–179. doi: 10.1146/annurev.med.59.053006.104707. [ PubMed ] [ CrossRef ] [ Google Scholar ]
9. Ting CY, Liew SM, Price A, Gan GG, Bee-Lan Ong D, Tan SY, et al. Clinical significance of aberrant microRNAs expression in predicting disease relapse/refractoriness to treatment in diffuse large B-cell lymphoma: a meta-analysis. Crit Rev Oncol Hematol. 2019; 144 :102818. doi: 10.1016/j.critrevonc.2019.102818. [ PubMed ] [ CrossRef ] [ Google Scholar ]
10. Burger JA, Buggy JJ. Bruton tyrosine kinase inhibitor ibrutinib (PCI-32765) Leuk Lymphoma. 2013; 54 (11):2385–2391. doi: 10.3109/10428194.2013.777837. [ PubMed ] [ CrossRef ] [ Google Scholar ]
11. Wilson WH, Young RM, Schmitz R, Yang Y, Pittaluga S, Wright G, et al. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med. 2015; 21 (8):922–926. doi: 10.1038/nm.3884. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
12. Edgar R, Domrachev M, Lash AE. Gene expression omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002; 30 (1):207–210. doi: 10.1093/nar/30.1.207. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
13. Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, et al. NCBI GEO: archive for functional genomics data sets–update. Nucleic Acids Res. 2013; 41 :D991–D995. doi: 10.1093/nar/gks1193. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
14. da Huang W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009; 4 (1):44–57. doi: 10.1038/nprot.2008.211. [ PubMed ] [ CrossRef ] [ Google Scholar ]
15. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019; 47 :D607–D613. doi: 10.1093/nar/gky1131. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
16. Almeida D, Azevedo V, Silva A, Baumbach J. PetriScape—a plugin for discrete Petri net simulations in Cytoscape. Journal of integrative bioinformatics. 2016; 13 (1):284. doi: 10.2390/biecoll-jib-2016-284. [ PubMed ] [ CrossRef ] [ Google Scholar ]
17. Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013 doi: 10.1126/scisignal.2004088. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
18. Wong N, Wang X. miRDB: an online resource for microRNA target prediction and functional annotations. Nucleic Acids Res. 2015; 43 :D146–D152. doi: 10.1093/nar/gku1104. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
19. Sauter CS, Matasar MJ, Schoder H, Devlin SM, Drullinsky P, Gerecitano J, et al. A phase 1 study of ibrutinib in combination with R-ICE in patients with relapsed or primary refractory DLBCL. Blood. 2018; 131 (16):1805–1808. doi: 10.1182/blood-2017-08-802561. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
20. Younes A, Sehn LH, Johnson P, Zinzani PL, Hong X, Zhu J, et al. Randomized phase III trial of ibrutinib and rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone in non-germinal center B-cell diffuse large B-cell lymphoma. J Clin Oncol. 2019; 37 (15):1285–1295. doi: 10.1200/jco.18.02403. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
21. Dreyling M, Jurczak W, Jerkeman M, Silva RS, Rusconi C, Trneny M, et al. Ibrutinib versus temsirolimus in patients with relapsed or refractory mantle-cell lymphoma: an international, randomised, open-label, phase 3 study. Lancet (London, England) 2016; 387 (10020):770–778. doi: 10.1016/s0140-6736(15)00667-4. [ PubMed ] [ CrossRef ] [ Google Scholar ]
22. Maddocks KJ, Ruppert AS, Lozanski G, Heerema NA, Zhao W, Abruzzo L, et al. Etiology of ibrutinib therapy discontinuation and outcomes in patients with chronic lymphocytic leukemia. JAMA Oncol. 2015; 1 (1):80–87. doi: 10.1001/jamaoncol.2014.218. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
23. Basso K, Dalla-Favera R. BCL6: master regulator of the germinal center reaction and key oncogene in B cell lymphomagenesis. Adv Immunol. 2010; 105 :193–210. doi: 10.1016/s0065-2776(10)05007-8. [ PubMed ] [ CrossRef ] [ Google Scholar ]
24. Hatzi K, Melnick A. Breaking bad in the germinal center: how deregulation of BCL6 contributes to lymphomagenesis. Trends Mol Med. 2014; 20 (6):343–352. doi: 10.1016/j.molmed.2014.03.001. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
25. Lossos IS, Jones CD, Warnke R, Natkunam Y, Kaizer H, Zehnder JL, et al. Expression of a single gene, BCL-6, strongly predicts survival in patients with diffuse large B-cell lymphoma. Blood. 2001; 98 (4):945–951. doi: 10.1182/blood.v98.4.945. [ PubMed ] [ CrossRef ] [ Google Scholar ]
26. Hurtz C, Chan LN, Geng H, Ballabio E, Xiao G, Deb G, et al. Rationale for targeting BCL6 in MLL-rearranged acute lymphoblastic leukemia. Genes Dev. 2019; 33 (17–18):1265–1279. doi: 10.1101/gad.327593.119. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
27. Eskandari S, Yazdanparast R. Bcl6 gene-silencing facilitates PMA-induced megakaryocyte differentiation in K562 cells. J Cell Commun Signal. 2017; 11 (4):357–367. doi: 10.1007/s12079-017-0395-5. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
28. 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 (7347):384–388. doi: 10.1038/nature09883. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
29. Wu HB, Lv WF, Wang YX, Li YY, Guo W. BCL6 promotes the methotrexate-resistance by upregulating ZEB1 expression in children with acute B lymphocytic leukemia. Eur Rev Med Pharmacol Sci. 2018; 22 (16):5240–5247. doi: 10.26355/eurrev_201808_15722. [ PubMed ] [ CrossRef ] [ Google Scholar ]
30. Devin J, Kassambara A, Bruyer A, Moreaux J, Bret C. Phenotypic characterization of diffuse large B-cell lymphoma cells and prognostic impact. J Clin Med. 2019 doi: 10.3390/jcm8071074. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
31. 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 (9):3351–3362. doi: 10.1172/jci85795. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
32. Parekh S, Polo JM, Shaknovich R, Juszczynski P, Lev P, Ranuncolo SM, et al. BCL6 programs lymphoma cells for survival and differentiation through distinct biochemical mechanisms. Blood. 2007; 110 (6):2067–2074. doi: 10.1182/blood-2007-01-069575. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
33. Shaffer AL, Yu X, He YS, Boldrick J, Chan EP, Staudt LM. BCL-6 represses genes that function in lymphocyte differentiation, inflammation, and cell cycle control. Immunity. 2000; 13 :199–212. doi: 10.1016/s1074-7613(00)00020-0. [ PubMed ] [ CrossRef ] [ Google Scholar ]
34. Tunyaplin C, Shaffer AL, Angelin-Duclos CD, Yu X, Staudt LM, Calame KL. Direct repression of prdm1 by Bcl-6 inhibits plasmacytic differentiation. J Immunol (Baltimore, Md: 1950) 2004; 173 (2):1158–1165. doi: 10.4049/jimmunol.173.2.1158. [ PubMed ] [ CrossRef ] [ Google Scholar ]
35. Niu H, Cattoretti G, Dalla-Favera R. BCL6 controls the expression of the B7–1/CD80 costimulatory receptor in germinal center B cells. J Exp Med. 2003; 198 (2):211–221. doi: 10.1084/jem.20021395. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
36. Basso K, Saito M, Sumazin P, Margolin AA, Wang K, Lim WK, et al. Integrated biochemical and computational approach identifies BCL6 direct target genes controlling multiple pathways in normal germinal center B cells. Blood. 2010; 115 (5):975–984. doi: 10.1182/blood-2009-06-227017. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
37. Basso K, Dalla-Favera R. Roles of BCL6 in normal and transformed germinal center B cells. Immunol Rev. 2012; 247 (1):172–183. doi: 10.1111/j.1600-065X.2012.01112.x. [ PubMed ] [ CrossRef ] [ Google Scholar ]
38. Shapiro-Shelef M, Lin KI, McHeyzer-Williams LJ, Liao J, McHeyzer-Williams MG, Calame K. Blimp-1 is required for the formation of immunoglobulin secreting plasma cells and pre-plasma memory B cells. Immunity. 2003; 19 (4):607–620. doi: 10.1016/s1074-7613(03)00267-x. [ PubMed ] [ CrossRef ] [ Google Scholar ]
39. Calado DP, Zhang B, Srinivasan L, Sasaki Y, Seagal J, Unitt C, et al. Constitutive canonical NF-κB activation cooperates with disruption of BLIMP1 in the pathogenesis of activated B cell-like diffuse large cell lymphoma. Cancer Cell. 2010; 18 (6):580–589. doi: 10.1016/j.ccr.2010.11.024. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
40. Mandelbaum J, Bhagat G, Tang H, Mo TW, Brahmachary M, Shen Q, et al. BLIMP1 is a tumor suppressor gene frequently disrupted in activated B cell-like diffuse large B cell lymphoma. Cancer Cell. 2010; 18 (6):568–579. doi: 10.1016/j.ccr.2010.10.030. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
41. Tam W, Gomez M, Chadburn A, Lee JW, Chan WC, Knowles DM. Mutational analysis of PRDM1 indicates a tumor-suppressor role in diffuse large B-cell lymphomas. Blood. 2006; 107 (10):4090–4100. doi: 10.1182/blood-2005-09-3778. [ PubMed ] [ CrossRef ] [ Google Scholar ]
42. Xia Y, Xu-Monette ZY, Tzankov A, Li X, Manyam GC, Murty V, et al. Loss of PRDM1/BLIMP-1 function contributes to poor prognosis of activated B-cell-like diffuse large B-cell lymphoma. Leukemia. 2017; 31 (3):625–636. doi: 10.1038/leu.2016.243. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
43. Zhang XY, Ma ZP, Cui WL, Pang XL, Chen R, Wang L, et al. Impact of PRDM1 gene inactivation on C-MYC regulation in diffuse large B-cell lymphoma. Chin J Pathol. 2018; 47 (1):25–31. doi: 10.3760/cma.j.issn.0529-5807.2018.01.006. [ PubMed ] [ CrossRef ] [ Google Scholar ]
44. Low MSY, Brodie EJ, Fedele PL, Liao Y, Grigoriadis G, Strasser A, et al. IRF4 activity is required in established plasma cells to regulate gene transcription and mitochondrial homeostasis. Cell Rep. 2019; 29 (9):2634–45 e5. doi: 10.1016/j.celrep.2019.10.097. [ PubMed ] [ CrossRef ] [ Google Scholar ]
45. Angelin-Duclos C, Cattoretti G, Lin KI, Calame K. Commitment of B lymphocytes to a plasma cell fate is associated with Blimp-1 expression in vivo. J Immunol (Baltimore, Md: 1950) 2000; 165 (10):5462–5471. doi: 10.4049/jimmunol.165.10.5462. [ PubMed ] [ CrossRef ] [ Google Scholar ]
46. Shapiro-Shelef M, Calame K. Regulation of plasma-cell development. Nat Rev Immunol. 2005; 5 (3):230–242. doi: 10.1038/nri1572. [ PubMed ] [ CrossRef ] [ Google Scholar ]
47. Klein U, Dalla-Favera R. Germinal centres: role in B-cell physiology and malignancy. Nat Rev Immunol. 2008; 8 (1):22–33. doi: 10.1038/nri2217. [ PubMed ] [ CrossRef ] [ Google Scholar ]
48. Heo MH, Park HY, Ko YH, Kim WS, Kim SJ. IRF4/MUM1 expression is associated with poor survival outcomes in patients with peripheral T-cell lymphoma. J Cancer. 2017; 8 (6):1018–1024. doi: 10.7150/jca.17358. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
49. Chang CC, Lorek J, Sabath DE, Li Y, Chitambar CR, Logan B, et al. Expression of MUM1/IRF4 correlates with clinical outcome in patients with B-cell chronic lymphocytic leukemia. Blood. 2002; 100 (13):4671–4675. doi: 10.1182/blood-2002-01-0104. [ PubMed ] [ CrossRef ] [ Google Scholar ]
50. Zhu YX, Shi CX, Bruins LA, Wang X, Riggs DL, Porter B, et al. Identification of lenalidomide resistance pathways in myeloma and targeted resensitization using cereblon replacement, inhibition of STAT3 or targeting of IRF4. Blood Cancer J. 2019; 9 (2):19. doi: 10.1038/s41408-019-0173-0. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
51. Bertrand E, Jouy N, Manier S, Guillemette F, Guidez S, Eileen B, et al. Role of IRF4 in resistance to immunomodulatory (IMid) compounds® in Waldenström’s macroglobulinemia. Onco Targets Ther. 2017; 8 (68):112917–112927. doi: 10.18632/oncotarget.22872. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
52. Yang Y, Shaffer AL, III, Emre NT, et al. Exploiting synthetic lethality for the therapy of ABC diffuse large B cell lymphoma. Cancer Cell. 2012; 21 (6):723–737. doi: 10.1016/j.ccr.2012.05.024. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
53. Lin J, Lwin T, Zhao JJ, et al. Follicular dendritic cell-induced microRNA-mediated upregulation of PRDM1 and downregulation of BCL-6 in non-Hodgkin’s B-cell lymphomas. Leukemia. 2011; 251 (1):145–152. doi: 10.1038/leu.2010.230. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
54. Zhang R, Xu J, Zhao J, Bai J. Mir-30d suppresses cell proliferation of colon cancer cells by inhibiting cell autophagy and promoting cell apoptosis. Tumour Biol. 2017; 39 (6):1010428317703984. doi: 10.1177/1010428317703984. [ PubMed ] [ CrossRef ] [ Google Scholar ]
55. Rodríguez-González FG, Sieuwerts AM, Smid M, Look MP, Meijer-van Gelder ME, de Weerd V, et al. MicroRNA-30c expression level is an independent predictor of clinical benefit of endocrine therapy in advanced estrogen receptor positive breast cancer. Breast Cancer Res Treat. 2011; 127 (1):43–51. doi: 10.1007/s10549-010-0940-x. [ PubMed ] [ CrossRef ] [ Google Scholar ]
56. Gu YF, Zhang H, Su D, Mo ML, Song P, Zhang F, et al. miR-30b and miR-30c expression predicted response to tyrosine kinase inhibitors as first line treatment in non-small cell lung cancer. Chin Med J. 2013; 126 (23):4435–4439. [ PubMed ] [ Google Scholar ]
57. Gaziel-Sovran A, Segura MF, Di Micco R, Collins MK, Hanniford D, Vega-Saenz de Miera E, et al. miR-30b/30d regulation of GalNAc transferases enhances invasion and immunosuppression during metastasis. Cancer Cell. 2011; 20 (1):104–118. doi: 10.1016/j.ccr.2011.05.027. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]

Articles from Medical Oncology (Northwood, London, England) are provided here courtesy of Springer