314
Views
14
CrossRef citations to date
0
Altmetric
Reviews

Targeted Agents in Preclinical and Early Clinical Development for the Treatment of Cancer Bone Metastases

Pages 319-334 | Received 18 Nov 2015, Accepted 13 Jan 2016, Published online: 06 Feb 2016

References

  • Mundy GR. Metastasis to bone: causes, consequences and therapeutic opportunities. Nat Rev Cancer. 2002;2:584–593.
  • Weilbaecher KN, Guise TA, McCauley LK. Cancer to bone: a fatal attraction. Nat Rev Cancer. 2011;11:411–425.
  • Coleman RE. Clinical features of metastatic bone disease and risk of skeletal morbidity. Clin Cancer Res. 2006;12:6243s– 6249s.
  • Wong MH, Pavlakis N. Optimal management of bone metastases in breast cancer patients. Breast Cancer Targets Ther. 2011;3:35.
  • Sun L, Yu S. Efficacy and safety of denosumab versus zoledronic acid in patients with bone metastases: a systematic review and meta-analysis. Am J Clin Oncol. 2013;36:399–403.
  • Coleman R, Gnant M, Morgan G, et al. Effects of bone-targeted agents on cancer progression and mortality. J Natl Cancer Inst. 2012;104:1059–1067.
  • Costa AG, Cusano NE, Silva BC, et al. Cathepsin K: its skeletal actions and role as a therapeutic target in osteoporosis. Nat Rev Rheumatol. 2011;7:447–456.
  • Saftig P, Hunziker E, Wehmeyer O, et al. Impaired osteoclastic bone resorption leads to osteopetrosis in cathepsin-K-deficient mice. Proc Natl Acad Sci U S A. 1998;95:13453–13458.
  • Gelb BD, Shi G-P, Chapman HA, et al. Pycnodysostosis, a lysosomal disease caused by cathepsin K deficiency. Science. 1996;273:1236–1238.
  • Le Gall C, Bellahcene A, Bonnelye E, et al. A cathepsin K inhibitor reduces breast cancer induced osteolysis and skeletal tumor burden. Cancer Res. 2007;67:9894–9902.
  • Littlewood-Evans AJ, Bilbe G, Bowler WB, et al. The osteoclast-associated protease cathepsin K is expressed in human breast carcinoma. Cancer Res. 1997;57:5386–5390.
  • Brubaker KD, Vessella RL, True LD, et al. Cathepsin K mRNA and protein expression in prostate cancer progression. J Bone Miner Res. 2003;18:222–230.
  • Chapurlat RD. Odanacatib: a review of its potential in the management of osteoporosis in postmenopausal women. Ther Adv Musculoskelet Dis. 2015;7:103–109.
  • Jensen AB, Wynne C, Ramirez G, et al. The cathepsin K inhibitor odanacatib suppresses bone resorption in women with breast cancer and established bone metastases: results of a 4-week, double-blind, randomized, controlled trial. Clin Breast Cancer. 2010;10:452–458.
  • Mayer EL, Krop IE. Advances in targeting Src in the treatment of breast cancer and other solid malignancies. Clin Cancer Res. 2010;16:3526–3532.
  • Zhang S, Yu D. Targeting Src family kinases in anti-cancer therapies: turning promise into triumph. Trends Pharmacol Sci. 2012;33:122–128.
  • Soriano P, Montgomery C, Geske R, et al. Targeted disruption of the c-src proto-oncogene leads to osteopetrosis in mice. Cell. 1991;64:693–702.
  • Boyce BF, Yoneda T, Lowe C, et al. Requirement of pp60c-src expression for osteoclasts to form ruffled borders and resorb bone in mice. J Clin Invest. 1992;90:1622–1627.
  • Myoui A, Nishimura R, Williams PJ, et al. C-src tyrosine kinase activity is associated with tumor colonization in bone and lung in an animal model of human breast cancer metastasis. Cancer Res. 2003;63:5028–5033.
  • Zhang XHF, Wang Q, Gerald W, et al. Latent bone metastasis in breast cancer tied to Src-dependent survival signals. Cancer Cell. 2009;16:67–78.
  • Koreckij T, Nguyen H, Brown LG, et al. Dasatinib inhibits the growth of prostate cancer in bone and provides additional protection from osteolysis. Br J Cancer. 2009;101:263–268.
  • Rabbani SA, Valentino M-L, Arakelian A, et al. SKI-606 (Bosutinib) blocks prostate cancer invasion, growth, and metastasis in vitro and in vivo through regulation of genes involved in cancer growth and skeletal metastasis. Mol Cancer Ther. 2010;9:1147–1157.
  • Yang JC, Bai L, Yap S, et al. Effect of the specific src family kinase inhibitor saracatinib on osteolytic lesions using the PC-3 bone model. Mol Cancer Ther. 2010;9:1629–1637.
  • Takahashi S, Miyazaki M, Okamoto I, et al. Phase I study of dasatinib (BMS-354825) in Japanese patients with solid tumors. Cancer Sci. 2011;102:2058–2064.
  • Yu EY, Wilding G, Posadas E, et al. Phase II study of dasatinib in patients with metastatic castration-resistant prostate cancer. Clin Cancer Res. 2009;15:7421–7428.
  • Araujo JC, Trudel GC, Saad F, et al. Docetaxel and dasatinib or placebo in men with metastatic castration-resistant prostate cancer (READY): a randomised, double-blind phase 3 trial. Lancet Oncol. 2013;14:1307–1316.
  • Barczyk M, Carracedo S, Gullberg D. Integrins. Cell Tissue Res. 2010;339:269–280.
  • Desgrosellier JS, Cheresh DA. Integrins in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer. 2010;10:9–22.
  • Schneider JG, Amend SR, Weilbaecher KN. Integrins and bone metastasis: integrating tumor cell and stromal cell interactions. Bone. 2011;48:54–65.
  • Xu F, Teitelbaum SL. Osteoclasts: new insights. Bone Res. 2013;1:11–26.
  • Brooks P, Clark R, Cheresh D. Requirement of vascular integrin alpha v beta 3 for angiogenesis. Science. 1994;264:569–571.
  • Pécheur I, Peyruchaud O, Serre CM, et al. Integrin alpha(v)beta3 expression confers on tumor cells a greater propensity to metastasize to bone. Faseb J. 2002;16:1266–1268.
  • Zhao Y, Bachelier R, Treilleux I, et al. Tumor αvβ3 integrin is a therapeutic target for breast cancer bone metastases. Cancer Res. 2007;67:5821–5830.
  • Bakewell SJ, Nestor P, Prasad S, et al. Platelet and osteoclast beta3 integrins are critical for bone metastasis. Proc Natl Acad Sci U S A. 2003;100:14205–14210.
  • Mulgrew K, Kinneer K, Yao X-T, et al. Direct targeting of αvβ3 integrin on tumor cells with a monoclonal antibody, Abegrin. Mol Cancer Ther. 2006;5:3122–3129.
  • Gramoun A, Shorey S, Bashutski JD, et al. Effects of Vitaxin, a novel therapeutic in trial for metastatic bone tumors, on osteoclast functions in vitro. J Cell Biochem. 2007;102:341–352.
  • Bretschi M, Merz M, Komljenovic D, et al. Cilengitide inhibits metastatic bone colonization in a nude rat model. Oncol Rep. 2011;26:843–851.
  • Forgac M. Vacuolar ATPases: rotary proton pumps in physiology and pathophysiology. Nat Rev Mol Cell Biol. 2007;8:917–929.
  • Keeling DJ, Herslof M, Ryberg B, et al. Vacuolar H+-ATPases. Targets for drug discovery? Ann N Y Acad Sci. 1997;834:600–608.
  • Li YP, Chen W, Liang Y, et al. Atp6i-deficient mice exhibit severe osteopetrosis due to loss of osteoclast-mediated extracellular acidification. Nat Genet. 1999;23:447–451.
  • Visentin L, Dodds RA, Valente M, et al. A selective inhibitor of the osteoclastic V-H+-ATPase prevents bone loss in both thyroparathyroidectomized and ovariectomized rats. J Clin Invest. 2000;106:309–318.
  • Niikura K, Takeshita N, Takano M. A vacuolar ATPase inhibitor, FR167356, prevents bone resorption in ovariectomized rats with high potency and specificity: potential for clinical application. J Bone Miner Res. 2005;20:1579–1588.
  • Nishisho T, Hata K, Nakanishi M, et al. The a3 isoform vacuolar type H+-ATPase promotes distant metastasis in the mouse B16 melanoma cells. Mol Cancer Res. 2011;9:845–855.
  • Capecci J, Forgac M. The function of vacuolar ATPase (V-ATPase) a subunit isoforms in invasiveness of MCF10a and MCF10CA1a human breast cancer cells. J Biol Chem. 2013;288:32731–32741.
  • Terpos E, Christoulas D. Effects of proteasome inhibitors on bone cancer. Bonekey Rep. 2013;2:395.
  • Mohty M, Malard F, Mohty B, et al. The effects of bortezomib on bone disease in patients with multiple myeloma. Cancer. 2014;120:618–623.
  • Zavrski I, Krebbel H, Wildemann B, et al. Proteasome inhibitors abrogate osteoclast differentiation and osteoclast function. Biochem Biophys Res Commun. 2005;333:200–205.
  • Garrett IR, Chen D, Gutierrez G, et al. Selective inhibitors of the osteoblast proteasome stimulate bone formation in vivo and in vitro. J Clin Invest. 2003;111:1771–1782.
  • Bellido T, Ali AA, Plotkin LI, et al. Proteasomal degradation of Runx2 shortens parathyroid hormone-induced anti-apoptotic signaling in osteoblasts. A putative explanation for why intermittent administration is needed for bone anabolism. J Biol Chem. 2003;278:50259–50272.
  • Qiang Y-W, Hu B, Chen Y, et al. Bortezomib induces osteoblast differentiation via Wnt-independent activation of -catenin/TCF signaling. Blood. 2009;113:4319–4330.
  • Oyajobi BO, Garrett IR, Gupta A, et al. Stimulation of new bone formation by the proteasome inhibitor, bortezomib: implications for myeloma bone disease. Br J Haematol. 2007;139:434–438.
  • 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.
  • Sunwoo JB, Chen Z, Dong G, et al. Novel proteasome inhibitor PS-341 inhibits activation of nuclear factor-kB, cell survival, tumor growth, and angiogenesis in squamous cell carcinoma. Clin Cancer Res. 2001;7:1419–1428.
  • Whang PG, Gamradt SC, Gates JJ, et al. Effects of the proteasome inhibitor bortezomib on osteolytic human prostate cancer cell metastases. Prostate Cancer Prostatic Dis. 2005;8:327–334.
  • Jones MD, Liu JC, Barthel TK, et al. A proteasome inhibitor, bortezomib, inhibits breast cancer growth and reduces osteolysis by downregulating metastatic genes. Clin Cancer Res. 2010;16:4978–4989.
  • Yang CH, Gonzalez-Angulo AM, Reuben JM, et al. Bortezomib (VELCADE) in metastatic breast cancer: pharmacodynamics, biological effects, and prediction of clinical benefits. Ann Oncol. 2006;17:813–817.
  • Pal A, Young MA, Donato NJ. Emerging potential of therapeutic targeting of ubiquitin-specific proteases in the treatment of cancer. Cancer Res. 2014;74:4955–4966.
  • Sabatini DM. mTOR and cancer: insights into a complex relationship. Nat Rev Cancer. 2006;6:729–734.
  • Porta C, Paglino C, Mosca A. Targeting PI3K/Akt/mTOR signaling in cancer. Front Oncol. 2014;4:1–11.
  • Kneissel M, Luong-Nguyen N-H, Baptist M, et al. Everolimus suppresses cancellous bone loss, bone resorption, and cathepsin K expression by osteoclasts. Bone. 2004;35:1144–1156.
  • Okui T, Shimo T, Fukazawa T, et al. Antitumor effect of temsirolimus against oral squamous cell carcinoma associated with bone destruction. Mol Cancer Ther. 2010;9:2960–2969.
  • Hussein O, Tiedemann K, Murshed M, et al. Rapamycin inhibits osteolysis and improves survival in a model of experimental bone metastases. Cancer Lett. 2012;314:176–184.
  • Morgan TM, Pitts TEM, Gross TS, et al. RAD001 (Everolimus) inhibits growth of prostate cancer in the bone and the inhibitory effects are increased by combination with docetaxel and zoledronic acid. Prostate. 2008;68:861–871.
  • Gnant M, Baselga J, Rugo HS, et al. Effect of Everolimus on bone marker levels and progressive disease in bone in BOLERO-2. J Natl Cancer Inst. 2013;105:654–663.
  • Ng JMY, Curran T. The Hedgehog’s tale: developing strategies for targeting cancer. Nat Rev Cancer. 2011;11:493–501.
  • Heller E, Hurchla MA, Xiang J, et al. Hedgehog signaling inhibition blocks growth of resistant tumors through effects on tumor microenvironment. Cancer Res. 2012;72:897–907.
  • Yaccoby S, Wezeman MJ, Zangari M, et al. Inhibitory effects of osteoblasts and increased bone formation on myeloma in novel culture systems and a myelomatous mouse model. Haematologica. 2006;91:192–199.
  • Krawetz R, Wu YE, Rancourt DE, et al. Osteoblasts suppress high bone turnover caused by osteolytic breast cancer in-vitro. Exp Cell Res. 2009;315:2333–2342.
  • Pennisi A, Ling W, Li X, et al. Consequences of daily administered parathyroid hormone on myeloma growth, bone disease, and molecular profiling of whole myelomatous bone. PLoS One. 2010;5:e15233.
  • Canalis E. Wnt signalling in osteoporosis: mechanisms and novel therapeutic approaches. Nat Rev Endocrinol. 2013;9:575–583.
  • Anastas JN, Moon RT. WNT signalling pathways as therapeutic targets in cancer. Nat Rev Cancer. 2013;13:11–26.
  • Tian E, Zhan F, Walker R, et al. The role of the Wnt-signalling antagonist DKK1 in the development of osteolytic lesions in multiple myeloma. N Engl J Med. 2003;349:2483–2494.
  • Voorzanger-Rousselot N, Goehrig D, Journe F, et al. Increased Dickkopf-1 expression in breast cancer bone metastases. Br J Cancer. 2007;97:964–970.
  • Thudi NK, Martin CK, Murahari S, et al. Dickkopf-1 (DKK-1) stimulated prostate cancer growth and metastasis and inhibited bone formation in osteoblastic bone metastases. Prostate. 2011;71:615–625.
  • Yaccoby S, Ling W, Zhan F, et al. Antibody-based inhibition of DKK1 suppresses tumor-induced bone resorption and multiple myeloma growth in vivo. Blood. 2007;109:2106–2111.
  • Iyer SP, Beck JT, Stewart AK, et al. A Phase IB multicentre dose-determination study of BHQ880 in combination with anti-myeloma therapy and zoledronic acid in patients with relapsed or refractory multiple myeloma and prior skeletal-related events. Br J Haematol. 2014;167:366–375.
  • Mendoza-Villanueva D, Zeef L, Shore P. Metastatic breast cancer cells inhibit osteoblast differentiation through the Runx2/CBFβ-dependent expression of the Wnt antagonist, sclerostin. Breast Cancer Res. 2011;13:R106.
  • McClung MR, Grauer A, Boonen S, et al. Romosozumab in postmenopausal women with low bone mineral density. N Engl J Med. 2014;370:412–420.
  • Leto G. Activin A and bone metastasis. J Cell Physiol. 2010;225:302–309.
  • Incorvaia L, Badalamenti G, Rini G, et al. MMP-2, MMP-9 and activin a blood levels in patients with breast cancer or prostate cancer metastatic to the bone. Anticancer Res. 2007;27:1519–1525.
  • Terpos E, Kastritis E, Christoulas D, et al. Circulating activin-A is elevated in patients with advanced multiple myeloma and correlates with extensive bone involvement and inferior survival; no alterations post-lenalidomide and dexamethasone therapy. Ann Oncol. 2012;23:2681–2686.
  • Chantry AD, Heath D, Mulivor AW, et al. Inhibiting activin-A signaling stimulates bone formation and prevents cancer-induced bone destruction in vivo. J Bone Miner Res. 2010;25:2633–2646.
  • Abdulkadyrov KM, Salogub GN, Khuazheva NK, et al. Sotatercept in patients with osteolytic lesions of multiple myeloma. Br J Haematol. 2014;165:814–823.
  • Rosanò L, Spinella F, Bagnato A. Endothelin 1 in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer. 2013;13:637–651.
  • Yin JJ, Mohammad KS, Kakonen SM, et al. A causal role for endothelin-1 in the pathogenesis of osteoblastic bone metastases. Proc Natl Acad Sci U S A. 2003;100:10954–10959.
  • Carducci MA, Saad F, Abrahamsson P-A, et al A phase 3 randomized controlled trial of the efficacy and safety of atrasentan in men with metastatic hormone-refractory prostate cancer. Cancer. 2007;110:1959–1966.
  • Quinn DI, Tangen CM, Hussain M, et al. Docetaxel and atrasentan versus docetaxel and placebo for men with advanced castration-resistant prostate cancer (SWOG S0421): a randomised phase 3 trial. Lancet Oncol. 2013;14:893–900.
  • Lara PN, Ely B, Quinn DI, et al. Serum biomarkers of bone metabolism in castration-resistant prostate cancer patients with skeletal metastases: results from SWOG 0421. J Natl Cancer Inst. 2014;106:dju013.
  • Nelson JB, Fizazi K, Miller K, et al. Phase 3, randomized, placebo-controlled study of zibotentan (ZD4054) in patients with castration-resistant prostate cancer metastatic to bone. Cancer. 2012;118:5709–5718.
  • Fizazi KS, Higano CS, Nelson JB, et al. Phase III, randomized, placebo-controlled study of docetaxel in combination with zibotentan in patients with metastatic castration-resistant prostate cancer. J Clin Oncol. 2013;31:1740–1747.
  • Ikushima H, Miyazono K. TGFβ signalling: a complex web in cancer progression. Nat Rev Cancer. 2010;10:415–424.
  • Korpal M, Yan J, Lu X, et al. Imaging transforming growth factor-β signaling dynamics and therapeutic response in breast cancer bone metastasis. Nat Med. 2009;15:960–966.
  • Juárez P, Guise TA. TGF-β in cancer and bone: implications for treatment of bone metastases. Bone. 2011;48:23–29.
  • Buijs JT, Stayrook KR, Guise TA. The role of TGF-β in bone metastasis: novel therapeutic perspectives. Bonekey Rep. 2012;1:1–10.
  • Pollak M. The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nat Rev Cancer. 2012;12:159–169.
  • Hiraga T, Myoui A, Hashimoto N, et al. Bone-derived IGF mediates crosstalk between bone and breast cancer cells in bony metastases. Cancer Res. 2012;72:4238–4249.
  • Goya M, Miyamoto S, Nagai K, et al. Growth inhibition of human prostate cancer cells in human adult bone implanted into nonobese diabetic/severe combined immunodeficient mice by a ligand-specific antibody to human insulin-like growth factors. Cancer Res. 2004;64:6252–6258.
  • Logan JG, Sophocleous A, Marino S, et al. Selective tyrosine kinase inhibition of insulin-like growth factor-1 receptor inhibits human and mouse breast cancer-induced bone cell activity, bone remodeling, and osteolysis. J Bone Miner Res. 2013;28:1229–1242.
  • Bowers LW, Rossi EL, O’Flanagan CH, et al. The role of the Insulin/IGF system in cancer: lessons learned from clinical trials and the energy balance-cancer link. Front Endocrinol. 2015;6:77.
  • Scala S. Molecular pathways: targeting the CXCR4-CXCL12 axis-untapped potential in the tumor microenvironment. Clin Cancer Res. 2015;21:4278–4285.
  • Shiozawa Y, Pienta KJ, Taichman RS. Hematopoietic stem cell niche is a potential therapeutic target for bone metastatic tumors. Clin Cancer Res. 2011;17:5553–5558.
  • Andre F, Xia W, Conforti R, et al. CXCR4 expression in early breast cancer and risk of distant recurrence. Oncologist. 2009;14:1182–1188.
  • Richert MM, Vaidya KS, Mills CN, et al. Inhibition of CXCR4 by CTCE-9908 inhibits breast cancer metastasis to lung and bone. Oncol Rep. 2009;21:761–767.
  • Gravina GL, Mancini A, Muzi P, et al. CXCR4 pharmacogical inhibition reduces bone and soft tissue metastatic burden by affecting tumor growth and tumorigenic potential in prostate cancer preclinical models. Prostate. 2015;75:1227–1246.
  • Ntziachristos P, Lim JS, Sage J, et al. From fly wings to targeted cancer therapies: a centennial for notch signaling. Cancer Cell. 2014;25:318–334.
  • Sethi N, Dai X, Winter CG, et al. Tumor-derived Jagged1 promotes osteolytic bone metastasis of breast cancer by engaging Notch signaling in bone cells. Cancer Cell. 2011;19:192–205.
  • Takebe N, Miele L, Harris PJ, et al. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol. 2015;12:445–464.
  • Okazaki M, Takeshita S, Kawai S, et al. Molecular cloning and characterization of OB-cadherin, a new member of cadherin family expressed in osteoblasts. J Biol Chem. 1994;269:12092–12098.
  • Pishvaian MJ, Feltes CM, Thompson P, et al. Cadherin-11 Is expressed in invasive breast cancer cell lines. Cancer Res. 1999;59:947–952.
  • Chu K, Cheng C-J, Ye X, et al. Cadherin-11 promotes the metastasis of prostate cancer cells to bone. Mol Cancer Res. 2008;6:1259–1267.
  • Huang C-F, Lira C, Chu K, et al. Cadherin-11 increases migration and invasion of prostate cancer cells and enhances their interaction with osteoblasts. Cancer Res. 2010;70:4580–4589.
  • Tamura D, Hiraga T, Myoui A, et al. Cadherin-11-mediated interactions with bone marrow stromal/osteoblastic cells support selective colonization of breast cancer cells in bone. Int J Oncol. 2008;33:17–24.
  • Lee Y-C, Bilen MA, Yu G, et al. Inhibition of cell adhesion by a cadherin-11 antibody thwarts bone metastasis. Mol Cancer Res. 2013;11:1401–1411.
  • Lee RJ, Smith MR. Targeting MET and vascular endothelial growth factor receptor signaling in castration-resistant prostate cancer. Cancer J. 2013;19:90–98.
  • Dai J, Zhang H, Karatsinides A, et al. Cabozantinib inhibits prostate cancer growth and prevents tumor-induced bone lesions. Clin Cancer Res. 2014;20:617–630.
  • Nguyen HM, Ruppender N, Zhang X, et al. Cabozantinib inhibits growth of androgen-sensitive and castration-resistant prostate cancer and affects bone remodeling. PLoS One. 2013;8:1–15.
  • Smith DC, Smith MR, Sweeney C, et al. Cabozantinib in patients with advanced prostate cancer: results of a phase II randomized discontinuation trial. J Clin Oncol. 2013;31:412–419.
  • Pichler M, Calin GA. MicroRNAs in cancer: from developmental genes in worms to their clinical application in patients. Br J Cancer. 2015;113:569-573.
  • Croset M, Kan C, Clézardin P. Tumour-derived miRNAs and bone metastasis. Bonekey Rep. 2015;4:688.
  • Ell B, Mercatali L, Ibrahim T, et al. Tumor-induced osteoclast miRNA changes as regulators and biomarkers of osteolytic bone metastasis. Cancer Cell. 2013;24:542–556.
  • Balic M, Lin H, Young L, et al. Most early disseminated cancer cells detected in bone marrow of breast cancer patients have a putative breast cancer stem cell phenotype. Clin Cancer Res. 2006;12:5615–5621.
  • Hiraga T, Ito S, Nakamura H. Cancer stem-like cell marker CD44 promotes bone metastases by enhancing tumorigenicity, cell motility, and hyaluronan production. Cancer Res. 2013;73:4112–4122.
  • Plaks V, Kong N, Werb Z. The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells? Cell Stem Cell. 2015;16:225–238.
  • Yoneda T, Tanaka S, Hata K. Role of RANKL/RANK in primary and secondary breast cancer. World J Orthop. 2013;4:178–185.

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.