Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-c4f8m Total loading time: 0 Render date: 2024-04-20T01:47:47.739Z Has data issue: false hasContentIssue false

9 - Principles of pathway directed therapy

from Section 2 - Biological basis for targeted therapies in myeloma

Published online by Cambridge University Press:  18 December 2013

Stephen A. Schey
Affiliation:
Department of Haematology, King’s College Hospital, London
Kwee L. Yong
Affiliation:
Department of Haematology, University College Hospital, London
Robert Marcus
Affiliation:
Department of Haematology, King’s College Hospital, London
Kenneth C. Anderson
Affiliation:
Dana-Farber Cancer Institute, Boston
Get access

Summary

Introduction

One of the hallmarks of the malignant phenotype is the ability of cells to grow in an autonomous manner with minimal cell death. This has been linked to diverse machinery of the proliferative and/or survival signaling pathways that become constitutively activated or deregulated in all human cancers. The proto-oncogenic driver mutations of Ras (H-, N-, K-ras, depending on tissue origin) or Raf gene are frequently tied to constitutive mitogen activated protein kinases (MAPK)/the extracellular signal regulated kinase (ERK1/2) signaling in a variety of tumors (Figure 9.1). Since the ERK signaling pathway is involved in both physiological and pathological cell proliferation, ERK1/2 inhibitors represent a desirable class of anti-neoplastic agents. Accumulating results have independently supported the promising anti-cancer effects of this class of inhibitor in numerous pre-clinical and clinical studies, i.e. CI-1040[1], selumetinib (AZD6244)[2–4], AS703026[5] and GSK1120212[6]. In fact, the MEK1/2 inhibitor CI-1040 (Pfizer) was the first signal transduction inhibitor tested in clinical trials in various advanced solid tumors[7]. Although no MEK1/2 inhibitors are approved for clinical use, kinase inhibitors that also inhibit Raf and VEGFR kinases (e.g. Sorafenib inhibiting multikinases)[8, 9] are successful anti-neoplastic agents against many types of cancer and FDA approved for the treatment of patients with unresectable hepatocellular carcinoma and advanced renal cell carcinoma [10, 11] (Figure 9.2). Such information support targeting this pathway, alone or in combination with other well-established growth and survival pathways, i.e. phosphoinositide 3-kinase (PI3K)/Akt/mTOR, STAT3, NFκB, heat shock proteins (Hsp90, Hsp70), histone deacetylases (HDAC), osteoblast (OB) as effective strategies across multiple cancers.

Type
Chapter
Information
Myeloma
Pathology, Diagnosis, and Treatment
, pp. 110 - 120
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Barrett, SD, Bridges, AJ, Dudley, DT, et al. The discovery of the benzhydroxamate MEK inhibitors CI-1040 and PD 0325901. Bioorg Med Chem Lett. 2008;18:6501–6504.CrossRefGoogle ScholarPubMed
Tai, YT, Fulciniti, M, Hideshima, T, et al. Targeting MEK induces myeloma-cell cytotoxicity and inhibits osteoclastogenesis. Blood. 2007;110:1656–1663.CrossRefGoogle ScholarPubMed
Kirkwood, JM, Bastholt, L, Robert, C, et al. Phase II, open-label, randomized trial of the MEK1/2 inhibitor selumetinib as monotherapy versus temozolomide in patients with advanced melanoma. Clin Cancer Res. 2012;18:555–567.CrossRefGoogle ScholarPubMed
Banerji, U, Camidge, DR, Verheul, HM, et al. The first-in-human study of the hydrogen sulfate (Hyd-sulfate) capsule of the MEK1/2 inhibitor AZD6244 (ARRY-142886): a phase I open-label multicenter trial in patients with advanced cancer. Clin Cancer Res. 2010;16:1613–1623.CrossRefGoogle ScholarPubMed
Yoon, J, Koo, KH, Choi, KY.MEK1/2 inhibitors AS703026 and AZD6244 may be potential therapies for KRAS mutated colorectal cancer that is resistant to EGFR monoclonal antibody therapy. Cancer Res. 2011;71:445–453.CrossRefGoogle ScholarPubMed
Gilmartin, AG, Bleam, MR, Groy, A, et al. GSK1120212 (JTP-74057) is an inhibitor of MEK activity and activation with favorable pharmacokinetic properties for sustained in vivo pathway inhibition. Clin Cancer Res. 2011;17:989–1000.CrossRefGoogle ScholarPubMed
Rinehart, J, Adjei, AA, Lorusso, PM, et al. Multicenter phase II study of the oral MEK inhibitor, CI-1040, in patients with advanced non-small-cell lung, breast, colon, and pancreatic cancer. J Clin Oncol. 2004;22:4456–4462.CrossRefGoogle ScholarPubMed
Adnane, L, Trail, PA, Taylor, I, Wilhelm, SM.Sorafenib (BAY 43-9006, Nexavar), a dual-action inhibitor that targets RAF/MEK/ERK pathway in tumor cells and tyrosine kinases VEGFR/PDGFR in tumor vasculature. Methods Enzymol. 2006;407:597–612.CrossRefGoogle Scholar
Wilhelm, SM, Adnane, L, Newell, P, Villanueva, A, Llovet, JM, Lynch, M.Preclinical overview of sorafenib, a multikinase inhibitor that targets both Raf and VEGF and PDGF receptor tyrosine kinase signaling. Mol Cancer Ther. 2008;7:3129–3140.CrossRefGoogle ScholarPubMed
Messmer, D, Fecteau, JF, O’Hayre, M, Bharati, IS, Handel, TM, Kipps, TJ.Chronic lymphocytic leukemia cells receive RAF-dependent survival signals in response to CXCL12 that are sensitive to inhibition by sorafenib. Blood. 2011;117:882–889.CrossRefGoogle ScholarPubMed
Widemann, BC, Kim, A, Fox, E, et al. A phase I trial and pharmacokinetic study of the Raf kinase and receptor tyrosine kinase inhibitor sorafenib in children with refractory solid tumors or refractory leukemias. Clin Cancer Res. 2012.
Flaherty, KT, Yasothan, U, Kirkpatrick, P.Vemurafenib. Nat Rev Drug Discov. 2011;10:811–812.CrossRefGoogle ScholarPubMed
Kim, K, Kong, SY, Fulciniti, M, et al. Blockade of the MEK/ERK signalling cascade by AS703026, a novel selective MEK1/2 inhibitor, induces pleiotropic anti-myeloma activity in vitro and in vivo. Br J Haematol. 2010;149:537–549.CrossRefGoogle ScholarPubMed
Chapman, MA, Lawrence, MS, Keats, JJ, et al. Initial genome sequencing and analysis of multiple myeloma. Nature. 2011;471:467–472.CrossRefGoogle ScholarPubMed
Hideshima, T, Mitsiades, C, Tonon, G, Richardson, PG, Anderson, KC.Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets. Nat Rev Cancer. 2007;7:585–598.CrossRefGoogle ScholarPubMed
Berns, K, Horlings, HM, Hennessy, BT, et al. A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer. Cancer Cell. 2007;12:395–402.CrossRefGoogle ScholarPubMed
Woyach, JA, Johnson, AJ, Byrd, JC.The B-cell receptor signaling pathway as a therapeutic target in CLL. Blood. 2012;120:1175–1184.CrossRefGoogle ScholarPubMed
Baumann, P, Mandl-Weber, S, Oduncu, F, Schmidmaier, R.The novel orally bioavailable inhibitor of phosphoinositol-3-kinase and mammalian target of rapamycin, NVP-BEZ235, inhibits growth and proliferation in multiple myeloma. Exp Cell Res. 2009;315:485–497.CrossRefGoogle ScholarPubMed
Leung, E, Kim, JE, Rewcastle, GW, Finlay, GJ, Baguley, BC.Comparison of the effects of the PI3K/mTOR inhibitors NVP-BEZ235 and GSK2126458 on tamoxifen-resistant breast cancer cells. Cancer Biol Ther. 2011;11:938–946.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
Jhaveri, K, Taldone, T, Modi, S, Chiosis, G.Advances in the clinical development of heat shock protein 90 (Hsp90) inhibitors in cancers. Biochim Biophys Acta. 2012;1823:742–755.CrossRefGoogle ScholarPubMed
Erlichman, C.Tanespimycin: the opportunities and challenges of targeting heat shock protein 90. Expert Opin Investig Drugs. 2009;18:861–868.CrossRefGoogle ScholarPubMed
Plesner, T, Lokhorst, HM, Gimsing, P, Nahi, H, Lisby, S, Richardson, P. Daratumumab, a CD38 mab, for the treatment of relapsed/refractory multiple myeloma patients: Preliminary efficacy data from a multicenter phase I/II study. J Clin Oncol 30, 2012 (suppl; abstr 8019) 2012.
Mitsiades, CS, Hideshima, T, Chauhan, D, et al. Emerging treatments for multiple myeloma: beyond immunomodulatory drugs and bortezomib. Semin Hematol. 2009;46:166–175.CrossRefGoogle ScholarPubMed
Allegra, A, Sant’antonio, E, Penna, G, et al. Novel therapeutic strategies in multiple myeloma: role of the heat shock protein inhibitors. Eur J Haematol. 2011;86:93–110.CrossRefGoogle ScholarPubMed
Ellis, LM, Hicklin, DJ.VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat Rev Cancer. 2008;8:579–591.CrossRefGoogle ScholarPubMed
Podar, K, Tai, YT, Hideshima, T, Vallet, S, Richardson, PG, Anderson, KC.Emerging therapies for multiple myeloma. Expert Opin Emerg Drugs. 2009;14:99–127.CrossRefGoogle ScholarPubMed
Anderson, KC.The 39th David A. Karnofsky Lecture: bench-to-bedside translation of targeted therapies in multiple myeloma. J Clin Oncol. 2012;30:445–452.CrossRefGoogle Scholar
Chauhan, D, Tian, Z, Nicholson, B, et al. A small molecule inhibitor of ubiquitin-specific protease-7 induces apoptosis in multiple myeloma cells and overcomes bortezomib resistance. Cancer Cell. 2012;22:345–358.CrossRefGoogle ScholarPubMed
McMillin, DW, Jacobs, HM, Delmore, JE, et al. Molecular and cellular effects of NEDD8-activating enzyme inhibition in myeloma. Mol Cancer Ther. 2012;11:942–951.CrossRefGoogle Scholar
Tai, YT, Anderson, KC.Antibody-based therapies in multiple myeloma. Bone Marrow Res. 2011;2011:924058.CrossRefGoogle ScholarPubMed
Tai, YT, Horton, HM, Kong, SY, et al. Potent in vitro and in vivo activity of an Fc-engineered humanized anti-HM1.24 antibody against multiple myeloma via augmented effector function. Blood. 2012;119:2074–2082.CrossRefGoogle ScholarPubMed
Raje, N, Roodman, GD.Advances in the biology and treatment of bone disease in multiple myeloma. Clin Cancer Res. 2011;17:1278–1286.CrossRefGoogle ScholarPubMed
Hideshima, T, Richardson, PG, Anderson, KC.Mechanism of action of proteasome inhibitors and deacetylase inhibitors and the biological basis of synergy in multiple myeloma. Mol Cancer Ther. 2011;10:2034–2042.CrossRefGoogle ScholarPubMed
Hideshima, T, Bradner, JE, Wong, J, et al. Small-molecule inhibition of proteasome and aggresome function induces synergistic antitumor activity in multiple myeloma. Proc Natl Acad Sci U S A. 2005;102:8567–8572.CrossRefGoogle ScholarPubMed
Santo, L, Hideshima, T, Kung, AL, et al. Preclinical activity, pharmacodynamic, and pharmacokinetic properties of a selective HDAC6 inhibitor, ACY-1215, in combination with bortezomib in multiple myeloma. Blood. 2012;119:2579–2589.CrossRefGoogle ScholarPubMed
Stewart, AK.Novel therapeutics in multiple myeloma. Hematology. 2012;17 Suppl 1:S105–108.Google ScholarPubMed
Shinohara, M, Koga, T, Okamoto, K, et al. Tyrosine kinases Btk and Tec regulate osteoclast differentiation by linking RANK and ITAM signals. Cell. 2008;132:794–806.CrossRefGoogle ScholarPubMed
de Rooij, MF, Kuil, A, Geest, CR, et al. 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.CrossRefGoogle ScholarPubMed
Tai, YT, Chang, BY, Kong, SY, 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.CrossRefGoogle ScholarPubMed
Treon, SP, Xu, L, Yang, G, et al. MYD88 L265P somatic mutation in Waldenstrom’s macroglobulinemia. N Engl J Med. 2012;367:826–833.CrossRefGoogle ScholarPubMed
Yang, G, Xu, L, Zhou, Y, et al. Participation of BTK in MYD88 signaling in malignant cells expressing the L265P mutation in Waldenstrom’s macroglobulinemia, and effect on tumor cells with BTK-inhibitor PCI-32765 in combination with MYD88 pathway inhibitors. J Clin Oncol. 2012 30:abstr 8106Google Scholar
Yang, Y, Shaffer, AL, 3rd, Emre, NC, et al. Exploiting synthetic lethality for the therapy of ABC diffuse large B cell lymphoma. Cancer Cell. 2012;21:723–737.CrossRefGoogle ScholarPubMed
Turner, JG, Dawson, J, Sullivan, DM.Nuclear export of proteins and drug resistance in cancer. Biochem Pharmacol. 2012;83:1021–1032.CrossRefGoogle Scholar
Xu, D, Farmer, A, Collett, G, Grishin, NV, Chook, YM.Sequence and structural analyses of nuclear export signals in the NESdb database. Mol Biol Cell. 2012;23:3677–93.CrossRefGoogle ScholarPubMed
Etchin, J, Sun, Q, Kentsis, A, et al. Anti-leukemic activity of nuclear export inhibitors that spare normal hematopoietic cells. Leukemia. 2013;27:66–74.CrossRefGoogle Scholar
Tai, YT, Landesman, Y, Acharya, C, et al. The nuclear export protein CRM1 (exportin 1) regulates multiple myeloma cell growth, osteoclastogenesis, and myeloma-induced osteolysis. Leukemia 2013 Apr 16. . [Epub ahead of print]
Tiedemann, RE, Zhu, YX, Schmidt, J, et al. Identification of molecular vulnerabilities in human multiple myeloma cells by RNA interference lethality screening of the druggable genome. Cancer Res. 2012;72:757–768.CrossRefGoogle ScholarPubMed
Henkin, RI.Vismodegib in advanced basal-cell carcinoma. N Engl J Med. 2012;367:970; author reply 970–971.Google ScholarPubMed
Lonial, S, Vij, R, Harousseau, JL, et al. Elotuzumab in combination with lenalidomide and low-dose dexamethasone in relapsed or refractory multiple myeloma. J Clin Oncol. 2012;30:1953–1959.CrossRefGoogle ScholarPubMed
Edwards, CM.BTK inhibition in myeloma: targeting the seed and the soil. Blood. 2012;120:1757–1759.CrossRefGoogle ScholarPubMed
Robak, T, Robak, E.Tyrosine kinase inhibitors as potential drugs for B-cell lymphoid malignancies and autoimmune disorders. Expert Opin Investig Drugs. 2012;21:921–947.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×