Role of Bruton's tyrosine kinase in B cells and malignancies

Simar Pal Singh, Floris Dammeijer, Rudi W Hendriks, Simar Pal Singh, Floris Dammeijer, Rudi W Hendriks

Abstract

Bruton's tyrosine kinase (BTK) is a non-receptor kinase that plays a crucial role in oncogenic signaling that is critical for proliferation and survival of leukemic cells in many B cell malignancies. BTK was initially shown to be defective in the primary immunodeficiency X-linked agammaglobulinemia (XLA) and is essential both for B cell development and function of mature B cells. Shortly after its discovery, BTK was placed in the signal transduction pathway downstream of the B cell antigen receptor (BCR). More recently, small-molecule inhibitors of this kinase have shown excellent anti-tumor activity, first in animal models and subsequently in clinical studies. In particular, the orally administered irreversible BTK inhibitor ibrutinib is associated with high response rates in patients with relapsed/refractory chronic lymphocytic leukemia (CLL) and mantle-cell lymphoma (MCL), including patients with high-risk genetic lesions. Because ibrutinib is generally well tolerated and shows durable single-agent efficacy, it was rapidly approved for first-line treatment of patients with CLL in 2016. To date, evidence is accumulating for efficacy of ibrutinib in various other B cell malignancies. BTK inhibition has molecular effects beyond its classic role in BCR signaling. These involve B cell-intrinsic signaling pathways central to cellular survival, proliferation or retention in supportive lymphoid niches. Moreover, BTK functions in several myeloid cell populations representing important components of the tumor microenvironment. As a result, there is currently a considerable interest in BTK inhibition as an anti-cancer therapy, not only in B cell malignancies but also in solid tumors. Efficacy of BTK inhibition as a single agent therapy is strong, but resistance may develop, fueling the development of combination therapies that improve clinical responses. In this review, we discuss the role of BTK in B cell differentiation and B cell malignancies and highlight the importance of BTK inhibition in cancer therapy.

Keywords: B cell development; B cell receptor signaling; Bruton’s tyrosine kinase; Chemokine receptor; Chronic lymphocytic leukemia; Ibrutinib; Leukemia; Lymphoma; Tumor microenvironment.

Conflict of interest statement

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

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

Figures

Fig. 1
Fig. 1
Domain structure of TEC kinase family members and key interacting partners of Bruton’s tyrosine kinase. a Schematic overview of the protein structure of BTK and other TEC kinase family members. Shown are five different domains, as explained in text, the Y223 autophosphorylation site, the Y551 phosphorylation site that activates BTK, and the C481 binding site of ibrutinib. b Schematic overview of the protein structure of key interacting partners of BTK. PH, pleckstrin homology; TH, TEC homology; BH, BTK homology; PRR, proline rich domain; SH2/SH3, SRC homology domains 2 and 3; Cys, cysteine-string motif
Fig. 2
Fig. 2
Role of Bruton’s tyrosine kinase downstream of the B cell receptor. Signaling cascade showing important events downstream of B cell receptor (BCR). Antigen engagement by the BCR results in the formation of a micro-signalosome whereby BTK activates four families of non-receptor protein tyrosine kinases that transduce key signaling events including phospholipase Cγ, mitogen-activated protein kinase (MAPK) activation, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-кB) pathway components and activation of the serine/threonine kinase AKT (PKB). In addition, BTK mediated signaling events are regulated by various phosphatases that can be recruited to the cell membrane, following crosslinking of inhibitory receptors, e.g., FcγRIIB that is exclusively expressed on B cells and signals upon immune complex binding. See text for details
Fig. 3
Fig. 3
Role of Bruton’s tyrosine kinase downstream of chemokine receptors, Toll-like receptors and activating Fcγ receptors. Signaling cascade showing important events downstream of (a) Chemokine receptors (e.g. CXCR4): upon chemokine binding to the extracellular domain Gα and Gβy subunits can independently activate PI3K, which results in activation of BTK, AKT and MAPK-dependent pathways. b Toll-like receptors: upon ligand recognition TLRs recruit different proteins including TIR, MYD88, IRAK1 and TIRAP/MAL, all of which interact with BTK and induce downstream activation of the transcription factor NF-κB. c Activating Fc receptors (e.g. FcγRI): Following FcγRI cross-linking, Src-kinases, SYK, PI3K-γ and BTK are activated. In contrast, inhibitory Fc-receptors (FcγRIIB) containing ITIM domains recruit phosphatases and reduce BTK activation (Fig. 2). See text for details
Fig. 4
Fig. 4
Stages of B cell differentiation and associated malignancies. Model of B cell development indicating different stages of B cell differentiation and important immune checkpoints where BTK plays a key role. Various B-cell malignancies are indicated, which are associated with abnormal BTK signaling at distinct stages of B-cell differentiation and activation. Note that the cellular origin of U-CLL is thought to be CD5+ mature B cells. Somatic hypermutation status of BCR and gene expression profiling indicates post-germinal center (GC) origin of M-CLL. See text for detailed information. CLP, common lymphoid progenitor; CSR, class switch recombination; FDC, follicular dendritic cell; SHM, somatic hypermutation

References

    1. Gross S, Rahal R, Stransky N, Lengauer C, Hoeflich KP. Targeting cancer with kinase inhibitors. J Clin Invest. 2015;125:1780–1789. doi: 10.1172/JCI76094.
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013.
    1. Vetrie D, Vorechovsky I, Sideras P, Holland J, Davies A, Flinter F, Hammarstrom L, Kinnon C, Levinsky R, Bobrow M, et al. The gene involved in X-linked agammaglobulinaemia is a member of the src family of protein-tyrosine kinases. Nature. 1993;361:226–233. doi: 10.1038/361226a0.
    1. Tsukada S, Saffran DC, Rawlings DJ, Parolini O, Allen RC, Klisak I, Sparkes RS, Kubagawa H, Mohandas T, Quan S, et al. Deficient expression of a B cell cytoplasmic tyrosine kinase in human X-linked agammaglobulinemia. Cell. 1993;72:279–290. doi: 10.1016/0092-8674(93)90667-F.
    1. Rawlings DJ, Saffran DC, Tsukada S, Largaespada DA, Grimaldi JC, Cohen L, Mohr RN, Bazan JF, Howard M, Copeland NG, et al. Mutation of unique region of Bruton's tyrosine kinase in immunodeficient XID mice. Science. 1993;261:358–361. doi: 10.1126/science.8332901.
    1. Thomas JD, Sideras P, Smith CI, Vorechovsky I, Chapman V, Paul WE. Colocalization of X-linked agammaglobulinemia and X-linked immunodeficiency genes. Science. 1993;261:355–358. doi: 10.1126/science.8332900.
    1. Scher I. The CBA/N mouse strain: an experimental model illustrating the influence of the X-chromosome on immunity. Adv Immunol. 1982;33:1–71. doi: 10.1016/S0065-2776(08)60834-2.
    1. Khan WN, Alt FW, Gerstein RM, Malynn BA, Larsson I, Rathbun G, Davidson L, Muller S, Kantor AB, Herzenberg LA, et al. Defective B cell development and function in Btk-deficient mice. Immunity. 1995;3:283–299. doi: 10.1016/1074-7613(95)90114-0.
    1. Hendriks RW, de Bruijn MF, Maas A, Dingjan GM, Karis A, Grosveld F. Inactivation of Btk by insertion of lacZ reveals defects in B cell development only past the pre-B cell stage. EMBO J. 1996;15:4862–4872.
    1. Middendorp S, Dingjan GM, Hendriks RW. Impaired precursor B cell differentiation in Bruton's tyrosine kinase-deficient mice. J Immunol. 2002;168:2695–2703. doi: 10.4049/jimmunol.168.6.2695.
    1. Byrd JC, Furman RR, Coutre SE, Flinn IW, Burger JA, Blum KA, Grant B, Sharman JP, Coleman M, Wierda WG, et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med. 2013;369:32–42. doi: 10.1056/NEJMoa1215637.
    1. Byrd JC, Harrington B, O'Brien S, Jones JA, Schuh A, Devereux S, Chaves J, Wierda WG, Awan FT, Brown JR, et al. Acalabrutinib (ACP-196) in relapsed chronic lymphocytic leukemia. N Engl J Med. 2016;374:323–332. doi: 10.1056/NEJMoa1509981.
    1. Burger JA, Tedeschi A, Barr PM, Robak T, Owen C, Ghia P, Bairey O, Hillmen P, Bartlett NL, Li J, et al. Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med. 2015;373:2425–2437. doi: 10.1056/NEJMoa1509388.
    1. Hendriks RW, Yuvaraj S, Kil LP. Targeting Bruton's tyrosine kinase in B cell malignancies. Nat Rev Cancer. 2014;14:219–232. doi: 10.1038/nrc3702.
    1. Kawakami Y, Yao L, Miura T, Tsukada S, Witte ON, Kawakami T. Tyrosine phosphorylation and activation of Bruton tyrosine kinase upon fc epsilon RI cross-linking. Mol Cell Biol. 1994;14:5108–5113. doi: 10.1128/MCB.14.8.5108.
    1. Hata D, Kawakami Y, Inagaki N, Lantz CS, Kitamura T, Khan WN, Maeda-Yamamoto M, Miura T, Han W, Hartman SE, et al. Involvement of Bruton's tyrosine kinase in FcepsilonRI-dependent mast cell degranulation and cytokine production. J Exp Med. 1998;187:1235–1247. doi: 10.1084/jem.187.8.1235.
    1. Jongstra-Bilen J, Puig Cano A, Hasija M, Xiao H, Smith CI, Cybulsky MI. Dual functions of Bruton's tyrosine kinase and Tec kinase during Fcgamma receptor-induced signaling and phagocytosis. J Immunol. 2008;181:288–298. doi: 10.4049/jimmunol.181.1.288.
    1. Wang D, Feng J, Wen R, Marine JC, Sangster MY, Parganas E, Hoffmeyer A, Jackson CW, Cleveland JL, Murray PJ, Ihle JN. Phospholipase Cgamma2 is essential in the functions of B cell and several fc receptors. Immunity. 2000;13:25–35. doi: 10.1016/S1074-7613(00)00005-4.
    1. Shinohara M, Koga T, Okamoto K, Sakaguchi S, Arai K, Yasuda H, Takai T, Kodama T, Morio T, Geha RS, et al. Tyrosine kinases Btk and Tec regulate osteoclast differentiation by linking RANK and ITAM signals. Cell. 2008;132:794–806. doi: 10.1016/j.cell.2007.12.037.
    1. Oda A, Ikeda Y, Ochs HD, Druker BJ, Ozaki K, Handa M, Ariga T, Sakiyama Y, Witte ON, Wahl MI. Rapid tyrosine phosphorylation and activation of Bruton's tyrosine/Tec kinases in platelets induced by collagen binding or CD32 cross-linking. Blood. 2000;95:1663–1670.
    1. Ito M, Shichita T, Okada M, Komine R, Noguchi Y, Yoshimura A, Morita R. Bruton's tyrosine kinase is essential for NLRP3 inflammasome activation and contributes to ischaemic brain injury. Nat Commun. 2015;6:7360. doi: 10.1038/ncomms8360.
    1. Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41:49–61. doi: 10.1016/j.immuni.2014.06.010.
    1. Mantovani A, Marchesi F, Malesci A, Laghi L, Allavena P. Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol. 2017;14:399–416. doi: 10.1038/nrclinonc.2016.217.
    1. Kokabee L, Wang X, Sevinsky CJ, Wang WL, Cheu L, Chittur SV, Karimipoor M, Tenniswood M, Conklin DS. Bruton's tyrosine kinase is a potential therapeutic target in prostate cancer. Cancer Biol Ther. 2015;16:1604–1615. doi: 10.1080/15384047.2015.1078023.
    1. Grassilli E, Pisano F, Cialdella A, Bonomo S, Missaglia C, Cerrito MG, Masiero L, Ianzano L, Giordano F, Cicirelli V, et al. A novel oncogenic BTK isoform is overexpressed in colon cancers and required for RAS-mediated transformation. Oncogene. 2016;35:4368–4378. doi: 10.1038/onc.2015.504.
    1. Zucha MA, Wu AT, Lee WH, Wang LS, Lin WW, Yuan CC, Yeh CT. Bruton's tyrosine kinase (Btk) inhibitor ibrutinib suppresses stem-like traits in ovarian cancer. Oncotarget. 2015;6:13255–13268. doi: 10.18632/oncotarget.3658.
    1. Wei L, Su YK, Lin CM, Chao TY, Huang SP, Huynh TT, Jan HJ, Whang-Peng J, Chiou JF, Wu AT, Hsiao M. Preclinical investigation of ibrutinib, a Bruton's kinase tyrosine (Btk) inhibitor, in suppressing glioma tumorigenesis and stem cell phenotypes. Oncotarget. 2016;7:69961–69975.
    1. Bradshaw JM. The Src, Syk, and Tec family kinases: distinct types of molecular switches. Cell Signal. 2010;22:1175–1184. doi: 10.1016/j.cellsig.2010.03.001.
    1. Hyvonen M, Saraste M. Structure of the PH domain and Btk motif from Bruton's tyrosine kinase: molecular explanations for X-linked agammaglobulinaemia. EMBO J. 1997;16:3396–3404. doi: 10.1093/emboj/16.12.3396.
    1. Rawlings DJ, Scharenberg AM, Park H, Wahl MI, Lin S, Kato RM, Fluckiger AC, Witte ON, Kinet JP. Activation of BTK by a phosphorylation mechanism initiated by SRC family kinases. Science. 1996;271:822–825. doi: 10.1126/science.271.5250.822.
    1. Park H, Wahl MI, Afar DE, Turck CW, Rawlings DJ, Tam C, Scharenberg AM, Kinet JP, Witte ON. Regulation of Btk function by a major autophosphorylation site within the SH3 domain. Immunity. 1996;4:515–525. doi: 10.1016/S1074-7613(00)80417-3.
    1. Marcotte DJ, Liu YT, Arduini RM, Hession CA, Miatkowski K, Wildes CP, Cullen PF, Hong V, Hopkins BT, Mertsching E, et al. Structures of human Bruton's tyrosine kinase in active and inactive conformations suggest a mechanism of activation for TEC family kinases. Protein Sci. 2010;19:429–439.
    1. Middendorp S, Dingjan GM, Maas A, Dahlenborg K, Hendriks RW. Function of Bruton's tyrosine kinase during B cell development is partially independent of its catalytic activity. J Immunol. 2003;171:5988–5996. doi: 10.4049/jimmunol.171.11.5988.
    1. Lam KP, Kuhn R, Rajewsky K. In Vivo ablation of surface immunoglobulin on mature B cells by inducible gene targeting results in rapid cell death. Cell. 1997;90:1073–1083. doi: 10.1016/S0092-8674(00)80373-6.
    1. Anderson JS, Teutsch M, Dong Z, Wortis HH. An essential role for Bruton's [corrected] tyrosine kinase in the regulation of B-cell apoptosis. Proc Natl Acad Sci U S A. 1996;93:10966–10971. doi: 10.1073/pnas.93.20.10966.
    1. Solvason N, Wu WW, Kabra N, Lund-Johansen F, Roncarolo MG, Behrens TW, Grillot DA, Nunez G, Lees E, Howard M. Transgene expression of bcl-xL permits anti-immunoglobulin (Ig)-induced proliferation in xid B cells. J Exp Med. 1998;187:1081–1091. doi: 10.1084/jem.187.7.1081.
    1. Brorson K, Brunswick M, Ezhevsky S, Wei DG, Berg R, Scott D, Stein KE. Xid affects events leading to B cell cycle entry. J Immunol. 1997;159:135–143.
    1. Glassford J, Soeiro I, Skarell SM, Banerji L, Holman M, Klaus GG, Kadowaki T, Koyasu S, Lam EW. BCR targets cyclin D2 via Btk and the p85alpha subunit of PI3-K to induce cell cycle progression in primary mouse B cells. Oncogene. 2003;22:2248–2259. doi: 10.1038/sj.onc.1206425.
    1. Spaargaren M, Beuling EA, Rurup ML, Meijer HP, Klok MD, Middendorp S, Hendriks RW, Pals ST. The B cell antigen receptor controls integrin activity through Btk and PLCgamma2. J Exp Med. 2003;198:1539–1550. doi: 10.1084/jem.20011866.
    1. Rolli V, Gallwitz M, Wossning T, Flemming A, Schamel WW, Zurn C, Reth M. Amplification of B cell antigen receptor signaling by a Syk/ITAM positive feedback loop. Mol Cell. 2002;10:1057–1069. doi: 10.1016/S1097-2765(02)00739-6.
    1. O'Rourke LM, Tooze R, Turner M, Sandoval DM, Carter RH, Tybulewicz VL, Fearon DT. CD19 as a membrane-anchored adaptor protein of B lymphocytes: costimulation of lipid and protein kinases by recruitment of Vav. Immunity. 1998;8:635–645. doi: 10.1016/S1074-7613(00)80568-3.
    1. Okada T, Maeda A, Iwamatsu A, Gotoh K, Kurosaki T. BCAP: the tyrosine kinase substrate that connects B cell receptor to phosphoinositide 3-kinase activation. Immunity. 2000;13:817–827. doi: 10.1016/S1074-7613(00)00079-0.
    1. Inabe K, Ishiai M, Scharenberg AM, Freshney N, Downward J, Kurosaki T. Vav3 modulates B cell receptor responses by regulating phosphoinositide 3-kinase activation. J Exp Med. 2002;195:189–200. doi: 10.1084/jem.20011571.
    1. Saito K, Scharenberg AM, Kinet JP. Interaction between the Btk PH domain and phosphatidylinositol-3,4,5-trisphosphate directly regulates Btk. J Biol Chem. 2001;276:16201–16206. doi: 10.1074/jbc.M100873200.
    1. Engels N, Konig LM, Heemann C, Lutz J, Tsubata T, Griep S, Schrader V, Wienands J. Recruitment of the cytoplasmic adaptor Grb2 to surface IgG and IgE provides antigen receptor-intrinsic costimulation to class-switched B cells. Nat Immunol. 2009;10:1018–1025. doi: 10.1038/ni.1764.
    1. Kuhn J, Wong LE, Pirkuliyeva S, Schulz K, Schwiegk C, Funfgeld KG, Keppler S, Batista FD, Urlaub H, Habeck M, et al. The adaptor protein CIN85 assembles intracellular signaling clusters for B cell activation. Sci Signal. 2016;9:ra66. doi: 10.1126/scisignal.aad6275.
    1. Guo B, Kato RM, Garcia-Lloret M, Wahl MI, Rawlings DJ. Engagement of the human pre-B cell receptor generates a lipid raft-dependent calcium signaling complex. Immunity. 2000;13:243–253. doi: 10.1016/S1074-7613(00)00024-8.
    1. Su YW, Zhang Y, Schweikert J, Koretzky GA, Reth M, Wienands J. Interaction of SLP adaptors with the SH2 domain of Tec family kinases. Eur J Immunol. 1999;29:3702–3711. doi: 10.1002/(SICI)1521-4141(199911)29:11<3702::AID-IMMU3702>;2-R.
    1. Chiu CW, Dalton M, Ishiai M, Kurosaki T, Chan AC. BLNK: molecular scaffolding through 'cis'-mediated organization of signaling proteins. EMBO J. 2002;21:6461–6472. doi: 10.1093/emboj/cdf658.
    1. Weber M, Treanor B, Depoil D, Shinohara H, Harwood NE, Hikida M, Kurosaki T, Batista FD. Phospholipase C-gamma2 and Vav cooperate within signaling microclusters to propagate B cell spreading in response to membrane-bound antigen. J Exp Med. 2008;205:853–868. doi: 10.1084/jem.20072619.
    1. Kim YJ, Sekiya F, Poulin B, Bae YS, Rhee SG. Mechanism of B-cell receptor-induced phosphorylation and activation of phospholipase C-gamma2. Mol Cell Biol. 2004;24:9986–9999. doi: 10.1128/MCB.24.22.9986-9999.2004.
    1. Hashimoto A, Okada H, Jiang A, Kurosaki M, Greenberg S, Clark EA, Kurosaki T. Involvement of guanosine triphosphatases and phospholipase C-gamma2 in extracellular signal-regulated kinase, c-Jun NH2-terminal kinase, and p38 mitogen-activated protein kinase activation by the B cell antigen receptor. J Exp Med. 1998;188:1287–1295. doi: 10.1084/jem.188.7.1287.
    1. Petro JB, Rahman SM, Ballard DW, Khan WN. Bruton’s tyrosine kinase is required for activation of IkappaB kinase and nuclear factor kappaB in response to B cell receptor engagement. J Exp Med. 2000;191:1745–1754. doi: 10.1084/jem.191.10.1745.
    1. Bajpai UD, Zhang K, Teutsch M, Sen R, Wortis HH. Bruton’s tyrosine kinase links the B cell receptor to nuclear factor kappaB activation. J Exp Med. 2000;191:1735–1744. doi: 10.1084/jem.191.10.1735.
    1. Manning BD, Toker A. AKT/PKB signaling: navigating the network. Cell. 2017;169:381–405. doi: 10.1016/j.cell.2017.04.001.
    1. Craxton A, Jiang A, Kurosaki T, Clark EA. Syk and Bruton's tyrosine kinase are required for B cell antigen receptor-mediated activation of the kinase Akt. J Biol Chem. 1999;274:30644–30650. doi: 10.1074/jbc.274.43.30644.
    1. Ellmeier W, Jung S, Sunshine MJ, Hatam F, Xu Y, Baltimore D, Mano H, Littman DR. Severe B cell deficiency in mice lacking the tec kinase family members Tec and Btk. J Exp Med. 2000;192:1611–1624. doi: 10.1084/jem.192.11.1611.
    1. de Bruijn MJ, Rip J, van der Ploeg EK, van Greuningen LW, Ta VT, Kil LP, Langerak AW, Rimmelzwaan GF, Ellmeier W, Hendriks RW, Corneth OB. Distinct and overlapping functions of TEC kinase and BTK in B cell receptor signaling. J Immunol. 2017;198:3058–3068. doi: 10.4049/jimmunol.1601285.
    1. Engels N, Konig LM, Schulze W, Radtke D, Vanshylla K, Lutz J, Winkler TH, Nitschke L, Wienands J. The immunoglobulin tail tyrosine motif upgrades memory-type BCRs by incorporating a Grb2-Btk signalling module. Nat Commun. 2014;5:5456. doi: 10.1038/ncomms6456.
    1. Nimmerjahn F, Ravetch JV. Fcgamma receptors as regulators of immune responses. Nat Rev Immunol. 2008;8:34–47. doi: 10.1038/nri2206.
    1. Amigorena S, Bonnerot C, Drake JR, Choquet D, Hunziker W, Guillet JG, Webster P, Sautes C, Mellman I, Fridman WH. Cytoplasmic domain heterogeneity and functions of IgG fc receptors in B lymphocytes. Science. 1992;256:1808–1812. doi: 10.1126/science.1535455.
    1. Muta T, Kurosaki T, Misulovin Z, Sanchez M, Nussenzweig MC, Ravetch JV. A 13-amino-acid motif in the cytoplasmic domain of fc gamma RIIB modulates B-cell receptor signalling. Nature. 1994;369:340. doi: 10.1038/369340a0.
    1. Bolland S, Ravetch JV. Inhibitory pathways triggered by ITIM-containing receptors. Adv Immunol. 1999;72:149–177. doi: 10.1016/S0065-2776(08)60019-X.
    1. Ono M, Bolland S, Tempst P, Ravetch JV. Role of the inositol phosphatase SHIP in negative regulation of the immune system by the receptor fc(gamma)RIIB. Nature. 1996;383:263–266. doi: 10.1038/383263a0.
    1. Ono M, Okada H, Bolland S, Yanagi S, Kurosaki T, Ravetch JV. Deletion of SHIP or SHP-1 reveals two distinct pathways for inhibitory signaling. Cell. 1997;90:293–301. doi: 10.1016/S0092-8674(00)80337-2.
    1. Liu W, Quinto I, Chen X, Palmieri C, Rabin RL, Schwartz OM, Nelson DL, Scala G. Direct inhibition of Bruton's tyrosine kinase by IBtk, a Btk-binding protein. Nat Immunol. 2001;2:939–946. doi: 10.1038/ni1001-939.
    1. Kang SW, Wahl MI, Chu J, Kitaura J, Kawakami Y, Kato RM, Tabuchi R, Tarakhovsky A, Kawakami T, Turck CW, et al. PKCbeta modulates antigen receptor signaling via regulation of Btk membrane localization. EMBO J. 2001;20:5692–5702. doi: 10.1093/emboj/20.20.5692.
    1. Belver L, de Yebenes VG, Ramiro AR. MicroRNAs prevent the generation of autoreactive antibodies. Immunity. 2010;33:713–722. doi: 10.1016/j.immuni.2010.11.010.
    1. Bottoni A, Rizzotto L, Lai TH, Liu C, Smith LL, Mantel R, Reiff S, El-Gamal D, Larkin K, Johnson AJ, et al. Targeting BTK through microRNA in chronic lymphocytic leukemia. Blood. 2016;128:3101–3112.
    1. Yu L, Mohamed AJ, Simonson OE, Vargas L, Blomberg KE, Bjorkstrand B, Arteaga HJ, Nore BF, Smith CI. Proteasome-dependent autoregulation of Bruton tyrosine kinase (Btk) promoter via NF-kappaB. Blood. 2008;111:4617–4626. doi: 10.1182/blood-2007-10-121137.
    1. Ritter SL, Hall RA. Fine-tuning of GPCR activity by receptor-interacting proteins. Nat Rev Mol Cell Biol. 2009;10:819–830. doi: 10.1038/nrm2803.
    1. Okada T, Ngo VN, Ekland EH, Forster R, Lipp M, Littman DR, Cyster JG. Chemokine requirements for B cell entry to lymph nodes and Peyer's patches. J Exp Med. 2002;196:65–75. doi: 10.1084/jem.20020201.
    1. Servant G, Weiner OD, Herzmark P, Balla T, Sedat JW, Bourne HR. Polarization of chemoattractant receptor signaling during neutrophil chemotaxis. Science. 2000;287:1037–1040. doi: 10.1126/science.287.5455.1037.
    1. Lowry WE, Huang XY, Protein G. Beta gamma subunits act on the catalytic domain to stimulate Bruton's agammaglobulinemia tyrosine kinase. J Biol Chem. 2002;277:1488–1492. doi: 10.1074/jbc.M110390200.
    1. Tsukada S, Simon MI, Witte ON, Katz A. Binding of beta gamma subunits of heterotrimeric G proteins to the PH domain of Bruton tyrosine kinase. Proc Natl Acad Sci U S A. 1994;91:11256–11260. doi: 10.1073/pnas.91.23.11256.
    1. Bence K, Ma W, Kozasa T, Huang XY. Direct stimulation of Bruton's tyrosine kinase by G(q)-protein alpha-subunit. Nature. 1997;389:296–299. doi: 10.1038/38520.
    1. de Gorter DJ, Beuling EA, Kersseboom R, Middendorp S, van Gils JM, Hendriks RW, Pals ST, Spaargaren M. Bruton's tyrosine kinase and phospholipase Cgamma2 mediate chemokine-controlled B cell migration and homing. Immunity. 2007;26:93–104. doi: 10.1016/j.immuni.2006.11.012.
    1. Rawlings DJ, Schwartz MA, Jackson SW, Meyer-Bahlburg A. Integration of B cell responses through toll-like receptors and antigen receptors. Nat Rev Immunol. 2012;12:282–294. doi: 10.1038/nri3190.
    1. Jefferies CA, Doyle S, Brunner C, Dunne A, Brint E, Wietek C, Walch E, Wirth T, O'Neill LA. Bruton's tyrosine kinase is a toll/interleukin-1 receptor domain-binding protein that participates in nuclear factor kappaB activation by toll-like receptor 4. J Biol Chem. 2003;278:26258–26264. doi: 10.1074/jbc.M301484200.
    1. Liu X, Zhan Z, Li D, Xu L, Ma F, Zhang P, Yao H, Cao X. Intracellular MHC class II molecules promote TLR-triggered innate immune responses by maintaining activation of the kinase Btk. Nat Immunol. 2011;12:416–424. doi: 10.1038/ni.2015.
    1. Gray P, Dunne A, Brikos C, Jefferies CA, Doyle SL, O'Neill LA. MyD88 adapter-like (mal) is phosphorylated by Bruton's tyrosine kinase during TLR2 and TLR4 signal transduction. J Biol Chem. 2006;281:10489–10495. doi: 10.1074/jbc.M508892200.
    1. Bournazos S, Wang TT, Ravetch JV. The role and function of Fcgamma receptors on myeloid cells. Microbiol Spectr. 2016;4
    1. Fearon ER, Winkelstein JA, Civin CI, Pardoll DM, Vogelstein B. Carrier detection in X-linked agammaglobulinemia by analysis of X-chromosome inactivation. N Engl J Med. 1987;316:427–431. doi: 10.1056/NEJM198702193160802.
    1. Puck JM, Nussbaum RL, Conley ME. Carrier detection in X-linked severe combined immunodeficiency based on patterns of X chromosome inactivation. J Clin Invest. 1987;79:1395–1400. doi: 10.1172/JCI112967.
    1. Melchers F, ten Boekel E, Seidl T, Kong XC, Yamagami T, Onishi K, Shimizu T, Rolink AG, Andersson J. Repertoire selection by pre-B-cell receptors and B-cell receptors, and genetic control of B-cell development from immature to mature B cells. Immunol Rev. 2000;175:33–46. doi: 10.1111/j.1600-065X.2000.imr017510.x.
    1. Hendriks RW, Middendorp S. The pre-BCR checkpoint as a cell-autonomous proliferation switch. Trends Immunol. 2004;25:249–256. doi: 10.1016/j.it.2004.02.011.
    1. Hendriks RW, Bredius RG, Pike-Overzet K, Staal FJ. Biology and novel treatment options for XLA, the most common monogenetic immunodeficiency in man. Expert Opin Ther Targets. 2011;15:1003–1021. doi: 10.1517/14728222.2011.585971.
    1. Herzog S, Reth M, Jumaa H. Regulation of B-cell proliferation and differentiation by pre-B-cell receptor signalling. Nat Rev Immunol. 2009;9:195–205. doi: 10.1038/nri2491.
    1. Ohnishi K, Melchers F. The nonimmunoglobulin portion of lambda5 mediates cell-autonomous pre-B cell receptor signaling. Nat Immunol. 2003;4:849–856. doi: 10.1038/ni959.
    1. Ubelhart R, Bach MP, Eschbach C, Wossning T, Reth M, Jumaa H. N-linked glycosylation selectively regulates autonomous precursor BCR function. Nat Immunol. 2010;11:759–765. doi: 10.1038/ni.1903.
    1. ten Boekel E, Yamagami T, Andersson J, Rolink AG, Melchers F. The formation and selection of cells expressing preB cell receptors and B cell receptors. Curr Top Microbiol Immunol. 1999;246:3–9.
    1. Wardemann H, Yurasov S, Schaefer A, Young JW, Meffre E, Nussenzweig MC. Predominant autoantibody production by early human B cell precursors. Science. 2003;301:1374–1377. doi: 10.1126/science.1086907.
    1. Jumaa H, Mitterer M, Reth M, Nielsen PJ. The absence of SLP65 and Btk blocks B cell development at the preB cell receptor-positive stage. Eur J Immunol. 2001;31:2164–2169. doi: 10.1002/1521-4141(200107)31:7<2164::AID-IMMU2164>;2-S.
    1. Jumaa H, Bossaller L, Portugal K, Storch B, Lotz M, Flemming A, Schrappe M, Postila V, Riikonen P, Pelkonen J, et al. Deficiency of the adaptor SLP-65 in pre-B-cell acute lymphoblastic leukaemia. Nature. 2003;423:452–456. doi: 10.1038/nature01608.
    1. Middendorp S, Zijlstra AJ, Kersseboom R, Dingjan GM, Jumaa H, Hendriks RW. Tumor suppressor function of Bruton tyrosine kinase is independent of its catalytic activity. Blood. 2005;105:259–265. doi: 10.1182/blood-2004-07-2708.
    1. Kersseboom R, Middendorp S, Dingjan GM, Dahlenborg K, Reth M, Jumaa H, Hendriks RW. Bruton's tyrosine kinase cooperates with the B cell linker protein SLP-65 as a tumor suppressor in pre-B cells. J Exp Med. 2003;198:91–98. doi: 10.1084/jem.20030615.
    1. Thompson EC, Cobb BS, Sabbattini P, Meixlsperger S, Parelho V, Liberg D, Taylor B, Dillon N, Georgopoulos K, Jumaa H, et al. Ikaros DNA-binding proteins as integral components of B cell developmental-stage-specific regulatory circuits. Immunity. 2007;26:335–344. doi: 10.1016/j.immuni.2007.02.010.
    1. Stadhouders R, de Bruijn MJ, Rother MB, Yuvaraj S, Ribeiro de Almeida C, Kolovos P, Van Zelm MC, van Ijcken W, Grosveld F, Soler E, Hendriks RW. Pre-B cell receptor signaling induces immunoglobulin kappa locus accessibility by functional redistribution of enhancer-mediated chromatin interactions. PLoS Biol. 2014;12:e1001791. doi: 10.1371/journal.pbio.1001791.
    1. Dingjan GM, Middendorp S, Dahlenborg K, Maas A, Grosveld F, Hendriks RW. Bruton's tyrosine kinase regulates the activation of gene rearrangements at the lambda light chain locus in precursor B cells in the mouse. J Exp Med. 2001;193:1169–1178. doi: 10.1084/jem.193.10.1169.
    1. Gay D, Saunders T, Camper S, Weigert M. Receptor editing: an approach by autoreactive B cells to escape tolerance. J Exp Med. 1993;177:999–1008. doi: 10.1084/jem.177.4.999.
    1. Nemazee D, Weigert M. Revising B cell receptors. J Exp Med. 2000;191:1813–1817. doi: 10.1084/jem.191.11.1813.
    1. Rolink A, Grawunder U, Haasner D, Strasser A, Melchers F. Immature surface Ig+ B cells can continue to rearrange kappa and lambda L chain gene loci. J Exp Med. 1993;178:1263–1270. doi: 10.1084/jem.178.4.1263.
    1. Petro JB, Gerstein RM, Lowe J, Carter RS, Shinners N, Khan WN. Transitional type 1 and 2 B lymphocyte subsets are differentially responsive to antigen receptor signaling. J Biol Chem. 2002;277:48009–48019. doi: 10.1074/jbc.M200305200.
    1. Levine MH, Haberman AM, Sant'Angelo DB, Hannum LG, Cancro MP, Janeway CA, Jr, Shlomchik MJ. A B-cell receptor-specific selection step governs immature to mature B cell differentiation. Proc Natl Acad Sci U S A. 2000;97:2743–2748. doi: 10.1073/pnas.050552997.
    1. Hammad H, Vanderkerken M, Pouliot P, Deswarte K, Toussaint W, Vergote K, Vandersarren L, Janssens S, Ramou I, Savvides SN, et al. Transitional B cells commit to marginal zone B cell fate by Taok3-mediated surface expression of ADAM10. Nat Immunol. 2017;18:313–320. doi: 10.1038/ni.3657.
    1. Gibb DR, El Shikh M, Kang DJ, Rowe WJ, El Sayed R, Cichy J, Yagita H, Tew JG, Dempsey PJ, Crawford HC, Conrad DH. ADAM10 is essential for Notch2-dependent marginal zone B cell development and CD23 cleavage in vivo. J Exp Med. 2010;207:623–635. doi: 10.1084/jem.20091990.
    1. Martin F, Kearney JF. Positive selection from newly formed to marginal zone B cells depends on the rate of clonal production, CD19, and btk. Immunity. 2000;12:39–49. doi: 10.1016/S1074-7613(00)80157-0.
    1. Corneth OB, Klein Wolterink RG, Hendriks RW. BTK signaling in B cell differentiation and autoimmunity. Curr Top Microbiol Immunol. 2016;393:67–105.
    1. Victora GD. SnapShot: the germinal center reaction. Cell. 2014;159:700–0. e701
    1. Oropallo MA, Cerutti A. Germinal center reaction: antigen affinity and presentation explain it all. Trends Immunol. 2014;35:287–289. doi: 10.1016/j.it.2014.06.001.
    1. Dingjan GM, Maas A, Nawijn MC, Smit L, Voerman JS, Grosveld F, Hendriks RW, Severe B. Cell deficiency and disrupted splenic architecture in transgenic mice expressing the E41K mutated form of Bruton's tyrosine kinase. EMBO J. 1998;17:5309–5320. doi: 10.1093/emboj/17.18.5309.
    1. Kersseboom R, Kil L, Flierman R, van der Zee M, Dingjan GM, Middendorp S, Maas A, Hendriks RW. Constitutive activation of Bruton's tyrosine kinase induces the formation of autoreactive IgM plasma cells. Eur J Immunol. 2010;40:2643–2654. doi: 10.1002/eji.201040521.
    1. Kil LP, de Bruijn MJ, van Nimwegen M, Corneth OB, van Hamburg JP, Dingjan GM, Thaiss F, Rimmelzwaan GF, Elewaut D, Delsing D, et al. Btk levels set the threshold for B-cell activation and negative selection of autoreactive B cells in mice. Blood. 2012;119:3744–3756. doi: 10.1182/blood-2011-12-397919.
    1. Corneth OB, de Bruijn MJ, Rip J, Asmawidjaja PS, Kil LP, Hendriks RW. Enhanced expression of Bruton's tyrosine kinase in B cells drives systemic autoimmunity by disrupting T cell homeostasis. J Immunol. 2016;197:58–67. doi: 10.4049/jimmunol.1600208.
    1. Ammann EM, Shanafelt TD, Wright KB, McDowell BD, Link BK, Chrischilles EA. Updating survival estimates in patients with chronic lymphocytic leukemia or small lymphocytic lymphoma (CLL/SLL) based on treatment-free interval length. Leuk Lymphoma. 2017:1–7.
    1. Kil LP, Yuvaraj S, Langerak AW, Hendriks RW. The role of B cell receptor stimulation in CLL pathogenesis. Curr Pharm Des. 2012;18:3335–3355. doi: 10.2174/138161212801227041.
    1. Zenz T, Eichhorst B, Busch R, Denzel T, Habe S, Winkler D, Buhler A, Edelmann J, Bergmann M, Hopfinger G, et al. TP53 mutation and survival in chronic lymphocytic leukemia. J Clin Oncol. 2010;28:4473–4479. doi: 10.1200/JCO.2009.27.8762.
    1. Gonzalez D, Martinez P, Wade R, Hockley S, Oscier D, Matutes E, Dearden CE, Richards SM, Catovsky D, Morgan GJ. Mutational status of the TP53 gene as a predictor of response and survival in patients with chronic lymphocytic leukemia: results from the LRF CLL4 trial. J Clin Oncol. 2011;29:2223–2229. doi: 10.1200/JCO.2010.32.0838.
    1. Robak P, Robak T. Novel synthetic drugs currently in clinical development for chronic lymphocytic leukemia. Expert Opin Investig Drugs. 2017;26:1249–1265. doi: 10.1080/13543784.2017.1384814.
    1. Malcikova J, Smardova J, Rocnova L, Tichy B, Kuglik P, Vranova V, Cejkova S, Svitakova M, Skuhrova Francova H, Brychtova Y, et al. Monoallelic and biallelic inactivation of TP53 gene in chronic lymphocytic leukemia: selection, impact on survival, and response to DNA damage. Blood. 2009;114:5307–5314. doi: 10.1182/blood-2009-07-234708.
    1. Zenz T, Krober A, Scherer K, Habe S, Buhler A, Benner A, Denzel T, Winkler D, Edelmann J, Schwanen C, et al. Monoallelic TP53 inactivation is associated with poor prognosis in chronic lymphocytic leukemia: results from a detailed genetic characterization with long-term follow-up. Blood. 2008;112:3322–3329. doi: 10.1182/blood-2008-04-154070.
    1. Chiorazzi N, Ferrarini M. Cellular origin(s) of chronic lymphocytic leukemia: cautionary notes and additional considerations and possibilities. Blood. 2011;117:1781–1791. doi: 10.1182/blood-2010-07-155663.
    1. Klein U, Tu Y, Stolovitzky GA, Mattioli M, Cattoretti G, Husson H, Freedman A, Inghirami G, Cro L, Baldini L, et al. Gene expression profiling of B cell chronic lymphocytic leukemia reveals a homogeneous phenotype related to memory B cells. J Exp Med. 2001;194:1625–1638. doi: 10.1084/jem.194.11.1625.
    1. Seifert M, Sellmann L, Bloehdorn J, Wein F, Stilgenbauer S, Durig J, Kuppers R. Cellular origin and pathophysiology of chronic lymphocytic leukemia. J Exp Med. 2012;209:2183–2198. doi: 10.1084/jem.20120833.
    1. Griffin DO, Holodick NE, Rothstein TL. Human B1 cells in umbilical cord and adult peripheral blood express the novel phenotype CD20+ CD27+ CD43+ CD70. J Exp Med. 2011;208:67–80. doi: 10.1084/jem.20101499.
    1. DiLillo DJ, Weinberg JB, Yoshizaki A, Horikawa M, Bryant JM, Iwata Y, Matsushita T, Matta KM, Chen Y, Venturi GM, et al. Chronic lymphocytic leukemia and regulatory B cells share IL-10 competence and immunosuppressive function. Leukemia. 2013;27:170–182. doi: 10.1038/leu.2012.165.
    1. Muggen AF, Singh SP, Hendriks RW, Langerak AW. Targeting signaling pathways in chronic lymphocytic leukemia. Curr Cancer Drug Targets. 2016;16:669–688. doi: 10.2174/1568009616666160408145623.
    1. Damle RN, Wasil T, Fais F, Ghiotto F, Valetto A, Allen SL, Buchbinder A, Budman D, Dittmar K, Kolitz J, et al. Ig V gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia. Blood. 1999;94:1840–1847.
    1. Agathangelidis A, Darzentas N, Hadzidimitriou A, Brochet X, Murray F, Yan XJ, Davis Z, van Gastel-Mol EJ, Tresoldi C, Chu CC, et al. Stereotyped B-cell receptors in one-third of chronic lymphocytic leukemia: a molecular classification with implications for targeted therapies. Blood. 2012;119:4467–4475. doi: 10.1182/blood-2011-11-393694.
    1. Murray F, Darzentas N, Hadzidimitriou A, Tobin G, Boudjogra M, Scielzo C, Laoutaris N, Karlsson K, Baran-Marzsak F, Tsaftaris A, et al. Stereotyped patterns of somatic hypermutation in subsets of patients with chronic lymphocytic leukemia: implications for the role of antigen selection in leukemogenesis. Blood. 2008;111:1524–1533. doi: 10.1182/blood-2007-07-099564.
    1. Hayakawa K, Formica AM, Colombo MJ, Shinton SA, Brill-Dashoff J, Morse Iii HC, Li YS, Hardy RR. Loss of a chromosomal region with synteny to human 13q14 occurs in mouse chronic lymphocytic leukemia that originates from early-generated B-1 B cells. Leukemia. 2016;30:1510–1519. doi: 10.1038/leu.2016.61.
    1. Chen SS, Batliwalla F, Holodick NE, Yan XJ, Yancopoulos S, Croce CM, Rothstein TL, Chiorazzi N. Autoantigen can promote progression to a more aggressive TCL1 leukemia by selecting variants with enhanced B-cell receptor signaling. Proc Natl Acad Sci U S A. 2013;110:E1500–E1507. doi: 10.1073/pnas.1300616110.
    1. Singh SP, Pillai SY, de Bruijn MJW, Stadhouders R, Corneth OBJ, van den Ham HJ, Muggen A, van IW SE, Kuil A, et al. Cell lines generated from a chronic lymphocytic leukemia mouse model exhibit constitutive Btk and Akt signaling. Oncotarget. 2017;8:71981–71995.
    1. Messmer BT, Albesiano E, Efremov DG, Ghiotto F, Allen SL, Kolitz J, Foa R, Damle RN, Fais F, Messmer D, et al. Multiple distinct sets of stereotyped antigen receptors indicate a role for antigen in promoting chronic lymphocytic leukemia. J Exp Med. 2004;200:519–525. doi: 10.1084/jem.20040544.
    1. Herve M, Xu K, Ng YS, Wardemann H, Albesiano E, Messmer BT, Chiorazzi N, Meffre E. Unmutated and mutated chronic lymphocytic leukemias derive from self-reactive B cell precursors despite expressing different antibody reactivity. J Clin Invest. 2005;115:1636–1643. doi: 10.1172/JCI24387.
    1. Lanemo Myhrinder A, Hellqvist E, Sidorova E, Soderberg A, Baxendale H, Dahle C, Willander K, Tobin G, Backman E, Soderberg O, et al. A new perspective: molecular motifs on oxidized LDL, apoptotic cells, and bacteria are targets for chronic lymphocytic leukemia antibodies. Blood. 2008;111:3838–3848. doi: 10.1182/blood-2007-11-125450.
    1. Hoogeboom R, van Kessel KP, Hochstenbach F, Wormhoudt TA, Reinten RJ, Wagner K, Kater AP, Guikema JE, Bende RJ, van Noesel CJ. A mutated B cell chronic lymphocytic leukemia subset that recognizes and responds to fungi. J Exp Med. 2013;210:59–70. doi: 10.1084/jem.20121801.
    1. Jimenez de Oya N, De Giovanni M, Fioravanti J, Ubelhart R, Di Lucia P, Fiocchi A, Iacovelli S, Efremov DG, Caligaris-Cappio F, Jumaa H, et al. Pathogen-specific B-cell receptors drive chronic lymphocytic leukemia by light-chain-dependent cross-reaction with autoantigens. EMBO Mol Med. 2017;9:1482–1490. doi: 10.15252/emmm.201707732.
    1. Duhren-von Minden M, Ubelhart R, Schneider D, Wossning T, Bach MP, Buchner M, Hofmann D, Surova E, Follo M, Kohler F, et al. Chronic lymphocytic leukaemia is driven by antigen-independent cell-autonomous signalling. Nature. 2012;489:309–312. doi: 10.1038/nature11309.
    1. Minici C, Gounari M, Ubelhart R, Scarfo L, Duhren-von Minden M, Schneider D, Tasdogan A, Alkhatib A, Agathangelidis A, Ntoufa S, et al. Distinct homotypic B-cell receptor interactions shape the outcome of chronic lymphocytic leukaemia. Nat Commun. 2017;8:15746. doi: 10.1038/ncomms15746.
    1. Herman SE, Gordon AL, Hertlein E, Ramanunni A, Zhang X, Jaglowski S, Flynn J, Jones J, Blum KA, Buggy JJ, et al. Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765. Blood. 2011;117:6287–6296. doi: 10.1182/blood-2011-01-328484.
    1. Ponader S, Chen SS, Buggy JJ, Balakrishnan K, Gandhi V, Wierda WG, Keating MJ, O'Brien S, Chiorazzi N, Burger JA. The Bruton tyrosine kinase inhibitor PCI-32765 thwarts chronic lymphocytic leukemia cell survival and tissue homing in vitro and in vivo. Blood. 2012;119:1182–1189. doi: 10.1182/blood-2011-10-386417.
    1. Kil LP, de Bruijn MJ, van Hulst JA, Langerak AW, Yuvaraj S, Hendriks RW. Bruton's tyrosine kinase mediated signaling enhances leukemogenesis in a mouse model for chronic lymphocytic leukemia. Am J Blood Res. 2013;3:71–83.
    1. Ramsay AD, Rodriguez-Justo M. Chronic lymphocytic leukaemia--the role of the microenvironment pathogenesis and therapy. Br J Haematol. 2013;162:15–24. doi: 10.1111/bjh.12344.
    1. Burger JA, Gribben JG. The microenvironment in chronic lymphocytic leukemia (CLL) and other B cell malignancies: insight into disease biology and new targeted therapies. Semin Cancer Biol. 2014;24:71–81. doi: 10.1016/j.semcancer.2013.08.011.
    1. de Rooij MF, Kuil A, Geest CR, Eldering E, Chang BY, Buggy JJ, Pals ST, Spaargaren M. The clinically active BTK inhibitor PCI-32765 targets B-cell receptor- and chemokine-controlled adhesion and migration in chronic lymphocytic leukemia. Blood. 2012;119:2590–2594. doi: 10.1182/blood-2011-11-390989.
    1. Cheah CY, Seymour JF, Wang ML. Mantle cell lymphoma. J Clin Oncol. 2016;34:1256–1269. doi: 10.1200/JCO.2015.63.5904.
    1. Bertoni F, Rinaldi A, Zucca E, Cavalli F. Update on the molecular biology of mantle cell lymphoma. Hematol Oncol. 2006;24:22–27. doi: 10.1002/hon.767.
    1. Meggendorfer M, Kern W, Haferlach C, Haferlach T, Schnittger S. SOX11 overexpression is a specific marker for mantle cell lymphoma and correlates with t(11;14) translocation, CCND1 expression and an adverse prognosis. Leukemia. 2013;27:2388–2391. doi: 10.1038/leu.2013.141.
    1. Navarro A, Clot G, Royo C, Jares P, Hadzidimitriou A, Agathangelidis A, Bikos V, Darzentas N, Papadaki T, Salaverria I, et al. Molecular subsets of mantle cell lymphoma defined by the IGHV mutational status and SOX11 expression have distinct biologic and clinical features. Cancer Res. 2012;72:5307–5316. doi: 10.1158/0008-5472.CAN-12-1615.
    1. Cinar M, Hamedani F, Mo Z, Cinar B, Amin HM, Alkan S. Bruton tyrosine kinase is commonly overexpressed in mantle cell lymphoma and its attenuation by Ibrutinib induces apoptosis. Leuk Res. 2013;37:1271–1277. doi: 10.1016/j.leukres.2013.07.028.
    1. Pighi C, Gu TL, Dalai I, Barbi S, Parolini C, Bertolaso A, Pedron S, Parisi A, Ren J, Cecconi D, et al. Phospho-proteomic analysis of mantle cell lymphoma cells suggests a pro-survival role of B-cell receptor signaling. Cell Oncol (Dordr) 2011;34:141–153. doi: 10.1007/s13402-011-0019-7.
    1. Boyd RS, Jukes-Jones R, Walewska R, Brown D, Dyer MJ, Cain K. Protein profiling of plasma membranes defines aberrant signaling pathways in mantle cell lymphoma. Mol Cell Proteomics. 2009;8:1501–1515. doi: 10.1074/mcp.M800515-MCP200.
    1. Chang BY, Francesco M, De Rooij MF, Magadala P, Steggerda SM, Huang MM, Kuil A, Herman SE, Chang S, Pals ST, et al. Egress of CD19(+)CD5(+) cells into peripheral blood following treatment with the Bruton tyrosine kinase inhibitor ibrutinib in mantle cell lymphoma patients. Blood. 2013;122:2412–2424. doi: 10.1182/blood-2013-02-482125.
    1. Treon SP, Xu L, Yang G, Zhou Y, Liu X, Cao Y, Sheehy P, Manning RJ, Patterson CJ, Tripsas C, et al. MYD88 L265P somatic mutation in Waldenstrom's macroglobulinemia. N Engl J Med. 2012;367:826–833. doi: 10.1056/NEJMoa1200710.
    1. Pasqualucci L, Trifonov V, Fabbri G, Ma J, Rossi D, Chiarenza A, Wells VA, Grunn A, Messina M, Elliot O, et al. Analysis of the coding genome of diffuse large B-cell lymphoma. Nat Genet. 2011;43:830–837. doi: 10.1038/ng.892.
    1. Puente XS, Pinyol M, Quesada V, Conde L, Ordonez GR, Villamor N, Escaramis G, Jares P, Bea S, Gonzalez-Diaz M, et al. Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature. 2011;475:101–105. doi: 10.1038/nature10113.
    1. Ngo VN, Young RM, Schmitz R, Jhavar S, Xiao W, Lim KH, Kohlhammer H, Xu W, Yang Y, Zhao H, et al. Oncogenically active MYD88 mutations in human lymphoma. Nature. 2011;470:115–119. doi: 10.1038/nature09671.
    1. Montesinos-Rongen M, Godlewska E, Brunn A, Wiestler OD, Siebert R, Deckert M. Activating L265P mutations of the MYD88 gene are common in primary central nervous system lymphoma. Acta Neuropathol. 2011;122:791–792. doi: 10.1007/s00401-011-0891-2.
    1. Yang G, Zhou Y, Liu X, Xu L, Cao Y, Manning RJ, Patterson CJ, Buhrlage SJ, Gray N, Tai YT, et al. A mutation in MYD88 (L265P) supports the survival of lymphoplasmacytic cells by activation of Bruton tyrosine kinase in Waldenstrom macroglobulinemia. Blood. 2013;122:1222–1232. doi: 10.1182/blood-2012-12-475111.
    1. Hunter ZR, Xu L, Yang G, Zhou Y, Liu X, Cao Y, Manning RJ, Tripsas C, Patterson CJ, Sheehy P, Treon SP. The genomic landscape of Waldenstrom macroglobulinemia is characterized by highly recurring MYD88 and WHIM-like CXCR4 mutations, and small somatic deletions associated with B-cell lymphomagenesis. Blood. 2014;123:1637–1646. doi: 10.1182/blood-2013-09-525808.
    1. Ngo HT, Leleu X, Lee J, Jia X, Melhem M, Runnels J, Moreau AS, Burwick N, Azab AK, Roccaro A, et al. SDF-1/CXCR4 and VLA-4 interaction regulates homing in Waldenstrom macroglobulinemia. Blood. 2008;112:150–158. doi: 10.1182/blood-2007-12-129395.
    1. Iqbal J, Shen Y, Huang X, Liu Y, Wake L, Liu C, Deffenbacher K, Lachel CM, Wang C, Rohr J, et al. Global microRNA expression profiling uncovers molecular markers for classification and prognosis in aggressive B-cell lymphoma. Blood. 2015;125:1137–1145. doi: 10.1182/blood-2014-04-566778.
    1. Dunleavy K, Wilson WH. Primary mediastinal B-cell lymphoma and mediastinal gray zone lymphoma: do they require a unique therapeutic approach? Blood. 2015;125:33–39. doi: 10.1182/blood-2014-05-575092.
    1. Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, Rosenwald A, Boldrick JC, 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. doi: 10.1038/35000501.
    1. Lenz G, Nagel I, Siebert R, Roschke AV, Sanger W, Wright GW, Dave SS, Tan B, Zhao H, Rosenwald A, et al. Aberrant immunoglobulin class switch recombination and switch translocations in activated B cell-like diffuse large B cell lymphoma. J Exp Med. 2007;204:633–643. doi: 10.1084/jem.20062041.
    1. Roschewski M, Staudt LM, Wilson WH. Diffuse large B-cell lymphoma-treatment approaches in the molecular era. Nat Rev Clin Oncol. 2014;11:12–23. doi: 10.1038/nrclinonc.2013.197.
    1. Davis RE, Brown KD, Siebenlist U, Staudt LM. Constitutive nuclear factor kappaB activity is required for survival of activated B cell-like diffuse large B cell lymphoma cells. J Exp Med. 2001;194:1861–1874. doi: 10.1084/jem.194.12.1861.
    1. Ngo VN, Davis RE, Lamy L, Yu X, Zhao H, Lenz G, Lam LT, Dave S, Yang L, Powell J, Staudt LM. A loss-of-function RNA interference screen for molecular targets in cancer. Nature. 2006;441:106–110. doi: 10.1038/nature04687.
    1. Compagno M, Lim WK, Grunn A, Nandula SV, Brahmachary M, Shen Q, Bertoni F, Ponzoni M, Scandurra M, Califano A, et al. Mutations of multiple genes cause deregulation of NF-kappaB in diffuse large B-cell lymphoma. Nature. 2009;459:717–721. doi: 10.1038/nature07968.
    1. Lenz G, Davis RE, Ngo VN, Lam L, George TC, Wright GW, Dave SS, Zhao H, Xu W, Rosenwald A, et al. Oncogenic CARD11 mutations in human diffuse large B cell lymphoma. Science. 2008;319:1676–1679. doi: 10.1126/science.1153629.
    1. Davis RE, Ngo VN, Lenz G, Tolar P, Young RM, Romesser PB, 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. doi: 10.1038/nature08638.
    1. Chen L, Monti S, Juszczynski P, Ouyang J, Chapuy B, Neuberg D, Doench JG, Bogusz AM, Habermann TM, Dogan A, et al. SYK inhibition modulates distinct PI3K/AKT- dependent survival pathways and cholesterol biosynthesis in diffuse large B cell lymphomas. Cancer Cell. 2013;23:826–838. doi: 10.1016/j.ccr.2013.05.002.
    1. Havranek O, Xu J, Kohrer S, Wang Z, Becker L, Comer JM, Henderson J, Ma W, Man Chun Ma J, Westin JR, et al. Tonic B-cell receptor signaling in diffuse large B-cell lymphoma. Blood. 2017;130:995–1006. doi: 10.1182/blood-2016-10-747303.
    1. Pfeifer M, Grau M, Lenze D, Wenzel SS, Wolf A, Wollert-Wulf B, Dietze K, Nogai H, Storek B, Madle H, et al. PTEN loss defines a PI3K/AKT pathway-dependent germinal center subtype of diffuse large B-cell lymphoma. Proc Natl Acad Sci U S A. 2013;110:12420–12425. doi: 10.1073/pnas.1305656110.
    1. Lenz G, Wright GW, Emre NC, Kohlhammer H, Dave SS, Davis RE, Carty S, Lam LT, Shaffer AL, Xiao W, et al. Molecular subtypes of diffuse large B-cell lymphoma arise by distinct genetic pathways. Proc Natl Acad Sci U S A. 2008;105:13520–13525. doi: 10.1073/pnas.0804295105.
    1. Grommes C, Pastore A, Palaskas N, Tang SS, Campos C, Schartz D, Codega P, Nichol D, Clark O, Hsieh WY, et al. Ibrutinib unmasks critical role of Bruton tyrosine kinase in primary CNS lymphoma. Cancer Discov. 2017;7:1018–1029. doi: 10.1158/-17-0613.
    1. Krysiak K, Gomez F, White BS, Matlock M, Miller CA, Trani L, Fronick CC, Fulton RS, Kreisel F, Cashen AF, et al. Recurrent somatic mutations affecting B-cell receptor signaling pathway genes in follicular lymphoma. Blood. 2017;129:473–483. doi: 10.1182/blood-2016-07-729954.
    1. Yang Y, Shi J, Gu Z, Salama ME, Das S, Wendlandt E, Xu H, Huang J, Tao Y, Hao M, et al. Bruton tyrosine kinase is a therapeutic target in stem-like cells from multiple myeloma. Cancer Res. 2015;75:594–604. doi: 10.1158/0008-5472.CAN-14-2362.
    1. Gu C, Peng H, Lu Y, Yang H, Tian Z, Yin G, Zhang W, Lu S, Zhang Y, Yang Y. BTK suppresses myeloma cellular senescence through activating AKT/P27/Rb signaling. Oncotarget. 2017;8:56858–56867.
    1. Tai YT, Chang BY, Kong SY, Fulciniti M, Yang G, Calle Y, Hu Y, Lin J, Zhao JJ, Cagnetta A, et al. Bruton tyrosine kinase inhibition is a novel therapeutic strategy targeting tumor in the bone marrow microenvironment in multiple myeloma. Blood. 2012;120:1877–1887. doi: 10.1182/blood-2011-12-396853.
    1. Yan Q, Huang Y, Watkins AJ, Kocialkowski S, Zeng N, Hamoudi RA, Isaacson PG, de Leval L, Wotherspoon A, Du MQ. BCR and TLR signaling pathways are recurrently targeted by genetic changes in splenic marginal zone lymphomas. Haematologica. 2012;97:595–598. doi: 10.3324/haematol.2011.054080.
    1. Clipson A, Wang M, de Leval L, Ashton-Key M, Wotherspoon A, Vassiliou G, Bolli N, Grove C, Moody S, Escudero-Ibarz L, et al. KLF2 mutation is the most frequent somatic change in splenic marginal zone lymphoma and identifies a subset with distinct genotype. Leukemia. 2015;29:1177–1185. doi: 10.1038/leu.2014.330.
    1. Pan Z, Scheerens H, Li SJ, Schultz BE, Sprengeler PA, Burrill LC, Mendonca RV, Sweeney MD, Scott KC, Grothaus PG, et al. Discovery of selective irreversible inhibitors for Bruton's tyrosine kinase. ChemMedChem. 2007;2:58–61. doi: 10.1002/cmdc.200600221.
    1. Honigberg LA, Smith AM, Sirisawad M, Verner E, Loury D, Chang B, Li S, Pan Z, Thamm DH, Miller RA, Buggy JJ. The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy. Proc Natl Acad Sci U S A. 2010;107:13075–13080. doi: 10.1073/pnas.1004594107.
    1. Advani RH, Buggy JJ, Sharman JP, Smith SM, Boyd TE, Grant B, Kolibaba KS, Furman RR, Rodriguez S, Chang BY, et al. Bruton tyrosine kinase inhibitor ibrutinib (PCI-32765) has significant activity in patients with relapsed/refractory B-cell malignancies. J Clin Oncol. 2013;31:88–94. doi: 10.1200/JCO.2012.42.7906.
    1. Wang ML, Rule S, Martin P, Goy A, Auer R, Kahl BS, Jurczak W, Advani RH, Romaguera JE, Williams ME, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2013;369:507–516. doi: 10.1056/NEJMoa1306220.
    1. Treon SP, Tripsas CK, Meid K, Warren D, Varma G, Green R, Argyropoulos KV, Yang G, Cao Y, Xu L, et al. Ibrutinib in previously treated Waldenstrom's macroglobulinemia. N Engl J Med. 2015;372:1430–1440. doi: 10.1056/NEJMoa1501548.
    1. Noy A, de Vos S, Thieblemont C, Martin P, Flowers CR, Morschhauser F, Collins GP, 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. doi: 10.1182/blood-2016-10-747345.
    1. Burger JA, Wiestner A. Targeting B cell receptor signalling in cancer: preclinical and clinical advances. Nat Rev Cancer. 2018;
    1. Mason JA, Hagel KR, Hawk MA, Schafer ZT. Metabolism during ECM detachment: Achilles heel of cancer cells? Trends Cancer. 2017;3:475–481. doi: 10.1016/j.trecan.2017.04.009.
    1. Pals ST, Kersten MJ, Spaargaren M. Targeting cell adhesion and homing as strategy to cure Waldenstrom's macroglobulinemia. Best Pract Res Clin Haematol. 2016;29:161–168. doi: 10.1016/j.beha.2016.08.019.
    1. Woyach JA, Furman RR, Liu TM, Ozer HG, Zapatka M, Ruppert AS, Xue L, Li DH, Steggerda SM, Versele M, et al. Resistance mechanisms for the Bruton's tyrosine kinase inhibitor ibrutinib. N Engl J Med. 2014;370:2286–2294. doi: 10.1056/NEJMoa1400029.
    1. Furman RR, Cheng S, Lu P, Setty M, Perez AR, Guo A, Racchumi J, Xu G, Wu H, Ma J, et al. Ibrutinib resistance in chronic lymphocytic leukemia. N Engl J Med. 2014;370:2352–2354. doi: 10.1056/NEJMc1402716.
    1. Kadri S, Lee J, Fitzpatrick C, Galanina N, Sukhanova M, Venkataraman G, Sharma S, Long B, Petras K, Theissen M, et al. Clonal evolution underlying leukemia progression and Richter transformation in patients with ibrutinib-relapsed CLL. Blood Adv. 2017;1:715–727. doi: 10.1182/bloodadvances.2016003632.
    1. Wilson WH, Young RM, Schmitz R, Yang Y, Pittaluga S, Wright G, Lih CJ, Williams PM, Shaffer AL, Gerecitano J, et al. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med. 2015;21:922–926. doi: 10.1038/nm.3884.
    1. Wu C, de Miranda NF, Chen L, Wasik AM, Mansouri L, Jurczak W, Galazka K, Dlugosz-Danecka M, Machaczka M, Zhang H, et al. Genetic heterogeneity in primary and relapsed mantle cell lymphomas: impact of recurrent CARD11 mutations. Oncotarget. 2016;7:38180–38190.
    1. Maddocks KJ, Ruppert AS, Lozanski G, Heerema NA, Zhao W, Abruzzo L, Lozanski A, Davis M, Gordon A, Smith LL, et al. Etiology of Ibrutinib therapy discontinuation and outcomes in patients with chronic lymphocytic leukemia. JAMA Oncol. 2015;1:80–87. doi: 10.1001/jamaoncol.2014.218.
    1. Coutre SE, Furman RR, Flinn IW, Burger JA, Blum K, Sharman J, Jones J, Wierda W, Zhao W, Heerema NA, et al. Extended treatment with single-agent Ibrutinib at the 420 mg dose leads to durable responses in chronic lymphocytic leukemia/small lymphocytic lymphoma. Clin Cancer Res. 2017;23:1149–1155. doi: 10.1158/1078-0432.CCR-16-1431.
    1. Brown JR, Hillmen P, O'Brien S, Barrientos JC, Reddy NM, Coutre SE, Tam CS, Mulligan SP, Jaeger U, Barr PM, et al. Extended follow-up and impact of high-risk prognostic factors from the phase 3 RESONATE study in patients with previously treated CLL/SLL. Leukemia. 2017;
    1. Mulligan SP, Ward CM, Whalley D, Hilmer SN. Atrial fibrillation, anticoagulant stroke prophylaxis and bleeding risk with ibrutinib therapy for chronic lymphocytic leukaemia and lymphoproliferative disorders. Br J Haematol. 2016;175:359–364. doi: 10.1111/bjh.14321.
    1. Jones JA, Hillmen P, Coutre S, Tam C, Furman RR, Barr PM, Schuster SJ, Kipps TJ, Flinn IW, Jaeger U, et al. Use of anticoagulants and antiplatelet in patients with chronic lymphocytic leukaemia treated with single-agent ibrutinib. Br J Haematol. 2017;178:286–291. doi: 10.1111/bjh.14660.
    1. Wiczer TE, Levine LB, Brumbaugh J, Coggins J, Zhao Q, Ruppert AS, Rogers K, McCoy A, Mousa L, Guha A, et al. Cumulative incidence, risk factors, and management of atrial fibrillation in patients receiving ibrutinib. Blood Adv. 2017;1:1739–1748.
    1. McMullen JR, Boey EJ, Ooi JY, Seymour JF, Keating MJ, Tam CS. Ibrutinib increases the risk of atrial fibrillation, potentially through inhibition of cardiac PI3K-Akt signaling. Blood. 2014;124:3829–3830. doi: 10.1182/blood-2014-10-604272.
    1. Byrd JC, Furman RR, Coutre SE, Burger JA, Blum KA, Coleman M, Wierda WG, Jones JA, Zhao W, Heerema NA, et al. Three-year follow-up of treatment-naïve and previously treated patients with CLL and SLL receiving single-agent ibrutinib. Blood. 2015;125:2497–2506. doi: 10.1182/blood-2014-10-606038.
    1. O'Brien SM, Furman RR, Coutre SE, Flinn IW, Burger J, Blum K, Sharman J, Wierda WG, Jones J, Zhao W, et al. Five-year experience with single-agent ibrutinib in patients with previously untreated and relapsed/refractory chronic lymphocytic leukemia/small lymphocytic leukemia. Blood. 2016;128
    1. Herman SEM, Montraveta A, Niemann CU, Mora-Jensen H, Gulrajani M, Krantz F, Mantel R, Smith LL, McClanahan F, Harrington BK, et al. The Bruton tyrosine kinase (BTK) inhibitor Acalabrutinib demonstrates potent on-target effects and efficacy in two mouse models of chronic lymphocytic leukemia. Clin Cancer Res. 2017;23:2831–2841. doi: 10.1158/1078-0432.CCR-16-0463.
    1. Quek LS, Bolen J, Watson SP. A role for Bruton's tyrosine kinase (Btk) in platelet activation by collagen. Curr Biol. 1998;8:1137–1140. doi: 10.1016/S0960-9822(98)70471-3.
    1. Futatani T, Watanabe C, Baba Y, Tsukada S, Ochs HD. Bruton's tyrosine kinase is present in normal platelets and its absence identifies patients with X-linked agammaglobulinaemia and carrier females. Br J Haematol. 2001;114:141–149. doi: 10.1046/j.1365-2141.2001.02905.x.
    1. Bye AP, Unsworth AJ, Desborough MJ, Hildyard CAT, Appleby N, Bruce D, Kriek N, Nock SH, Sage T, Hughes CE, Gibbins JM. Severe platelet dysfunction in NHL patients receiving ibrutinib is absent in patients receiving acalabrutinib. Blood Adv. 2017;1:2610–2623.
    1. Tillman BF, Pauff JM, Satyanarayana G, Talbott M, Warner JL. Systematic review of infectious events with the BTK inhibitor ibrutinib in the treatment of haematologic malignancies. Eur J Haematol. 2017;
    1. Long M, Beckwith K, Do P, Mundy BL, Gordon A, Lehman AM, Maddocks KJ, Cheney C, Jones JA, Flynn JM, et al. Ibrutinib treatment improves T cell number and function in CLL patients. J Clin Invest. 2017;127:3052–3064. doi: 10.1172/JCI89756.
    1. Schutt SD, Fu J, Nguyen H, Bastian D, Heinrichs J, Wu Y, Liu C, DG MD, Pidala J, Yu XZ. Inhibition of BTK and ITK with Ibrutinib is effective in the prevention of chronic graft-versus-host disease in mice. PLoS One. 2015;10:e0137641. doi: 10.1371/journal.pone.0137641.
    1. Dubovsky JA, Flynn R, Du J, Harrington BK, Zhong Y, Kaffenberger B, Yang C, Towns WH, Lehman A, Johnson AJ, et al. Ibrutinib treatment ameliorates murine chronic graft-versus-host disease. J Clin Invest. 2014;124:4867–4876. doi: 10.1172/JCI75328.
    1. Ryan CE, Sahaf B, Logan AC, O'Brien S, Byrd JC, Hillmen P, Brown JR, Dyer MJ, Mato AR, Keating MJ, et al. Ibrutinib efficacy and tolerability in patients with relapsed chronic lymphocytic leukemia following allogeneic HCT. Blood. 2016;128:2899–2908. doi: 10.1182/blood-2016-06-715284.
    1. Kohrt HE, Sagiv-Barfi I, Rafiq S, Herman SE, Butchar JP, Cheney C, Zhang X, Buggy JJ, Muthusamy N, Levy R, et al. Ibrutinib antagonizes rituximab-dependent NK cell-mediated cytotoxicity. Blood. 2014;123:1957–1960. doi: 10.1182/blood-2014-01-547869.
    1. Wu J, Liu C, Tsui ST, Liu D. Second-generation inhibitors of Bruton tyrosine kinase. J Hematol Oncol. 2016;9:80. doi: 10.1186/s13045-016-0313-y.
    1. Harrington BK, Gardner HL, Izumi R, Hamdy A, Rothbaum W, Coombes KR, Covey T, Kaptein A, Gulrajani M, Van Lith B, et al. Preclinical evaluation of the novel BTK inhibitor Acalabrutinib in canine models of B-cell non-Hodgkin lymphoma. PLoS One. 2016;11:e0159607. doi: 10.1371/journal.pone.0159607.
    1. Wang M, Rule S, Zinzani PL, Goy A, Casasnovas O, Smith SD, Damaj G, Doorduijn J, Lamy T, Morschhauser F, et al. Acalabrutinib in relapsed or refractory mantle cell lymphoma (ACE-LY-004): a single-arm, multicentre, phase 2 trial. Lancet. 2017;
    1. Acalabrutinib approved for MCL. Cancer Discov. 2018;8:OF6.
    1. Thompson PA, Burger JA. Bruton's tyrosine kinase inhibitors: first and second generation agents for patients with chronic lymphocytic leukemia (CLL) Expert Opin Investig Drugs. 2018;27:31–42. doi: 10.1080/13543784.2018.1404027.
    1. Walter HS, Rule SA, Dyer MJ, Karlin L, Jones C, Cazin B, Quittet P, Shah N, Hutchinson CV, Honda H, et al. A phase 1 clinical trial of the selective BTK inhibitor ONO/GS-4059 in relapsed and refractory mature B-cell malignancies. Blood. 2016;127:411–419. doi: 10.1182/blood-2015-08-664086.
    1. Koprulu AD, Ellmeier W. The role of Tec family kinases in mononuclear phagocytes. Crit Rev Immunol. 2009;29:317–333. doi: 10.1615/CritRevImmunol.v29.i4.30.
    1. Mirsafian H, Ripen AM, Leong WM, Chear CT, Bin Mohamad S, Merican AF. Transcriptome profiling of monocytes from XLA patients revealed the innate immune function dysregulation due to the BTK gene expression deficiency. Sci Rep. 2017;7:6836. doi: 10.1038/s41598-017-06342-5.
    1. Doyle SL, Jefferies CA, Feighery C, O'Neill LA. Signaling by toll-like receptors 8 and 9 requires Bruton's tyrosine kinase. J Biol Chem. 2007;282:36953–36960. doi: 10.1074/jbc.M707682200.
    1. Sochorova K, Horvath R, Rozkova D, Litzman J, Bartunkova J, Sediva A, Spisek R. Impaired toll-like receptor 8-mediated IL-6 and TNF-alpha production in antigen-presenting cells from patients with X-linked agammaglobulinemia. Blood. 2007;109:2553–2556. doi: 10.1182/blood-2006-07-037960.
    1. Lougaris V, Baronio M, Vitali M, Tampella G, Cattalini M, Tassone L, Soresina A, Badolato R, Plebani A. Bruton tyrosine kinase mediates TLR9-dependent human dendritic cell activation. J Allergy Clin Immunol. 2014;133:1644–1650. doi: 10.1016/j.jaci.2013.12.1085.
    1. Marron TU, Martinez-Gallo M, Yu JE, Cunningham-Rundles C. Toll-like receptor 4-, 7-, and 8-activated myeloid cells from patients with X-linked agammaglobulinemia produce enhanced inflammatory cytokines. J Allergy Clin Immunol. 2012;129:184–190. doi: 10.1016/j.jaci.2011.10.009.
    1. Taneichi H, Kanegane H, Sira MM, Futatani T, Agematsu K, Sako M, Kaneko H, Kondo N, Kaisho T, Miyawaki T. Toll-like receptor signaling is impaired in dendritic cells from patients with X-linked agammaglobulinemia. Clin Immunol. 2008;126:148–154. doi: 10.1016/j.clim.2007.10.005.
    1. Ni Gabhann J, Hams E, Smith S, Wynne C, Byrne JC, Brennan K, Spence S, Kissenpfennig A, Johnston JA, Fallon PG, Jefferies CA. Btk regulates macrophage polarization in response to lipopolysaccharide. PLoS One. 2014;9:e85834. doi: 10.1371/journal.pone.0085834.
    1. Gunderson AJ, Kaneda MM, Tsujikawa T, Nguyen AV, Affara NI, Ruffell B, Gorjestani S, Liudahl SM, Truitt M, Olson P, et al. Bruton tyrosine kinase-dependent immune cell cross-talk drives pancreas cancer. Cancer Discov. 2016;6:270–285. doi: 10.1158/-15-0827.
    1. Fiorcari S, Maffei R, Audrito V, Martinelli S, Ten Hacken E, Zucchini P, Grisendi G, Potenza L, Luppi M, Burger JA, et al. Ibrutinib modifies the function of monocyte/macrophage population in chronic lymphocytic leukemia. Oncotarget. 2016;7:65968–65981. doi: 10.18632/oncotarget.11782.
    1. Andreu P, Johansson M, Affara NI, Pucci F, Tan T, Junankar S, Korets L, Lam J, Tawfik D, DeNardo DG, et al. FcRgamma activation regulates inflammation-associated squamous carcinogenesis. Cancer Cell. 2010;17:121–134. doi: 10.1016/j.ccr.2009.12.019.
    1. Pucci F, Garris C, Lai CP, Newton A, Pfirschke C, Engblom C, Alvarez D, Sprachman M, Evavold C, Magnuson A, et al. SCS macrophages suppress melanoma by restricting tumor-derived vesicle-B cell interactions. Science. 2016;352:242–246. doi: 10.1126/science.aaf1328.
    1. Coffelt SB, Wellenstein MD, de Visser KE. Neutrophils in cancer: neutral no more. Nat Rev Cancer. 2016;16:431–446. doi: 10.1038/nrc.2016.52.
    1. Marron TU, Rohr K, Martinez-Gallo M, Yu J, Cunningham-Rundles C. TLR signaling and effector functions are intact in XLA neutrophils. Clin Immunol. 2010;137:74–80. doi: 10.1016/j.clim.2010.06.011.
    1. Farrar JE, Rohrer J, Conley ME. Neutropenia in X-linked agammaglobulinemia. Clin Immunol Immunopathol. 1996;81:271–276. doi: 10.1006/clin.1996.0188.
    1. Fiedler K, Sindrilaru A, Terszowski G, Kokai E, Feyerabend TB, Bullinger L, Rodewald HR, Brunner C. Neutrophil development and function critically depend on Bruton tyrosine kinase in a mouse model of X-linked agammaglobulinemia. Blood. 2011;117:1329–1339. doi: 10.1182/blood-2010-04-281170.
    1. Mangla A, Khare A, Vineeth V, Panday NN, Mukhopadhyay A, Ravindran B, Bal V, George A, Rath S. Pleiotropic consequences of Bruton tyrosine kinase deficiency in myeloid lineages lead to poor inflammatory responses. Blood. 2004;104:1191–1197. doi: 10.1182/blood-2004-01-0207.
    1. Mueller H, Stadtmann A, Van Aken H, Hirsch E, Wang D, Ley K, Zarbock A. Tyrosine kinase Btk regulates E-selectin-mediated integrin activation and neutrophil recruitment by controlling phospholipase C (PLC) gamma2 and PI3Kgamma pathways. Blood. 2010;115:3118–3127. doi: 10.1182/blood-2009-11-254185.
    1. Volmering S, Block H, Boras M, Lowell CA, Zarbock A. The neutrophil Btk Signalosome regulates integrin activation during sterile inflammation. Immunity. 2016;44:73–87. doi: 10.1016/j.immuni.2015.11.011.
    1. Stiff A, Trikha P, Wesolowski R, Kendra K, Hsu V, Uppati S, McMichael E, Duggan M, Campbell A, Keller K, et al. Myeloid-derived suppressor cells express Bruton's tyrosine kinase and can be depleted in tumor-bearing hosts by Ibrutinib treatment. Cancer Res. 2016;76:2125–2136. doi: 10.1158/0008-5472.CAN-15-1490.
    1. Sagiv-Barfi I, Kohrt HE, Czerwinski DK, Ng PP, Chang BY, Levy R. Therapeutic antitumor immunity by checkpoint blockade is enhanced by ibrutinib, an inhibitor of both BTK and ITK. Proc Natl Acad Sci U S A. 2015;112:E966–E972. doi: 10.1073/pnas.1500712112.
    1. Gao W, Wang M, Wang L, Lu H, Wu S, Dai B, Ou Z, Zhang L, Heymach JV, Gold KA, et al. Selective antitumor activity of ibrutinib in EGFR-mutant non-small cell lung cancer cells. J Natl Cancer Inst. 2014;106
    1. Ruella M, Kenderian SS, Shestova O, Fraietta JA, Qayyum S, Zhang Q, Maus MV, Liu X, Nunez-Cruz S, Klichinsky M, et al. The addition of the BTK inhibitor Ibrutinib to anti-CD19 chimeric antigen receptor T cells (CART19) improves responses against mantle cell lymphoma. Clin Cancer Res. 2016;22:2684–2696. doi: 10.1158/1078-0432.CCR-15-1527.
    1. Fraietta JA, Beckwith KA, Patel PR, Ruella M, Zheng Z, Barrett DM, Lacey SF, Melenhorst JJ, McGettigan SE, Cook DR, et al. Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood. 2016;127:1117–1127. doi: 10.1182/blood-2015-11-679134.
    1. Dubovsky JA, Beckwith KA, Natarajan G, Woyach JA, Jaglowski S, Zhong Y, Hessler JD, Liu TM, Chang BY, Larkin KM, et al. Ibrutinib is an irreversible molecular inhibitor of ITK driving a Th1-selective pressure in T lymphocytes. Blood. 2013;122:2539–2549. doi: 10.1182/blood-2013-06-507947.
    1. Byrd JC, Brown JR, O'Brien S, Barrientos JC, Kay NE, Reddy NM, Coutre S, Tam CS, Mulligan SP, Jaeger U, et al. Ibrutinib versus ofatumumab in previously treated chronic lymphoid leukemia. N Engl J Med. 2014;371:213–223. doi: 10.1056/NEJMoa1400376.
    1. Dreyling M, Jurczak W, Jerkeman M, Silva RS, Rusconi C, Trneny M, Offner F, Caballero D, Joao C, Witzens-Harig M, et al. Ibrutinib versus temsirolimus in patients with relapsed or refractory mantle-cell lymphoma: an international, randomised, open-label, phase 3 study. Lancet. 2016;387:770–778. doi: 10.1016/S0140-6736(15)00667-4.
    1. Burger JA, Keating MJ, Wierda WG, Hartmann E, Hoellenriegel J, Rosin NY, de Weerdt I, Jeyakumar G, Ferrajoli A, Cardenas-Turanzas M, et al. Safety and activity of ibrutinib plus rituximab for patients with high-risk chronic lymphocytic leukaemia: a single-arm, phase 2 study. Lancet Oncol. 2014;15:1090–1099. doi: 10.1016/S1470-2045(14)70335-3.
    1. Chanan-Khan A, Cramer P, Demirkan F, Fraser G, Silva RS, Grosicki S, Pristupa A, Janssens A, Mayer J, Bartlett NL, et al. Ibrutinib combined with bendamustine and rituximab compared with placebo, bendamustine, and rituximab for previously treated chronic lymphocytic leukaemia or small lymphocytic lymphoma (HELIOS): a randomised, double-blind, phase 3 study. Lancet Oncol. 2016;17:200–211. doi: 10.1016/S1470-2045(15)00465-9.
    1. Wang ML, Lee H, Chuang H, Wagner-Bartak N, Hagemeister F, Westin J, Fayad L, Samaniego F, Turturro F, Oki Y, et al. Ibrutinib in combination with rituximab in relapsed or refractory mantle cell lymphoma: a single-centre, open-label, phase 2 trial. Lancet Oncol. 2016;17:48–56. doi: 10.1016/S1470-2045(15)00438-6.
    1. Tam C, Grigg AP, Opat S, Ku M, Gilbertson M, Anderson MA, Seymour JF, Ritchie DS, Dicorleto C, Dimovski B, et al. The BTK inhibitor, Bgb-3111, is safe, tolerable, and highly active in patients with relapsed/refractory B-cell malignancies: initial report of a phase 1 first-in-human trial. Blood. 2015;126
    1. Noy A, de Vos S, Thieblemont C, Martin P, Flowers C, Morschhauser F, Collins GP, Ma S, Coleman M, Peles S, et al. Single-agent Ibrutinib demonstrates efficacy and safety in patients with relapsed/refractory marginal zone lymphoma: a multicenter, open-label, phase 2 study. Blood. 2016;128:1213.
    1. Secchiero P, Voltan R, Rimondi E, Melloni E, Athanasakis E, Tisato V, Gallo S, Rigolin GM, Zauli G. The gamma-secretase inhibitors enhance the anti-leukemic activity of ibrutinib in B-CLL cells. Oncotarget. 2017;8:59235–59245. doi: 10.18632/oncotarget.19494.
    1. Rotin LE, Gronda M, MacLean N, Hurren R, Wang X, Lin FH, Wrana J, Datti A, Barber DL, Minden MD, et al. Ibrutinib synergizes with poly(ADP-ribose) glycohydrolase inhibitors to induce cell death in AML cells via a BTK-independent mechanism. Oncotarget. 2016;7:2765–2779. doi: 10.18632/oncotarget.6409.
    1. Kelly PN, Romero DL, Yang Y, Shaffer AL, 3rd, Chaudhary D, Robinson S, Miao W, Rui L, Westlin WF, Kapeller R, Staudt LM. Selective interleukin-1 receptor-associated kinase 4 inhibitors for the treatment of autoimmune disorders and lymphoid malignancy. J Exp Med. 2015;212:2189–2201. doi: 10.1084/jem.20151074.
    1. Goldstein RL, Yang SN, Taldone T, Chang B, Gerecitano J, Elenitoba-Johnson K, Shaknovich R, Tam W, Leonard JP, Chiosis G, et al. Pharmacoproteomics identifies combinatorial therapy targets for diffuse large B cell lymphoma. J Clin Invest. 2015;125:4559–4571. doi: 10.1172/JCI80714.
    1. Patel V, Keating MJ, Wierda WG, Gandhi V. Preclinical combination of TP-0903, an AXL inhibitor and B-PAC-1, a procaspase-activating compound with ibrutinib in chronic lymphocytic leukemia. Leuk Lymphoma. 2016;57:1494–1497. doi: 10.3109/10428194.2015.1102243.
    1. Lamothe B, Cervantes-Gomez F, Sivina M, Wierda WG, Keating MJ, Gandhi V. Proteasome inhibitor carfilzomib complements ibrutinib's action in chronic lymphocytic leukemia. Blood. 2015;125:407–410. doi: 10.1182/blood-2014-07-585364.
    1. Hing ZA, Mantel R, Beckwith KA, Guinn D, Williams E, Smith LL, Williams K, Johnson AJ, Lehman AM, Byrd JC, et al. Selinexor is effective in acquired resistance to ibrutinib and synergizes with ibrutinib in chronic lymphocytic leukemia. Blood. 2015;125:3128–3132. doi: 10.1182/blood-2015-01-621391.
    1. Cervantes-Gomez F, Lamothe B, Woyach JA, Wierda WG, Keating MJ, Balakrishnan K, Gandhi V. Pharmacological and protein profiling suggests Venetoclax (ABT-199) as optimal partner with Ibrutinib in chronic lymphocytic leukemia. Clin Cancer Res. 2015;21:3705–3715. doi: 10.1158/1078-0432.CCR-14-2809.
    1. Zhao X, Bodo J, Sun D, Durkin L, Lin J, Smith MR, Hsi ED. Combination of ibrutinib with ABT-199: synergistic effects on proliferation inhibition and apoptosis in mantle cell lymphoma cells through perturbation of BTK, AKT and BCL2 pathways. Br J Haematol. 2015;168:765–768. doi: 10.1111/bjh.13149.
    1. Rushworth SA, Bowles KM, Barrera LN, Murray MY, Zaitseva L, MacEwan DJ. BTK inhibitor ibrutinib is cytotoxic to myeloma and potently enhances bortezomib and lenalidomide activities through NF-kappaB. Cell Signal. 2013;25:106–112. doi: 10.1016/j.cellsig.2012.09.008.
    1. Sagiv-Barfi I, Kohrt HE, Burckhardt L, Czerwinski DK, Levy R. Ibrutinib enhances the antitumor immune response induced by intratumoral injection of a TLR9 ligand in mouse lymphoma. Blood. 2015;125:2079–2086. doi: 10.1182/blood-2014-08-593137.
    1. Xargay-Torrent S, Lopez-Guerra M, Rosich L, Montraveta A, Roldan J, Rodriguez V, Villamor N, Aymerich M, Lagisetti C, Webb TR, et al. The splicing modulator sudemycin induces a specific antitumor response and cooperates with ibrutinib in chronic lymphocytic leukemia. Oncotarget. 2015;6:22734–22749. doi: 10.18632/oncotarget.4212.
    1. Yahiaoui A, Meadows SA, Sorensen RA, Cui ZH, Keegan KS, Brockett R, Chen G, Queva C, Li L, Tannheimer SL. PI3Kdelta inhibitor idelalisib in combination with BTK inhibitor ONO/GS-4059 in diffuse large B cell lymphoma with acquired resistance to PI3Kdelta and BTK inhibitors. PLoS One. 2017;12:e0171221. doi: 10.1371/journal.pone.0171221.

Source: PubMed

3
Subskrybuj