421
Views
19
CrossRef citations to date
0
Altmetric
Review

Emerging protein kinase inhibitors for the treatment of multiple myeloma

, , &
Pages 133-152 | Received 05 May 2019, Accepted 19 Jul 2019, Published online: 29 Jul 2019

References

  • Moreau P, San Miguel J, Sonneveld P, et al. Multiple myeloma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017;28:iv52–iv61.
  • Manning G, Whyte DB, Martinez R, et al. The protein kinase complement of the human genome. Science. 2002;298:1912–1934.
  • Roskoski R. Classification of small molecule protein kinase inhibitors based upon the structures of their drug-enzyme complexes. Pharmacol Res. 2016;103:26–48.
  • Kumar SK, Callander NS, Alsina M, et al. NCCN guidelines insights: multiple myeloma, version 3.2018. J Natl Compr Cancer Netw. 2018;16:11–20.
  • Rask-Andersen M, Masuram S, Schiöth HB. The druggable genome: evaluation of drug targets in clinical trials suggests major shifts in molecular class and indication. Annu Rev Pharmacol Toxicol. 2013;54:9–26.
  • Raje N, Berdeja J, Lin Y, et al. Anti-BCMA CAR T-Cell Therapy bb2121 in relapsed or refractory multiple myeloma. N Engl J Med. 2019;380:1726–1737.
  • Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell. 2010;141:1117–1134.
  • Podar K, Anderson KC. Caveolin-1 as a potential new therapeutic target in multiple myeloma. Cancer Lett. 2006;233:10–15.
  • Smadja NV, Bastard C, Brigaudeau C, et al. Hypodiploidy is a major prognostic factor in multiple myeloma. Blood. 2001;98:2229–2238.
  • Chell V, Balmanno K, Little AS, et al. Tumour cell responses to new fibroblast growth factor receptor tyrosine kinase inhibitors and identification of a gatekeeper mutation in FGFR3 as a mechanism of acquired resistance. Oncogene. 2013;32:3059–3070.
  • Hideshima T, Mitsiades C, Tonon G, et al. Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets. Nat Rev Cancer. 2007;7:585–598.
  • Keats JJ, Reiman T, Maxwell CA, et al. In multiple myeloma, t(4;14)(p16;q32) is an adverse prognostic factor irrespective of FGFR3 expression. Blood. 2003;101:1520–1529.
  • Wildes TM, Procknow E, Gao F, et al. Dasatinib in relapsed or plateau-phase multiple myeloma. Leuk Lymphoma. 2009;50:137–140.
  • Facon T, Leleu X, Stewart AK, et al. Dasatinib in combination with lenalidomide and dexamethasone in patients with relapsed or refractory multiple myeloma: preliminary results of a phase I study. Blood. 2009;114:1876.
  • Trudel S, Bergsagel PL, Singhal S, et al. A phase I study of the safety and pharmacokinetics of escalating doses of MFGR1877S, a Fibroblast Growth Factor Receptor 3 (FGFR3) antibody, in patients with relapsed or refractory t(4;14)-positive multiple myeloma. Blood. 2012;120:4029.
  • Smith DC, Britten C, Garon EB, et al. A phase I study of XL228, a multitargeted protein kinase inhibitor, in patients (pts) with solid tumors or multiple myeloma. J Clin Oncol. 2017;28:3105.
  • Dispenzieri A, Gertz MA, Lacy MQ, et al. A phase II trial of imatinib in patients with refractory/relapsed myeloma. Leuk Lymphoma. 2006;47:39–42.
  • Lendvai N, Yee AJ, Tsakos I, et al. Phase IB study of cabozantinib in patients with relapsed and/or refractory multiple myeloma. Blood. 2016;127:2355–2356.
  • Richardson P, Lonial S, Jakubowiak A, et al. Multi-center phase II study of perifosine (KRX-0401) alone and in combination with dexamethasone (dex) for patients with relapsed or relapsed/refractory multiple myeloma (MM): promising activity as combination therapy with manageable toxicity. Blood. 2007;110:1164.
  • Richardson PG, Wolf J, Jakubowiak A, et al. Perifosine plus bortezomib and dexamethasone in patients with relapsed/refractory multiple myeloma previously treated with bortezomib: results of a multicenter phase I/II trial. J Clin Oncol. 2011;29:4243–4249.
  • Spencer A, Yoon -S-S, Harrison SJ, et al. Novel AKT Inhibitor GSK2110183 shows favorable safety, pharmacokinetics, and clinical activity in multiple myeloma. Preliminary results from a phase I first-time-in-human study. Blood. 2011;118:1856.
  • Yee AJ, Mahindra AK, Richardson PG, et al. Biomarker correlation with outcomes in patients with relapsed or refractory multiple Myeloma on a phase I study of everolimus in combination with lenalidomide. Blood. 2011;118:3966.
  • Hofmeister CC, Yang X, Pichiorri F, et al. Phase I trial of lenalidomide and CCI-779 in patients with relapsed multiple Myeloma: evidence for Lenalidomide–CCI-779 Interaction via P-Glycoprotein. J Clin Oncol. 2011;29:3427–3434.
  • Ghobrial IM, Siegel D, Vij R, et al. TAK-228 (formerly MLN0128), an investigational oral dual TORC1/2 inhibitor: A phase I dose escalation study in patients with relapsed or refractory multiple myeloma, non-Hodgkin lymphoma, or Waldenström’s macroglobulinemia. Am J Hematol. 2016 Jun;91(4):400–405.
  • Chari A, Larson S, Holkova B, et al. Phase 1 trial of ibrutinib and carfilzomib combination therapy for relapsed or relapsed and refractory multiple myeloma. Leuk Lymphoma. 2018;59:2588–2594.
  • Holkova B, Perkins EB, Ramakrishnan V, et al. Phase I trial of bortezomib (PS-341; NSC 681239) and alvocidib (flavopiridol; NSC 649890) in patients with recurrent or refractory B-cell neoplasms. Clin Cancer Res. 2011;17:3388–3397.
  • Hofmeister CC, Poi M, Bowers MA, et al. A phase I trial of flavopiridol in relapsed multiple myeloma. Cancer Chemother Pharmacol. 2014;73:249–257.
  • Tong W-G, Chen R, Plunkett W, et al. Phase I and pharmacologic study of SNS-032, a potent and selective Cdk2, 7, and 9 inhibitor, in patients with advanced chronic lymphocytic leukemia and multiple myeloma. J Clin Oncol. 2010;28:3015–3022.
  • Hofmeister CC, Berdeja JG, Vesole DH, et al. TG02, an Oral CDK9-Inhibitor, in Combination with Carfilzomib Demonstrated Objective Responses in Carfilzomib Refractory Multiple Myeloma Patients. Blood. 2015;126:3052.
  • Roschewski M, Farooqui M, Aue G, et al. Phase I study of ON 01910.Na (Rigosertib), a multikinase PI3K inhibitor in relapsed/refractory B-cell malignancies. Leukemia. 2013;27:1920–1923.
  • Rosenthal A, Kumar S, Hofmeister C, et al. A Phase Ib Study of the combination of the Aurora Kinase Inhibitor Alisertib (MLN8237) and Bortezomib in Relapsed Multiple Myeloma. Br J Haematol. 2016;174:323–325.
  • Podar K, Anderson KC. The pathophysiologic role of VEGF in hematologic malignancies: therapeutic implications. Blood. 2005;105:1383–1395.
  • Kumar S, Gertz MA, Dispenzieri A, et al. Prognostic value of bone marrow angiogenesis in patients with multiple myeloma undergoing high-dose therapy. Bone Marrow Transplant. 2004;34:235–239.
  • Ghobrial IM, Siegel DS, Vij R, et al. TAK-228 (formerly MLN0128), an investigational oral dual TORC1/2 inhibitor: A phase I dose escalation study in patients with relapsed or refractory multiple myeloma, non-Hodgkin lymphoma, or Waldenström’s macroglobulinemia. Am J Hematol. 2016;91:400–405.
  • Zangari M, Anaissie E, Stopeck A, et al. Phase II study of SU5416, a small molecule vascular endothelial growth factor tyrosine kinase receptor inhibitor, in patients with refractory multiple myeloma. Clin Cancer Res. 2004;10:88–95.
  • Kropff M, Kienast J, Bisping G, et al. An open-label dose-escalation study of BIBF 1120 in patients with relapsed or refractory multiple myeloma. Anticancer Res. 2009;29:4233–4238.
  • Vij R, Ansstas G, Mosley JC, et al. Efficacy and tolerability of PTK787/ZK 222584 in a phase II study of post-transplant maintenance therapy in patients with multiple myeloma following high-dose chemotherapy and autologous stem cell transplant. Leuk Lymphoma. 2010;51:1577–1579.
  • Scheid C, Reece D, Beksac M, et al. Phase 2 study of dovitinib in patients with relapsed or refractory multiple myeloma with or without t(4;14) translocation. Eur J Haematol. 2015;95:316–324.
  • Prince HM, Hönemann D, Spencer A, et al. Vascular endothelial growth factor inhibition is not an effective therapeutic strategy for relapsed or refractory multiple myeloma: A phase 2 study of pazopanib (GW786034). Blood. 2009;113:4819–4820.
  • Kovacs MJ, Reece DE, Marcellus D, et al. A phase II study of ZD6474 (Zactima, a selective inhibitor of VEGFR and EGFR tyrosine kinase in patients with relapsed multiple myeloma–NCIC CTG IND.145. Invest New Drugs. 2006;24:529–535.
  • Kumar S, Flynn PJ, Chng WJJ, et al. A Phase II Trial of Sunitinib (SU11248) in Multiple Myeloma. Blood. 2009;114:4954.
  • Srkalovic G, Hussein MA, Hoering A, et al. A phase II trial of BAY 43-9006 (sorafenib) (NSC-724772) in patients with relapsing and resistant multiple myeloma: SWOG S0434. Cancer Med. 2014;3:1275–1283.
  • Yordanova A, Hose D, Neben K, et al. Sorafenib in patients with refractory or recurrent multiple myeloma. Hematol Oncol. 2013;31:197–200.
  • Kumar SK, Jett J, Marks R, et al. Phase 1 study of sorafenib in combination with bortezomib in patients with advanced malignancies. Invest New Drugs. 2013;31:1201–1206.
  • Gambella M, Palumbo A, Rocci A. MET/HGF pathway in multiple myeloma: from diagnosis to targeted therapy? Expert Rev Mol Diagn. 2015;15:881–893.
  • Mahtouk K, Tjin EPM, Spaargaren M, et al. The HGF/MET pathway as target for the treatment of multiple myeloma and B-cell lymphomas. Biochim Biophys Acta. 2010;1806:208–219.
  • Lath DL, Buckle CH, Evans HR, et al. ARQ-197, a small-molecule inhibitor of c-Met, reduces tumour burden and prevents myeloma-induced bone disease in vivo. PLoS One. 2018;13:e0199517.
  • Baljevic M, Zaman S, Baladandayuthapani V, et al. Phase II study of the c-MET inhibitor tivantinib (ARQ 197) in patients with relapsed or relapsed/refractory multiple myeloma. Ann Hematol. 2017;96:977–985.
  • Sprynski AC, Hose D, Caillot L, et al. The role of IGF-1 as a major growth factor for myeloma cell lines and the prognostic relevance of the expression of its receptor. Blood. 2009;113:4614–4626.
  • Sprynski AC, Hose D, Kassambara A, et al. Insulin is a potent myeloma cell growth factor through insulin/IGF-1 hybrid receptor activation. Leukemia. 2010;24:1940–1950.
  • Lacy MQ, Alsina M, Fonseca R, et al. Phase I, pharmacokinetic and pharmacodynamic study of the anti-insulinlike growth factor type 1 receptor monoclonal antibody CP-751,871 in patients with multiple myeloma. J Clin Oncol. 2008;26:3196–3203.
  • Ullrich A, Gray A, Tam AW, et al. Insulin-like growth factor I receptor primary structure: comparison with insulin receptor suggests structural determinants that define functional specificity. Embo J. 1986;5:2503–2512.
  • Clemmons DR. Involvement of insulin-like growth factor-I in the control of glucose homeostasis. Curr Opin Pharmacol. 2006;6:620–625.
  • Massagué J. TGF-beta signal transduction. Annu Rev Biochem. 1998;67:753–791.
  • Podar K, Raje N, Anderson KC. Inhibition of the TGF-beta signaling pathway in tumor cells. Recent Results Cancer Res. 2007;172:77–97.
  • Urashima M, Ogata A, Chauhan D, et al. Transforming growth factor-beta1: differential effects on multiple myeloma versus normal B cells. Blood. 1996;87:1928–1938.
  • Brown RD, Pope B, Murray A, et al. Dendritic cells from patients with myeloma are numerically normal but functionally defective as they fail to up-regulate CD80 (B7-1) expression after huCD40LT stimulation because of inhibition by transforming growth factor-beta1 and interleukin-10. Blood. 2001;98:2992–2998.
  • Otsuki T, Yamada O, Kurebayashi J, et al. Expression and In VitroModification of Parathyroid Hormone-Related Protein (PTHrP) and PTH/PTHrP-Receptor in Human Myeloma Cells. Leuk Lymphoma. 2001;41:397–409.
  • Takeuchi K, Abe M, Hiasa M, et al. Tgf-Beta inhibition restores terminal osteoblast differentiation to suppress myeloma growth. PLoS One. 2010;5:e9870.
  • de Weers M, Mensink RG, Kraakman ME, et al. Mutation analysis of the Bruton’s tyrosine kinase gene in X-linked agammaglobulinemia: identification of a mutation which affects the same codon as is altered in immunodeficient xid mice. Hum Mol Genet. 1994;3:161–166.
  • Liu Y, Dong Y, Jiang Q-L, et al. Bruton’s tyrosine kinase: potential target in human multiple myeloma. Leuk Lymphoma. 2014;55:177–181.
  • Tai Y-T, Anderson KC. Bruton’s tyrosine kinase: oncotarget in myeloma. Oncotarget. 2012;3:913–914.
  • Richardson PG, Schlossman RL, Roy AN, et al. Patient-reported outcomes of multiple myeloma patients treated with panobinostat after ≥2 lines of therapy based on the international phase 3, randomized, double-blind, placebo-controlled PANORAMA-1 trial. Br J Haematol. 2018;181:628–636.
  • Eda H, Santo L, Cirstea DD, et al. A novel Bruton’s tyrosine kinase inhibitor CC-292 in combination with the proteasome inhibitor carfilzomib impacts the bone microenvironment in a multiple myeloma model with resultant antimyeloma activity. Leukemia. 2014;28:1892–1901.
  • Taga T, Hibi M, Hirata Y, et al. Interleukin-6 triggers the association of its receptor with a possible signal transducer, gp130. Cell. 1989;58:573–581.
  • Zhong Z, Wen Z, Darnell JE. Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science. 1994;264:95–98.
  • Ferrajoli A, Faderl S, Ravandi F, et al. The JAK-STAT pathway: a therapeutic target in hematological malignancies. Curr Cancer Drug Targets. 2006;6:671–679.
  • Podar K, Hideshima T, Chauhan D, et al. Targeting signalling pathways for the treatment of multiple myeloma. Expert Opin Ther Targets. 2005;9:359–381.
  • Catlett-Falcone R, Landowski TH, Oshiro MM, et al. Constitutive activation of Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells. Immunity. 1999;10:105–115.
  • Quintanilla-Martinez L, Kremer M, Specht K, et al. Analysis of signal transducer and activator of transcription 3 (Stat 3) pathway in multiple myeloma: stat 3 activation and cyclin D1 dysregulation are mutually exclusive events. Am J Pathol. 2003;162:1449–1461.
  • Hideshima T, Richardson P, Chauhan D, et al. The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells. Cancer Res. 2001;61:3071–3076.
  • Le Gouill S, Podar K, Harousseau J-L, et al. Mcl-1 regulation and its role in multiple myeloma. Cell Cycle. 2004;3:1259–1262.
  • Swords R, Kelly K, Carew J, et al. The Pim kinases: new targets for drug development. Curr Drug Targets. 2011;12:2059–2066.
  • Lu J, Zavorotinskaya T, Dai Y, et al. Pim2 is required for maintaining multiple myeloma cell growth through modulating TSC2 phosphorylation. Blood. 2013;122:1610–1620.
  • Keane NA, Reidy M, Natoni A, et al. Targeting the Pim kinases in multiple myeloma. Blood Cancer J. 2015;5:e325.
  • Raab MS, Thomas SK, Ocio EM, et al. The first-in-human study of the pan-PIM kinase inhibitor PIM447 in patients with relapsed and/or refractory multiple myeloma. Leukemia. 2019. Epub ahead of print
  • Manni S, Carrino M, Semenzato G, et al. Old and young actors playing novel roles in the drama of multiple myeloma bone marrow microenvironment dependent drug resistance. Int J Mol Sci. 2018;19:E1512.
  • Morgan GJ, Walker BA, Davies FE. The genetic architecture of multiple myeloma. Nat Rev Cancer. 2012;12:335–348.
  • Alsina M, Fonseca R, Wilson EF, et al. Farnesyltransferase inhibitor tipifarnib is well tolerated, induces stabilization of disease, and inhibits farnesylation and oncogenic/tumor survival pathways in patients with advanced multiple myeloma. Blood. 2004;103:3271–3277.
  • Andrulis M, Lehners N, Capper D, et al. Targeting the BRAF V600E Mutation in Multiple Myeloma. Cancer Discov. 2013;3:862–869.
  • Sharman JP, Chmielecki J, Morosini D, et al. Vemurafenib response in 2 patients with posttransplant refractory BRAF V600E-mutated multiple myeloma. Clin Lymphoma Myeloma Leuk. 2014;14:e161–3.
  • Hatzivassiliou G, Song K, Yen I, et al. RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature. 2010;464:431–435.
  • Heuck C, Jethava Y, Khan RZ, et al. Targeted MEK Inhibition in Patients with Previously Treated Multiple Myeloma. Blood. 2014;124:4775.
  • Mey UJM, Renner C, von Moos R. Vemurafenib in combination with cobimetinib in relapsed and refractory extramedullary multiple myeloma harboring the BRAF V600E mutation. Hematol Oncol. 2017;35:890–893.
  • Holkova B, Zingone A, Kmieciak M, et al. A Phase II Trial of AZD6244 (Selumetinib, ARRY-142886), an Oral MEK1/2 Inhibitor, in Relapsed/Refractory Multiple Myeloma. Clin Cancer Res. 2016;22:1067–1075.
  • Hyman DM, Puzanov I, Subbiah V, et al. Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. N Engl J Med. 2015;373:726–736.
  • Keane NA, S V G, Krawczyk J, et al. AKT as a therapeutic target in multiple myeloma. Expert Opin Ther Targets. 2014;18:897–915.
  • Hyun T, Yam A, Pece S, et al. Loss of PTEN expression leading to high Akt activation in human multiple myelomas. Blood. 2000;96:3560–3568.
  • Hideshima T, Nakamura N, Chauhan D, et al. Biologic sequelae of interleukin-6 induced PI3-K/Akt signaling in multiple myeloma. Oncogene. 2001;20:5991–6000.
  • Hirai H, Sootome H, Nakatsuru Y, et al. MK-2206, an allosteric Akt inhibitor, enhances antitumor efficacy by standard chemotherapeutic agents or molecular targeted drugs in vitro and in vivo. Mol Cancer Ther. 2010;9:1956–1967.
  • Ramakrishnan V, Kimlinger T, Haug J, et al. Anti-Myeloma Activity of Akt Inhibition Is Linked to the Activation Status of PI3K/Akt and MEK/ERK Pathway. PLoS One. 2012;7:e50005.
  • Balasis ME, Forinash KD, Chen YA, et al. Combination of Farnesyltransferase and Akt Inhibitors Is Synergistic in Breast Cancer Cells and Causes Significant Breast Tumor Regression in ErbB2 Transgenic Mice. Clin Cancer Res. 2011;17:2852–2862.
  • Jakubowiak AJ, Richardson PG, Zimmerman T, et al. Perifosine plus lenalidomide and dexamethasone in relapsed and relapsed/refractory multiple myeloma: a Phase I Multiple Myeloma Research Consortium study. Br J Haematol. 2012;158:472–480.
  • Richardson PG, Eng C, Kolesar J, et al. Perifosine, an oral, anti-cancer agent and inhibitor of the Akt pathway: mechanistic actions, pharmacodynamics, pharmacokinetics, and clinical activity. Expert Opin Drug Metab Toxicol. 2012;8:623–633.
  • Mimura N, Hideshima T, Shimomura T, et al. Selective and potent Akt inhibition triggers anti-myeloma activities and enhances fatal endoplasmic reticulum stress induced by proteasome inhibition. Cancer Res. 2014;74:4458–4469.
  • Song IS, Jeong YJ, Jeong SH, et al. Pharmacologic inhibition of AKT leads to cell death in relapsed multiple myeloma. Cancer Lett. 2018;432:205–215.
  • Spencer A, Yoon SS, Harrison SJ, et al. The novel AKT inhibitor afuresertib shows favorable safety, pharmacokinetics, and clinical activity in multiple myeloma. Blood. 2014;124:2190–2195.
  • Tolcher AW, Patnaik A, Papadopoulos KP, et al. Phase I study of the MEK inhibitor trametinib in combination with the AKT inhibitor afuresertib in patients with solid tumors and multiple myeloma. Cancer Chemother Pharmacol. 2015;75:183–189.
  • Jacinto E, Loewith R, Schmidt A, et al. Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin insensitive. Nat Cell Biol. 2004;6:1122–1128.
  • Guertin DA, Sabatini DM. Defining the role of mTOR in cancer. Cancer Cell. 2007;12:9–22.
  • Laplante M, Sabatini DM. MTOR signaling in growth control and disease. Cell. 2012;149:274–293.
  • Li J, Zhu J, Cao B, et al. The mTOR signaling pathway is an emerging therapeutic target in multiple myeloma. Curr Pharm Des. 2014;20:125–135.
  • Frost P, Moatamed F, Hoang B, et al. In vivo antitumor effects of the mTOR inhibitor CCI-779 against human multiple myeloma cells in a xenograft model. Blood. 2004;104:4181–4187.
  • Farag SS, Zhang S, Jansak BS, et al. Phase II trial of temsirolimus in patients with relapsed or refractory multiple myeloma. Leuk Res. 2009;33:1475–1480.
  • Günther A, Baumann P, Burger R, et al. Activity of everolimus (RAD001) in relapsed and/or refractory multiple myeloma: a phase I study. Haematologica. 2015;100:541–547.
  • Ghobrial IM, Weller E, Vij R, et al. Weekly bortezomib in combination with temsirolimus in relapsed or relapsed and refractory multiple myeloma: a multicentre, phase 1/2, open-label, dose-escalation study. Lancet Oncol. 2011;12:263–272.
  • Yee AJ, Hari P, Marcheselli R, et al. Outcomes in patients with relapsed or refractory multiple myeloma in a phase I study of everolimus in combination with lenalidomide. Br J Haematol. 2014;166:401–409.
  • Hoang B, Frost P, Shi Y, et al. Targeting TORC2 in multiple myeloma with a new mTOR kinase inhibitor. Blood. 2010;116:4560–4568.
  • Cirstea D, Santo L, Hideshima T, et al. Delineating the mTOR Kinase Pathway Using a Dual TORC1/2 Inhibitor, AZD8055, in Multiple Myeloma. Mol Cancer Ther. 2014;13:2489–2500.
  • Bendell JC, Kelley RK, Shih KC, et al. A phase i dose-escalation study to assess safety, tolerability, pharmacokinetics, and preliminary efficacy of the dual mTORC1/mTORC2 kinase inhibitor CC-223 in patients with advanced solid tumors or multiple myeloma. Cancer. 2015;121:3481–3490.
  • Yang C, Huang X, Liu H, et al. PDK1 inhibitor GSK2334470 exerts antitumor activity in multiple myeloma and forms a novel multitargeted combination with dual mTORC1/C2 inhibitor PP242. Oncotarget. 2017;8:39185–39197.
  • Swords R, Mahalingam D, O’Dwyer M, et al. Cdc7 kinase - a new target for drug development. Eur J Cancer. 2010;46:33–40.
  • Zhan F, Huang Y, Colla S, et al. The molecular classification of multiple myeloma. Blood. 2006;108:2020–2028.
  • Dispenzieri A, Gertz MA, Lacy MQ, et al. Flavopiridol in patients with relapsed or refractory multiple myeloma: A phase 2 trial with clinical and pharmacodynamic end-points. Haematologica. 2006;91:390–393.
  • Niesvizky R, Badros AZ, Costa LJ, et al. Phase 1/2 study of cyclin-dependent kinase (CDK)4/6 inhibitor palbociclib (PD-0332991) with bortezomib and dexamethasone in relapsed/refractory multiple myeloma. Leuk Lymphoma. 2015;56:3320–3328.
  • Kumar SK, LaPlant B, Chng WJ, et al. Dinaciclib, a novel CDK inhibitor, demonstrates encouraging single-agent activity in patients with relapsed multiple myeloma. Blood. 2015;125:443–448.
  • Zhu YX, Tiedemann R, Shi C-X, et al. RNAi screen of the druggable genome identifies modulators of proteasome inhibitor sensitivity in myeloma including CDK5. Blood. 2011;117:3847–3857.
  • Hari PN, Landau H, Richardson PG, et al. A phase I/II open-label multicenter study of the cyclin kinase inhibitor AT7519M alone and in combination with bortezomib in patients with previously treated multiple myeloma. Blood. 2013;122:1976.
  • Raje N, Hideshima T, Mukherjee S, et al. Preclinical activity of P276-00, a novel small-molecule cyclin-dependent kinase inhibitor in the therapy of multiple myeloma. Leukemia. 2009;23:961–970.
  • Alvarez-Fernandez S, Ortiz-Ruiz MJ, Parrott T, et al. Potent antimyeloma activity of a novel ERK5/CDK inhibitor. Clin Cancer Res. 2013;19:2677–2687.
  • Cirstea D, Hideshima T, Santo L, et al. Small-molecule multi-targeted kinase inhibitor RGB-286638 triggers P53-dependent and -independent anti-multiple myeloma activity through inhibition of transcriptional CDKs. Leukemia. 2013;27:2366–2375.
  • McMillin DW, Delmore J, Negri J, et al. Molecular and cellular effects of multi-targeted cyclin-dependent kinase inhibition in myeloma: biological and clinical implications. Br J Haematol. 2011;152:420–432.
  • Natoni A, Coyne MRE, Jacobsen A, et al. Characterization of a dual CDC7/CDK9 inhibitor in multiple myeloma cellular models. Cancers (Basel). 2013;5:901–918.
  • Liu H-Y, Tuckett AZ, Fennell M, et al. Repurposing of the CDK inhibitor PHA-767491 as a NRF2 inhibitor drug candidate for cancer therapy via redox modulation. Invest New Drugs. 2018;36:590–600.
  • Menichincheri M, Bargiotti A, Berthelsen J, et al. First Cdc7 kinase inhibitors: pyrrolopyridinones as potent and orally active antitumor agents. 2. Lead discovery. J Med Chem. 2009;52:293–307.
  • Carmena M, Earnshaw WC. The cellular geography of aurora kinases. Nat Rev Mol Cell Biol. 2003;4:842–854.
  • Brown JR, Koretke KK, Birkeland ML, et al. Evolutionary relationships of Aurora kinases: implications for model organism studies and the development of anti-cancer drugs. BMC Evol Biol. 2004;4:39.
  • Kollareddy M, Dzubak P, Zheleva D, et al. Aurora kinases: structure, functions and their association with cancer. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 2008;152:27–33.
  • Malumbres M. Physiological Relevance of Cell Cycle Kinases. Physiol Rev. 2011;91:973–1007.
  • Craig SN, Wyatt MD, McInnes C. Current assessment of polo-like kinases as anti-tumor drug targets. Expert Opin Drug Discov. 2014;9:773–789.
  • Gautschi O, Heighway J, Mack PC, et al. Aurora kinases as anticancer drug targets. Clin Cancer Res. 2008;14:1639–1648.
  • Farag SS. The potential role of Aurora kinase inhibitors in haematological malignancies. Br J Haematol. 2011;155:561–579.
  • Maes A, Menu E, De Veirman K, et al. The therapeutic potential of cell cycle targeting in multiple myeloma. Oncotarget. 2017;8:90501–90520.
  • Stewart HJ, Kishikova L, Powell FL, et al. The polo-like kinase inhibitor BI 2536 exhibits potent activity against malignant plasma cells and represents a novel therapy in multiple myeloma. Exp Hematol. 2011;39:330–338.
  • Evans RP, Dueck G, Sidhu R, et al. Expression, adverse prognostic significance and therapeutic small molecule inhibition of Polo-like kinase 1 in multiple myeloma. Leuk Res. 2011;35:1637–1643.
  • Zhang G, Zhang Z, Liu Z. Scytonemin inhibits cell proliferation and arrests cell cycle through downregulating Plk1 activity in multiple myeloma cells. Tumor Biol. 2013;34:2241–2247.
  • Hay AE, Murugesan A, DiPasquale AM, et al. A phase II study of AT9283, an aurora kinase inhibitor, in patients with relapsed or refractory multiple myeloma: NCIC clinical trials group IND.191. Leuk Lymphoma. 2016;57:1463–1466.
  • Ganguly R, Hong CS, Smith LGF, et al. Maternal embryonic leucine zipper kinase: key kinase for stem cell phenotype in glioma and other cancers. Mol Cancer Ther. 2014;13:1393–1398.
  • Muller J, Bolomsky A, Dubois S, et al. Maternal embryonic leucine zipper kinase inhibitor OTSSP167 has preclinical activity in multiple myeloma bone disease. Haematologica. 2018;103:1359–1368.
  • Podar K, Raab MS, Chauhan D, et al. The therapeutic role of targeting protein kinase C in solid and hematologic malignancies. Expert Opin Investig Drugs. 2007;16:1693–1707.
  • Podar K, Raab MS, Zhang J, et al. Targeting PKC in multiple myeloma: in vitro and in vivo effects of the novel, orally available small-molecule inhibitor enzastaurin (LY317615.HCl). Blood. 2007;109:1669–1677.
  • Neri A, Marmiroli S, Tassone P, et al. The oral protein-kinase C beta inhibitor enzastaurin (LY317615) suppresses signalling through the AKT pathway, inhibits proliferation and induces apoptosis in multiple myeloma cell lines. Leuk Lymphoma. 2008;49:1374–1383.
  • Jourdan E, Leblond V, Maisonneuve H, et al. A multicenter phase II study of single-agent enzastaurin in previously treated multiple myeloma. Leuk Lymphoma. 2014;55:2013–2017.
  • Ghobrial IM, Munshi NC, Harris BN, et al. A phase I safety study of enzastaurin plus bortezomib in the treatment of relapsed or refractory multiple myeloma. Am J Hematol. 2011;86:573–578.
  • Hideshima T, Akiyama M, Hayashi T, et al. Targeting p38 MAPK inhibits multiple myeloma cell growth in the bone marrow milieu. Blood. 2003;101:703–705.
  • He J, Liu Z, Zheng Y, et al. p38 MAPK in myeloma cells regulates osteoclast and osteoblast activity and induces bone destruction. Cancer Res. 2012;72:6393–6402.
  • Liu H, He J, Yang J. Tumor cell p38 MAPK: A trigger of cancer bone osteolysis. Cancer Cell Microenviron. 2015;2:e464.
  • Ishitsuka K, Hideshima T, Neri P, et al. p38 mitogen-activated protein kinase inhibitor LY2228820 enhances bortezomib-induced cytotoxicity and inhibits osteoclastogenesis in multiple myeloma; therapeutic implications. Br J Haematol. 2008;141:598–606.
  • Vanderkerken K, Medicherla S, Coulton L, et al. Inhibition of p38α mitogen-activated protein kinase prevents the development of osteolytic bone disease, reduces tumor burden, and increases survival in murine models of multiple myeloma. Cancer Res. 2007;67:4572–4577.
  • Mitsiades CS, Ocio EM, Pandiella A, et al. Aplidin, a marine organism-derived compound with potent antimyeloma activity in vitro and in vivo. Cancer Res. 2008;68:5216–5225.
  • Siegel DS, Krishnan A, Lonial S, et al. Phase II trial of SCIO-469 as monotherapy (M) or in combination with bortezomib (MB) in relapsed refractory multiple myeloma (MM). Blood. 2006;108:3580.
  • Mateos MV, Cibeira MT, Richardson PG, et al. Phase II clinical and pharmacokinetic study of plitidepsin 3-hour infusion every two weeks alone or with dexamethasone in relapsed and refractory multiple myeloma. Clin Cancer Res. 2010;16:3260–3269.
  • Baumann P, Mandl-Weber S, Emmerich B, et al. Activation of adenosine monophosphate activated protein kinase inhibits growth of multiple myeloma cells. Exp Cell Res. 2007;313:3592–3603.
  • Baumann P, Mandl-Weber S, Emmerich B, et al. Inhibition of adenosine monophosphate-activated protein kinase induces apoptosis in multiple myeloma cells. Anticancer Drugs. 2007;18:405–410.
  • Zi F-M, He J-S, Li Y, et al. Metformin displays anti-myeloma activity and synergistic effect with dexamethasone in in vitro and in vivo xenograft models. Cancer Lett. 2015;356:443–453.
  • Wang Y, Xu W, Yan Z, et al. Metformin induces autophagy and G0/G1 phase cell cycle arrest in myeloma by targeting the AMPK/mTORC1 and mTORC2 pathways. J Exp Clin Cancer Res. 2018;37:63.
  • Wu W, Merriman K, Nabaah A, et al. The association of diabetes and anti-diabetic medications with clinical outcomes in multiple myeloma. Br J Cancer. 2014;111:628–636.
  • Suzuki A, Lu J, Kusakai G-I, et al. ARK5 is a tumor invasion-associated factor downstream of Akt signaling. Mol Cell Biol. 2004;24:3526–3535.
  • Suzuki A, Iida S, Kato-Uranishi M, et al. ARK5 is transcriptionally regulated by the Large-MAF family and mediates IGF-1-induced cell invasion in multiple myeloma: ARK5 as a new molecular determinant of malignant multiple myeloma. Oncogene. 2005;24:6936–6944.
  • Perumal D, Kuo P-Y, Leshchenko VV, et al. Dual targeting of CDK4 and ARK5 using a novel kinase inhibitor ON123300 exerts potent anticancer activity against multiple myeloma. Cancer Res. 2016;76:1225–1236.
  • David J-P, Mehic D, Bakiri L, et al. Essential role of RSK2 in c-Fos-dependent osteosarcoma development. J Clin Invest. 2005;115:664–672.
  • Kang S, Elf S, Lythgoe K, et al. p90 ribosomal S6 kinase 2 promotes invasion and metastasis of human head and neck squamous cell carcinoma cells. J Clin Invest. 2010;120:1165–1177.
  • Zhu YX, Yin H, Bruins LA, et al. RNA interference screening identifies lenalidomide sensitizers in multiple myeloma, including RSK2. Blood. 2015;125:483–491.
  • Matthews GM, de Matos Simoes R, Dhimolea E, et al. NF-κB dysregulation in multiple myeloma. Semin Cancer Biol. 2016;39:68–76.
  • Piazza F, Manni S, Semenzato G. Novel players in multiple myeloma pathogenesis: role of protein kinases CK2 and GSK3. Leuk Res. 2013;37:221–227.
  • Manni S, Toscani D, Mandato E, et al. Bone marrow stromal cell-fueled multiple myeloma growth and osteoclastogenesis are sustained by protein kinase CK2. Leukemia. 2014;28:2094–2097.
  • Chon HJ, Bae KJ, Lee Y, et al. The casein kinase 2 inhibitor, CX-4945, as an anti-cancer drug in treatment of human hematological malignancies. Front Pharmacol. 2015;6:70.
  • Papaioannou A, Chevet E. Driving cancer tumorigenesis and metastasis through UPR signaling. Curr Top Microbiol Immunol. 2018;414:159–192.
  • Vincenz L, Jager R, O’Dwyer M, et al. Endoplasmic reticulum stress and the unfolded protein response: targeting the achilles heel of multiple myeloma. Mol Cancer Ther. 2013;12:831–843.
  • Usmani SZ, Chiosis G. HSP90 inhibitors as therapy for multiple myeloma. Clin Lymphoma Myeloma Leuk. 2011;11:S77–81.
  • Manni S, Brancalion A, Tubi LQ, et al. Protein kinase CK2 protects multiple myeloma cells from ER stress-induced apoptosis and from the cytotoxic effect of HSP90 inhibition through regulation of the unfolded protein response. Clin Cancer Res. 2012;18:1888–1900.
  • Tiedemann RE, Zhu YX, Schmidt J, et al. Kinome-wide RNAi studies in human multiple myeloma identify vulnerable kinase targets, including a lymphoid-restricted kinase, GRK6. Blood. 2010;115:1594–1604.
  • de Boussac H, Bruyer A, Robert N, et al. Kinome expression profiling in multiple myeloma identifies new therapeutic targets. Blood. 2017;130:1766.
  • Vikova V, Jourdan M, Robert N, et al. Comprehensive characterization of the mutational landscape in multiple myeloma cell lines reveals potential drivers and pathways associated with tumor progression and drug resistance. Theranostics. 2019;9:540–553.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.