B Cells and B-Cell Receptor Signaling: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , ,

B-cell receptor (BCR) is a B cell hallmark surface complex regulating multiple cellular processes in normal as well as malignant B cells. Igα (CD79a)/Igβ (CD79b) are essential components of BCR that are indispensable for its functionality, signal initiation, and signal transduction. CD79a/CD79b-mediated BCR signaling is required for the survival of normal as well as malignant B cells via a wide signaling network. Studies identified the great complexity of this signaling network and revealed the emerging role of CD79a/CD79b in signal integration.

  • B cell
  • B-cell receptor (BCR)
  • signaling
  • lymphoid malignancies
  • non-Hodgkin lymphoma (NHL)

1. Introduction

B-cell receptor (BCR) signaling plays a critical role at multiple stages of B cell life cycle [1]. BCR is a hallmark molecule of B cells that determines cellular fate and regulates B cell survival, fitness, activation, development, and transformation into immunoglobulin-secreting cells [2]. Structurally, BCR is composed of a membrane-bound immunoglobulin (Ig) molecule noncovalently linked with Igα (CD79a) and Igβ (CD79b) transmembrane signaling subunits [2][3][4]. BCR has a central role within the adaptive immune system and antigen recognition. Antigen binding triggers antigen induced BCR signaling, activating an array of signaling cascades and cellular processes. These include BCR-mediated antigen capture, recognition, uptake, and processing (also making B cells effective antigen-presenting cells) [5]. Importantly, BCR is also a source of baseline antigen-independent signaling, the so-called “tonic” BCR signaling. Tonic BCR signaling is necessary for correct B-cell development and for survival of normal mature B cells [6]. At the same time, altered BCR signaling has been implicated in the pathogenesis of autoimmune inflammatory disorders and B-cell derived malignancies [7][8][9][10].

2. BCR Signaling in Normal B Cells

BCR signaling is of critical importance for normal B cell performance at all different stages of their development. BCR can induce a wide array of cellular responses related to the complexity of the intracellular BCR signaling network. Individual features of BCR signaling are highly dependent on a specific stage of B cell development and activation status [11]. Importantly, pre-BCR, which is structurally similar to BCR but contains a surrogate light chain (SLC) made up of the invariant proteins λ5 (CD179b) and VpreB (CD179a) instead of the Ig light chain in BCR [12], is transiently expressed in early developmental stages and is necessary for pro-B to pre-B transition and pre-B cell expansion [13][14]. Correct assembly and proper SLC replacement with the conventional light chain in pre-BCR is an imperative for further B cell development [15].
BCR signaling is involved in the prevention of naïve B cell premature activation and expansion of autoreactive clones. At the same time, low-level “tonic” BCR signaling is essential for naïve B cell survival [16]. BCR antigen binding promotes mature B cell activation and further differentiation of naïve B cells via activation of PLC-γ2, PI3K/AKT and MAPK signaling pathways [17]. To ensure proper functionality of activated B cells, BCR signaling sustains survival, stimulates cell growth, and supports other related cellular adaptations via an array of signaling cascades including Ca2+ signaling, NF-κB activation, PI3K/AKT/mTOR, NFAT, ERK, and MAPK signaling [18]. Furthermore, BCR is vital for antigen presentation and subsequent T cell response activation and for B cell differentiation into antibody-producing plasma cells [19]. In particular, BCR-mediated antigen internalization is followed by intracellular antigen processing and subsequent surface presentation to CD4+ and CD8+ T cells [20]. BCR signaling also critically regulates activation-induced cytidine deaminase (AID)-mediated immunoglobulin class switch recombination [21][22]. Combination of activated BCR signaling with either a T cell-dependent (follicular T helpers) or T cell-independent (lipopolysaccharides or glycolipids) signal is crucial for B cell differentiation into antibody-secreting plasma cells or memory B cells [23]. BCR signaling driven plasma cell differentiation requires transcription factor Ets1 downregulation via Lyn-, PI3K-, BTK-, IKK2- and JNK-dependent pathways [24].
Additionally, BCR signaling contributes to the regulation of multiple other cellular processes in normal B cells including metabolism. For instance, it induces PI3K/AKT-dependent activation of glycolysis, oxidative phosphorylation, and glucose uptake [25][26]. BCR signaling also activates c-Myc with resulting enhancement of glycolysis and mitochondrial biogenesis [27]. BCR-initiated Ca2+ mobilization regulates metabolic reprogramming of naïve B cells which is required for their growth and further differentiation [18]. Maintenance of the balance between cellular growth and catabolic and anabolic processes is critical for correct B cell functionality and is primarily sustained via c-Myc and mTORC1 activity (which are both adjusted through BCR signaling) [28]. Importantly, BCR signaling was also implicated in metabolic regulation via autophagy upregulation [29]. BCR-mediated autophagy has been reported to be required for B cell activation [28].
Besides survival, activation, and proliferation, BCR signaling may prime B cells to anergy and cellular death to ensure B cell tolerance [30]. For instance, BCR signaling has been suggested to serve as a B cell quality control. Only moderate-intensity BCR signaling promotes positive selection, while BCR ligation downregulates BCR expression, reduces pro-survival PI3K/AKT signaling, and provides negative selection [31]. Inappropriately activated BCR may lead to B cell apoptosis [32]. BCR-mediated pro-apoptotic signaling has been associated with Ca2+-dependent and mitochondrial pathways [30].
Therefore, not only the type of BCR signaling, but also its intensity varies during B cell development and can determine the cell fate of B cells and their involvement in the immune response.

3. B-Cell Derived Malignancies and BCR Signaling

According to the recently updated 5th classification of lymphoid neoplasms (the World Health Organization Classification of Haematolymphoid Tumours), B-cell malignancies include the following categories: tumor-like lesions with B-cell predominance, precursor B-cell neoplasms (B lymphoblastic leukemias), mature B-cell neoplasms, and plasma cell neoplasms and other diseases with paraproteins [33]. Mature B-cell neoplasms include, e.g., pre-neoplastic and neoplastic small lymphocytic proliferations (e.g., chronic lymphocytic leukemia, CLL), multiple types of non-Hodgkin lymphomas (NHLs), and Hodgkin lymphomas [33]. The most common subtypes of NHL are diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL), diagnosed in approximately 25–30% and 20% of NHL patients, respectively [34][35][36][37]. In most cases, B-cell-derived tumors retain surface expression of BCR which variably supports malignant cell growth and survival [38][39]. BCR signaling has been shown to drive the growth and evolution of B-cell acute lymphoblastic leukemia (B-ALL), chronic lymphocytic leukemia (CLL), and multiple types of NHLs [12][40][41][42]. Pathogenic BCR signaling has been extensively studied and clearly demonstrated for DLBCL. Additionally, there is evidence that BCR supports tumor cell growth and survival in mantle cell lymphoma (MCL), FL, Burkitt’s lymphoma, and marginal zone lymphoma [43][44][45][46].
BCR signaling supports tumor cell growth and survival via various mechanisms. The first described tumorigenic mode of BCR signaling was the so-called “chronic active” BCR signaling triggered by self-antigen binding. Chronic active BCR signaling supports the viability and growth of malignant B cells mainly through the NF-κB signaling pathway [47][48]. Recently, it was shown that frequent lymphoma-associated mutations of MYD88 (myeloid differentiation primary response 88) adaptor protein lead to its spontaneous association with Toll-like receptor 9 (TLR9) and BCR, forming a My-T-BCR complex capable to trigger NF-κB activation [49]. Moreover, it was shown that lymphoma growth is also supported by antigen-independent, constitutive, lower intensity “tonic” BCR signaling. Tonic BCR signaling supports the growth and survival of tumor cells mostly via the PI3K/AKT/FOXO1 signaling pathway [50][51]. Antigen-independent cell autonomous BCR signaling with features of antigen-triggered BCR signaling was identified in CLL [40]. Importantly, in DLBCL, the type of BCR signaling (antigen driven or similar vs. tonic) reflects gene expression profiling-based cell-of-origin classification into the activated B cell like (ABC) DLBCL subtype and germinal center B cell like (GCB) DLBCL subtype, respectively [52].
Given the importance of BCR signaling in B-cell derived malignancies, its inhibition is one of the novel therapeutic approaches. It is represented mainly by three BTK (Bruton’s tyrosine kinase) inhibitors (ibrutinib, acalabrutinib, and zanubrutinib) approved and frequently used in the treatment of certain B-cell derived neoplasms. BTK inhibitors are effective; however, their toxicity and common resistance development represent substantial challenges that motivate the search for additional BCR signaling targeted inhibitors [53].
Important considerations regarding types of BCR signaling come from genomic studies, as documented in DLBCL. Distinct patterns of BCR signaling are reflected in tumor mutational patterns, which further expand the above-mentioned cell-of-origin DLBCL classification. Based on the spectrum of somatic alterations, genomic studies identified five to seven distinct genetic DLBCL subtypes [54][55][56][57][58]. The MCD (combined MYD88L265P and CD79B mutations), N1 (mutated NOTCH1), and A53 (aneuploid and TP53 inactivation) subtypes are significantly overlapping with the ABC DLBCL subtype, whereas EZB (mutated EZH2 and translocated BCL2), ST2 (mutated SGK1 and TET2), and BN2 (translocated BCL6 and mutated NOTCH2) are overlapping with GCB DLBCL [56]. Alternative classifications were published by Chapuy et al., including clusters 1 to 5 (BN2-DLBCL, A53-DLBCL, EZB-DLBCL, ST2-DLBCL, and MCD-DLBCL, respectively); and by Lacy et al., including MYD88, BCL2, SOCS1/SGK1, TET2/SGK1, and NOTCH2 clusters [55][58]. Pedrosa et al. later attempted to unite and simplify the existing classifications through the assessment of the mutational status of only 26 genes and BCL2 and BCL6 translocation status to facilitate their clinical implementation (two-step genetic DLBCL classifier; 2-S). The suggested 2-S subtypes are N12-S, EZB2-S, MCD2-S, BN22-S, and ST22-S [54]. Importantly, none of the above-mentioned genetic studies were able to assign all cases, leaving a substantial proportion of tumors unclassified. On the other hand, genetic studies provided insights into the contribution of CD79a and CD79b (and their mutations) towards tumorigenesis and lymphoma development [54][55][56][57][58].

This entry is adapted from the peer-reviewed paper 10.3390/ijms25010010

References

  1. Tanaka, S.; Baba, Y. B Cell Receptor Signaling. Adv. Exp. Med. Biol. 2020, 1254, 23–36.
  2. Huse, K.; Bai, B.; Hilden, V.I.; Bollum, L.K.; Våtsveen, T.K.; Munthe, L.A.; Smeland, E.B.; Irish, J.M.; Wälchli, S.; Myklebust, J.H. Mechanism of CD79A and CD79B Support for IgM+ B Cell Fitness through B Cell Receptor Surface Expression. J. Immunol. 2022, 209, 2042–2053.
  3. Chen, R.; Zhou, D.; Wang, L.; Zhu, L.; Ye, X. MYD88(L265P) and CD79B double mutations type (MCD type) of diffuse large B-cell lymphoma: Mechanism, clinical characteristics, and targeted therapy. Ther. Adv. Hematol. 2022, 13, 20406207211072839.
  4. Feng, Y.; Wang, Y.; Zhang, S.; Haneef, K.; Liu, W. Structural and immunogenomic insights into B-cell receptor activation. J. Genet. Genom. 2020, 47, 27–35.
  5. Avalos, A.M.; Ploegh, H.L. Early BCR Events and Antigen Capture, Processing, and Loading on MHC Class II on B Cells. Front. Immunol. 2014, 5, 92.
  6. Srinivasan, L.; Sasaki, Y.; Calado, D.P.; Zhang, B.; Paik, J.H.; DePinho, R.A.; Kutok, J.L.; Kearney, J.F.; Otipoby, K.L.; Rajewsky, K. PI3 kinase signals BCR-dependent mature B cell survival. Cell 2009, 139, 573–586.
  7. Burger, J.A.; Wiestner, A. Targeting B cell receptor signalling in cancer: Preclinical and clinical advances. Nat. Rev. Cancer 2018, 18, 148–167.
  8. Young, R.M.; Staudt, L.M. Targeting pathological B cell receptor signalling in lymphoid malignancies. Nat. Rev. Drug Discov. 2013, 12, 229–243.
  9. Puri, K.D.; Di Paolo, J.A.; Gold, M.R. B-cell receptor signaling inhibitors for treatment of autoimmune inflammatory diseases and B-cell malignancies. Int. Rev. Immunol. 2013, 32, 397–427.
  10. Rawlings, D.J.; Metzler, G.; Wray-Dutra, M.; Jackson, S.W. Altered B cell signalling in autoimmunity. Nat. Rev. Immunol. 2017, 17, 421–436.
  11. Liu, W.; Tolar, P.; Song, W.; Kim, T.J. Editorial: BCR Signaling and B Cell Activation. Front. Immunol. 2020, 11, 45.
  12. Köhrer, S.; Havranek, O.; Seyfried, F.; Hurtz, C.; Coffey, G.P.; Kim, E.; Ten Hacken, E.; Jäger, U.; Vanura, K.; O’Brien, S.; et al. Pre-BCR signaling in precursor B-cell acute lymphoblastic leukemia regulates PI3K/AKT, FOXO1 and MYC, and can be targeted by SYK inhibition. Leukemia 2016, 30, 1246–1254.
  13. Zhang, M.; Srivastava, G.; Lu, L. The pre-B cell receptor and its function during B cell development. Cell Mol. Immunol. 2004, 1, 89–94.
  14. Winkler, T.H.; Mårtensson, I.-L. The Role of the Pre-B Cell Receptor in B Cell Development, Repertoire Selection, and Tolerance. Front. Immunol. 2018, 9, 2423.
  15. Keren, Z.; Melamed, D. Antigen receptor signaling competence and the determination of B cell fate in B-lymphopoiesis. Histol. Histopathol. 2005, 20, 187–196.
  16. Neys, S.F.H.; Heukels, P.; van Hulst, J.A.C.; Rip, J.; Wijsenbeek, M.S.; Hendriks, R.W.; Corneth, O.B.J. Aberrant B Cell Receptor Signaling in Naïve B Cells from Patients with Idiopathic Pulmonary Fibrosis. Cells 2021, 10, 1321.
  17. Wen, Y.; Jing, Y.; Yang, L.; Kang, D.; Jiang, P.; Li, N.; Cheng, J.; Li, J.; Li, X.; Peng, Z.; et al. The regulators of BCR signaling during B cell activation. Blood Sci. 2019, 1, 119–129.
  18. Berry, C.T.; Liu, X.; Myles, A.; Nandi, S.; Chen, Y.H.; Hershberg, U.; Brodsky, I.E.; Cancro, M.P.; Lengner, C.J.; May, M.J.; et al. BCR-Induced Ca2+ Signals Dynamically Tune Survival, Metabolic Reprogramming, and Proliferation of Naive B Cells. Cell Rep. 2020, 31, 107474.
  19. McShane, A.N.; Malinova, D. The Ins and Outs of Antigen Uptake in B cells. Front. Immunol. 2022, 13, 892169.
  20. Rastogi, I.; Jeon, D.; Moseman, J.E.; Muralidhar, A.; Potluri, H.K.; McNeel, D.G. Role of B cells as antigen presenting cells. Front. Immunol. 2022, 13, 954936.
  21. Chen, Z.; Wang, J.H. How the Signaling Crosstalk of B Cell Receptor (BCR) and Co-Receptors Regulates Antibody Class Switch Recombination: A New Perspective of Checkpoints of BCR Signaling. Front. Immunol. 2021, 12, 663443.
  22. Vlachiotis, S.; Abolhassani, H. Transcriptional regulation of B cell class-switch recombination: The role in development of noninfectious complications. Expert Rev. Clin. Immunol. 2022, 18, 1145–1154.
  23. Tsai, D.-Y.; Hung, K.-H.; Chang, C.-W.; Lin, K.-I. Regulatory mechanisms of B cell responses and the implication in B cell-related diseases. J. Biomed. Sci. 2019, 26, 64.
  24. Luo, W.; Mayeux, J.; Gutierrez, T.; Russell, L.; Getahun, A.; Müller, J.; Tedder, T.; Parnes, J.; Rickert, R.; Nitschke, L.; et al. A balance between B cell receptor and inhibitory receptor signaling controls plasma cell differentiation by maintaining optimal Ets1 levels. J. Immunol. 2014, 193, 909–920.
  25. Kluckova, K.; D’Avola, A.; Riches, J.C. Advances in Understanding of Metabolism of B-Cell Lymphoma: Implications for Therapy. Cancers 2022, 14, 5552.
  26. Doughty, C.A.; Bleiman, B.F.; Wagner, D.J.; Dufort, F.J.; Mataraza, J.M.; Roberts, M.F.; Chiles, T.C. Antigen receptor-mediated changes in glucose metabolism in B lymphocytes: Role of phosphatidylinositol 3-kinase signaling in the glycolytic control of growth. Blood 2006, 107, 4458–4465.
  27. Iperi, C.; Bordron, A.; Dueymes, M.; Pers, J.-O.; Jamin, C. Metabolic Program of Regulatory B Lymphocytes and Influence in the Control of Malignant and Autoimmune Situations. Front. Immunol. 2021, 12, 735463.
  28. Raza, I.G.A.; Clarke, A.J. B Cell Metabolism and Autophagy in Autoimmunity. Front. Immunol. 2021, 12, 681105.
  29. Watanabe, K.; Tsubata, T. Autophagy connects antigen receptor signaling to costimulatory signaling in B lymphocytes. Autophagy 2009, 5, 108–110.
  30. Eeva, J.; Pelkonen, J. Mechanisms of B cell receptor induced apoptosis. Apoptosis 2004, 9, 525–531.
  31. Nemazee, D. Mechanisms of central tolerance for B cells. Nat. Rev. Immunol. 2017, 17, 281–294.
  32. Yam-Puc, J.C.; Zhang, L.; Maqueda-Alfaro, R.A.; Garcia-Ibanez, L.; Zhang, Y.; Davies, J.; Senis, Y.A.; Snaith, M.; Toellner, K.-M. Enhanced BCR signaling inflicts early plasmablast and germinal center B cell death. iScience 2021, 24, 102038.
  33. Alaggio, R.; Amador, C.; Anagnostopoulos, I.; Attygalle, A.D.; Araujo, I.B.O.; Berti, E.; Bhagat, G.; Borges, A.M.; Boyer, D.; Calaminici, M.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid Neoplasms. Leukemia 2022, 36, 1720–1748.
  34. Susanibar-Adaniya, S.; Barta, S.K. 2021 Update on Diffuse large B cell lymphoma: A review of current data and potential applications on risk stratification and management. Am. J. Hematol. 2021, 96, 617–629.
  35. Liu, Y.; Barta, S.K. Diffuse large B-cell lymphoma: 2019 update on diagnosis, risk stratification, and treatment. Am. J. Hematol. 2019, 94, 604–616.
  36. Gupta, G.; Garg, V.; Mallick, S.; Gogia, A. Current trends in diagnosis and management of follicular lymphoma. Am. J. Blood Res. 2022, 12, 105–124.
  37. Carbone, A.; Roulland, S.; Gloghini, A.; Younes, A.; von Keudell, G.; López-Guillermo, A.; Fitzgibbon, J. Follicular lymphoma. Nat. Rev. Dis. Primers 2019, 5, 83.
  38. Profitós-Pelejà, N.; Santos, J.C.; Marín-Niebla, A.; Roué, G.; Ribeiro, M.L. Regulation of B-Cell Receptor Signaling and Its Therapeutic Relevance in Aggressive B-Cell Lymphomas. Cancers 2022, 14, 860.
  39. Niemann, C.U.; Wiestner, A. B-cell receptor signaling as a driver of lymphoma development and evolution. Semin. Cancer Biol. 2013, 23, 410–421.
  40. Dühren-von Minden, M.; Übelhart, R.; Schneider, D.; Wossning, T.; Bach, M.P.; Buchner, M.; Hofmann, D.; Surova, E.; Follo, M.; Köhler, F.; et al. Chronic lymphocytic leukaemia is driven by antigen-independent cell-autonomous signalling. Nature 2012, 489, 309–312.
  41. Young, R.M.; Phelan, J.D.; Wilson, W.H.; Staudt, L.M. Pathogenic B-cell receptor signaling in lymphoid malignancies: New insights to improve treatment. Immunol. Rev. 2019, 291, 190–213.
  42. Valla, K.; Flowers, C.R.; Koff, J.L. Targeting the B cell receptor pathway in non-Hodgkin lymphoma. Expert Opin. Investig. Drugs 2018, 27, 513–522.
  43. Fichtner, M.; Dreyling, M.; Binder, M.; Trepel, M. The role of B cell antigen receptors in mantle cell lymphoma. J. Hematol. Oncol. 2017, 10, 164.
  44. Sachen, K.L.; Strohman, M.J.; Singletary, J.; Alizadeh, A.A.; Kattah, N.H.; Lossos, C.; Mellins, E.D.; Levy, S.; Levy, R. Self-antigen recognition by follicular lymphoma B-cell receptors. Blood 2012, 120, 4182–4190.
  45. Corso, J.; Pan, K.T.; Walter, R.; Doebele, C.; Mohr, S.; Bohnenberger, H.; Ströbel, P.; Lenz, C.; Slabicki, M.; Hüllein, J.; et al. Elucidation of tonic and activated B-cell receptor signaling in Burkitt’s lymphoma provides insights into regulation of cell survival. Proc. Natl. Acad. Sci. USA 2016, 113, 5688–5693.
  46. Noy, A.; de Vos, S.; Thieblemont, C.; Martin, P.; Flowers, C.R.; Morschhauser, F.; Collins, G.P.; Ma, S.; Coleman, M.; Peles, S.; et al. Targeting Bruton tyrosine kinase with ibrutinib in relapsed/refractory marginal zone lymphoma. Blood 2017, 129, 2224–2232.
  47. Bogusz, A.M.; Baxter, R.H.; Currie, T.; Sinha, P.; Sohani, A.R.; Kutok, J.L.; Rodig, S.J. Quantitative immunofluorescence reveals the signature of active B-cell receptor signaling in diffuse large B-cell lymphoma. Clin. Cancer Res. 2012, 18, 6122–6135.
  48. Davis, R.E.; Ngo, V.N.; Lenz, G.; Tolar, P.; Young, R.M.; Romesser, P.B.; Kohlhammer, H.; Lamy, L.; Zhao, H.; Yang, Y.; et al. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature 2010, 463, 88–92.
  49. Phelan, J.D.; Young, R.M.; Webster, D.E.; Roulland, S.; Wright, G.W.; Kasbekar, M.; Shaffer, A.L., 3rd; Ceribelli, M.; Wang, J.Q.; Schmitz, R.; et al. A multiprotein supercomplex controlling oncogenic signalling in lymphoma. Nature 2018, 560, 387–391.
  50. Havranek, O.; Xu, J.; Köhrer, S.; Wang, Z.; Becker, L.; Comer, J.M.; Henderson, J.; Ma, W.; Man Chun Ma, J.; Westin, J.R.; et al. Tonic B-cell receptor signaling in diffuse large B-cell lymphoma. Blood 2017, 130, 995–1006.
  51. Myers, D.R.; Zikherman, J.; Roose, J.P. Tonic Signals: Why Do Lymphocytes Bother? Trends Immunol. 2017, 38, 844–857.
  52. Alizadeh, A.A.; Eisen, M.B.; Davis, R.E.; Ma, C.; Lossos, I.S.; Rosenwald, A.; Boldrick, J.C.; Sabet, H.; Tran, T.; Yu, X.; et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 2000, 403, 503–511.
  53. Frustaci, A.M.; Deodato, M.; Zamprogna, G.; Cairoli, R.; Montillo, M.; Tedeschi, A. Next Generation BTK Inhibitors in CLL: Evolving Challenges and New Opportunities. Cancers 2023, 15, 1504.
  54. Pedrosa, L.; Fernández-Miranda, I.; Pérez-Callejo, D.; Quero, C.; Rodríguez, M.; Martín-Acosta, P.; Gómez, S.; González-Rincón, J.; Santos, A.; Tarin, C.; et al. Proposal and validation of a method to classify genetic subtypes of diffuse large B cell lymphoma. Sci. Rep. 2021, 11, 1886.
  55. Lacy, S.E.; Barrans, S.L.; Beer, P.A.; Painter, D.; Smith, A.G.; Roman, E.; Cooke, S.L.; Ruiz, C.; Glover, P.; Van Hoppe, S.J.L.; et al. Targeted sequencing in DLBCL, molecular subtypes, and outcomes: A Haematological Malignancy Research Network report. Blood 2020, 135, 1759–1771.
  56. Wright, G.W.; Huang, D.W.; Phelan, J.D.; Coulibaly, Z.A.; Roulland, S.; Young, R.M.; Wang, J.Q.; Schmitz, R.; Morin, R.D.; Tang, J.; et al. A Probabilistic Classification Tool for Genetic Subtypes of Diffuse Large B Cell Lymphoma with Therapeutic Implications. Cancer Cell 2020, 37, 551–568.e14.
  57. Schmitz, R.; Wright, G.W.; Huang, D.W.; Johnson, C.A.; Phelan, J.D.; Wang, J.Q.; Roulland, S.; Kasbekar, M.; Young, R.M.; Shaffer, A.L.; et al. Genetics and Pathogenesis of Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2018, 378, 1396–1407.
  58. Chapuy, B.; Stewart, C.; Dunford, A.J.; Kim, J.; Kamburov, A.; Redd, R.A.; Lawrence, M.S.; Roemer, M.G.M.; Li, A.J.; Ziepert, M.; et al. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat. Med. 2018, 24, 679–690.
More
This entry is offline, you can click here to edit this entry!
Video Production Service