404
Views
13
CrossRef citations to date
0
Altmetric
Review

Pharmacodynamics, clinical findings and approval status of current and emerging tyrosine-kinase inhibitors for pancreatic neuroendocrine tumors

, , , , , & show all
Pages 993-1004 | Received 25 Aug 2019, Accepted 02 Dec 2019, Published online: 12 Dec 2019

References

  • Yao JC, Hassan M, Phan A, et al. One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol. 2008;26:3063–3072.
  • Dasari A, Shen C, Halperin D, et al. Trend in the incidence, prevalence and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol. 2017;3(10):1335–1342.
  • Falconi M, Eriksson B, Kaltsas G, et al. Consensus guidelines update for the management of functional p- NETs (F-p-NETs) and non-functional p-NETs (NF-p-NETs). Neuroendocrinology. 2016;103(2):153–171.
  • Lloyd RV, Osamura R, Kloppel G, et al. WHO classification of tumours of endocrine organs. 4th ed. Lyon, France: IARC Press; 2017.
  • Tonini T, Rossi F, Claudio PP. Molecular basis of angiogenesis and cancer. Oncogene. 2003;22:6549–6556.
  • Scoazec JY. Angiogenesis in neuroendocrine tumors: therapeutic applications. Neuroendocrinology. 2013;97(1):45–56.
  • Marion-Audibert AM, Barel C, Gouysse G, et al. Low microvessel density is an unfavorable histoprognostic factor in pancreatic endocrine tumors. Gastroenterology. 2003;125:1094–1104.
  • Rubbia-Brandt L, Terris B, Giostra E, et al. Lymphatic vessel density and vascular endothelial growth factor-C expression correlate with malignant behavior in human pancreatic endocrine tumors. Clin Cancer Res. 2004;10:6919–6928.
  • Zhang J, Jia Z, Li Q, et al. Elevated expression of vascular endothelial growth factor correlates with increased angiogenesis and decreased progression-free survival among patients with low-grade neuro- endocrine tumors. Cancer. 2007;109:1478–1486.
  • Konstantinova I, Lammert E. Microvascular development: learning from pancreatic islets. Bioessays. 2004;26:1069–1075.
  • Couvelard A, O’Toole D, Turley H, et al. Microvascular density and hypoxia-inducible factor pathway in pancreatic endocrine tumours: negative correlation of microvascular density and vegf expression with tumour progression. Br J Cancer. 2005;92(1):94–101.
  • Hanahan D. Heritable formation of pancreatic beta-cell tumours in transgenic mice expressing recombinant insulin/simian virus 40 oncogenes. Nature. 1985;315(6015):115–122.
  • Christofori G, Naik P, Hanahan D. Vascular endothelial growth factor and its receptors, flt-1 and flk-1, are expressed in normal pancreatic islets and throughout islet cell tumorigenesis. Mol Endocrinol. 1995;9(12):1760–1770.
  • Capurso G, Fazio N, Festa S, et al. Molecular target therapy for gastroenteropancreatic endocrine tumours: biological rationale and clinical perspectives. Crit Rev Oncol Hematol. 2009;72(2):110–124.
  • Faivre S, Djelloul S, Raymond E. New paradigms in anticancer therapy: targeting multiple signaling pathways with kinase inhibitors. Semin Oncol. 2006;33:407–420.
  • Marmor MD, Skaria KB, Yarden Y. Signal transduction and oncogen- esis by ErbB/HER receptors. Int J Radiat Oncol Biol Phys. 2004;58:903–913.
  • Kowanetz M, Ferrara N. Vascular endothelial growth factor signaling pathways: therapeutic perspective. Clin Cancer Res. 2006;12:5018–5022.
  • Shibuya M. Vascular endothelial growth factor (VEGF) and its receptor (VEGFR) signaling in angiogenesis: a crucial target for anti- and pro-angiogenic therapy. Genes Cancer. 2011;2:1097–1105.
  • Alitalo K, Carmeliet P. Molecular mechanisms of lymphangiogenesis in health and disease. Cancer Cell. 2002;1:219–227.
  • Fjallskog ML, Lejonklou MH, Oberg KE, et al. Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors. Clin Cancer Res. 2003;9:1469–1473.
  • Fjallskog ML, Hessman O, Eriksson B, et al. Upregulated expression of PDGF receptor beta in endocrine pancreatic tumors and metastases compared to normal endocrine pancreas. Acta Oncol. 2007;46:741–746.
  • Cavalcanti E, Ignazzi A, De Michele F, et al. PDGFRa expression as a novel therapeutic marker in well-differentiated neuroendocrine tumors. Cancer Biol Ther. 2019;20:423–430.
  • Chow LQM, Eckhardt SG. Sunitinib: from rational design to clinical efficacy. J Clin Oncol. 2007;25:884–896.
  • Mendel DB, Laird AD, Xin X, et al. In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship. Clin Cancer Res. 2003;9:327–337.
  • O’Farrell AM, Abrams TJ, Yuen HA, et al. SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent activity in vitro and in vivo. Blood. 2003;101:3597–3605.
  • Paez-Ribes M, Allen E, Hudock J, et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell. 2009;15(3):220–231.
  • Pietras K, Hanahan D. A multitargeted, metronomic, and maximum-tolerated dose “chemo- switch” regimen is antiangiogenic, producing objective responses and survival benefit in a mouse model of cancer. J Clin Oncol. 2005;23:939–952.
  • Couvelard A, O’Toole D, Turley H, et al. Microvascular density and hypoxia-inducible factor pathway in pancreatic endocrine tumours: negative correlation of microvascular density and VEGF expression with tumour progression. Br J Cancer. 2005;17(92):94–101.
  • Faivre S, Delbaldo C, Vera K, et al. Safety, pharmacokinetic, and antitumor activity of SU11248, a novel oral multitarget tyrosine kinase inhibitor, in patients with cancer. J Clin Oncol. 2006;24:25–35.
  • Kulke MH, Lenz HJ, Meropol NJ, et al. Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol. 2008;26:3403–3410.
  • Bello C, Deprimo S, Friece C, et al. Analysis of circulating biomarkers of sunitinib malate in patients with unresectable neuroendocrine tumors (NET): VEGF, IL-8, and soluble VEGF receptors 2 and 3. J Clin Oncol. 2006;24(18S):4045.
  • Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. New Engl J Med. 2011;364:501–513.
  • Blumenthal GM, Cortazar P, Zhang JJ, et al. FDA approval summary: sunitinib for the treatment of progressive well-differentiated locally advanced or metastatic pancreatic neuroendocrine tumors. Oncologist. 2012;17:1108–1113.
  • Raymond E, Kulke MH, Qin S, et al. Efficacy and safety of sunitinib in patients with well differentiated pancreatic neuroendocrine tumors. Neuroendocrinology. 2018;107:237–245.
  • Beuselinck B, Lambrechts D, Van Brussel T, et al. Efflux pump ABCB1 single nucleotide polymorphisms and dose reductions in patients with metastatic renal cell carcinoma treated with sunitinib. Acta Oncol. 2014;53:1413–1422.
  • Diekstra MH, Swen JJ, Boven E, et al. CYP3A5 and ABCB1 polymorphisms as predictors for sunitinib outcome in metastatic renal cell carcinoma. Eur Urol. 2015;68(4):621–629.
  • Maeng CH, Yi JH, Lee J, et al. Effects of single nucleotide polymorphisms on treatment outcomes and toxicity in patients treated with sunitinib. Anticancer Res. 2013;33:4619–4626.
  • Fazio N, Martini JF, Croitoru AE, et al. Pharmacogenomic analyses of sunitinib in patients with pancreatic neuroendocrine tumors. Future Oncol. 2019;15:1997–2007.
  • Jimenez-Fonseca P, Martin MN, Carmona-Bayonas A, et al. Biomarkers and polymorphisms in pancreatic neuroendocrine tumors treated with sunitinib. Oncotarget. 2018;9:36894–36905.
  • Valle J, Bornath I, Rosbrook B, et al. Sunitinib in patients with pancreatic neuroendocrine tumors: update of safety data. Future Oncol. 2019;15:1219–1230.
  • Elgendy M, Abdel-Aziz AK, Renne SL, et al. Dual modulation of MCL-1 and mTOR determines the response to sunitinib. J Clin Invest. 2017;127:153–168.
  • Bannen LC, Chan DS, Forsyth TP, et al. Inventors; Exelixis, Inc., assignee. c-Met modulators and methods of use. United States patent US 7579473; 2009 Aug 25 [cited 2011 Aug 8]. Available from: http://patents.justia.com/2009/07579473.html.
  • Yakes FM, Chen J, Tan J, et al. Cabozantinib (XL184), a novel MET and VEGFR2 inhibitor, simultaneously suppresses metastasis, angiogenesis, and tumor growth. Mol Cancer Ther. 2011;10(12):2298–2308.
  • Reuther C, Heinzle V, Spampatti M, et al. Cabozantinib and tivantinib, but not INC280, induce antiproliferative and antimigratory effects in human neuroendocrine tumor cells in vitro: evidence for ‘off-target’ effects not mediated by c-Met inhibition. Neuroendocrinology. 2016;103(3–4):383–401.
  • Sennino B, Ishiguro-Oonuma T, Wei Y, et al. Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors. Cancer Discov. 2012;2:270–287.
  • Chan JA, Faris JE, Murphy JE, et al. Phase II trial of cabozantinib in patients with carcinoid and pancreatic neuroendocrine tumors (pNET). J Clin Oncol. 2017;35(4_suppl):228.
  • Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674.
  • Raimondo L, D’Amato V, Servetto A, et al. Everolimus induces Met inactivation by disrupting the FKBP12/Met complex. Oncotarget. 2016 28;7(26):40073–40084.
  • Gupta A, Jarzab B, Capdevila J, et al. Population pharmacokinetic analysis of lenvatinib in healthy subjects and patients with cancer. Br J Clin Pharmacol. 2016;81:1124–1133.
  • Hussein Z, Mizuo H, Hayato S, et al. Clinical pharmacokinetic and pharmacodynamic profile of lenvatinib, an orally active, small-molecule, multitargeted tyrosine kinase inhibitor. Eur J Drug Metab Pharmacokinet. 2017;42:903–914.
  • Cabanillas ME, Habra MA. Lenvatinib: role in thyroid cancer and other solid tumors. Cancer Treat Rev. 2016;42:47–55.
  • Matsui J, Yamamoto Y, Funahashi Y, et al. E7080, a novel inhibitor that targets multiple kinases, has potent antitumor activities against stem cell factor producing human small cell lung cancer H146, based on angiogenesis inhibition. Int J Cancer. 2008;122(3):664–671.
  • Tohyama O, Matsui J, Kodama K, et al. Antitumor activity of lenvatinib (e7080): an angiogenesis inhibitor that targets multiple receptor tyrosine kinases in preclinical human thyroid cancer models. J Thyroid Res. 2014;2014:638747.
  • Yamamoto Y, Matsui J, Matsushima T, et al. Lenvatinib, an angiogenesis inhibitor targeting VEGFR/FGFR, shows broad antitumor activity in human tumor xenograft models associated with microvessel density and pericyte coverage. Vasc Cell. 2014;6:18.
  • Yamada K, Yamamoto N, Yamada Y, et al. Phase I dose-escalation study and biomarker analysis of E7080 in patients with advanced solid tumors. Clin Cancer Res. 2011;17(8):2528–2537.
  • Capdevila J, Fazio N, Lopez C, et al. Efficacy of lenvatinib in patients with advanced pancreatic (panNETs) and gastrointestinal (giNETs) grade 1/2 (G1/G2) neuroendocrine tumors: results of the international phase II TALENT trial (GETNE 1509). Ann Oncol. 2018;29(Suppl8):Abstract1307O.
  • Capdevila J, Fazio N, López-López C, et al. Progression-free survival (PFS) and subgroups analyses of lenvatinib in patients (pts) with G1/G2 advanced pancreatic (panNETs) and gastrointestinal (giNETs) neuroendocrine tumors (NETs): updated results from the phase II TALENT trial (GETNE 1509). J Clin Oncol. 2019;37(4 suppl):332.
  • Matsuki M, Adachi Y, Ozawa Y, et al. Targeting of tumor growth and angiogenesis underlies the enhanced antitumor activity of lenvatinib in combination with everolimus. Cancer Sci. 2017;108(4):763–771.
  • Smith NR, Baker D, Farren M, et al. Tumor stromal architecture can define the intrinsic tumor response to VEGF-targeted therapy. Clin Cancer Res. 2013;19(24):6943–6956.
  • Capozzi M, De Divitiis C, Ottaiano A, et al. Lenvatinib, a molecule with versatile application: from preclinical evidence to future development in anti-cancer treatment. Cancer Manag Res. 2019;11:3847–3860.
  • Verheijen RB, Beijnen JH, Schellens JHM, et al. Clinical pharmacokinetics and pharmacodynamics of pazopanib: towards optimized dosing. Clin Pharmacokinet. 2017 Sep;56(9):987–997.
  • Kumar R, Knick VB, Rudolph SK, et al. Pharmacokinetic-pharmacodynamic correlation from mouse to human with pazopanib, a multikinase angiogenesis inhibitor with potent antitumor and antiangiogenic activity. Mol Cancer Ther. 2007;6(7):2012–2021.
  • Hashimoto K, Man S, Xu P, et al. Potent preclinical impact of metronomic low-dose oral topotecan combined with the antiangiogenic drug pazopanib for the treatment of ovarian cancer. Mol Cancer Ther. 2010;9(9):996–1006.
  • Li H, Wozniak A, Sciot R, et al. Pazopanib, a receptor tyrosine kinase inhibitor, suppresses tumor growth through angiogenesis in dedifferentiated liposarcoma xenograft models. Transl Oncol. 2014;7(7):665–671.
  • Zhu X-D, Zhang J-B, Fan P-L, et al. Antiangiogenic effects of pazopanib in xenograft hepatocellular carcinoma models: evaluation by quantitative contrast-enhanced ultrasonography. BMC Cancer. 2011;11(1):28.
  • Ahn HK, Choi JY, Kim KM, et al. Phase II study of pazopanib monotherapy in metastatic gastroenteropancreatic neuroendocrine tumours. Br J Cancer. 2013 Sep 17;109(6):1414–1419.
  • Phan AT, Halperin DM, Chan JA, et al. Pazopanib and depot octreotide in advanced, well-differentiated neuroendocrine tumours: a multicentre, single-group, phase 2 study. Lancet Oncol. 2015 Jun;16(6):695–703.
  • Grande E, Capdevila J, Castellano D, et al. Pazopanib in pretreated advanced neuroendocrine tumors: a phase II, open-label trial of the Spanish task force group for neuroendocrine tumors (GETNE). Ann Oncol. 2015 Sep;26(9):1987–1993.
  • Park C, Ha SY, Kim ST, et al. Identification of the BRAF V600E mutation in gastroenteropancreatic neuroendocrine tumors. Oncotarget. 2016 Jan 26;7(4):4024–4035.
  • Grillo F, Florio T, Ferraù F, et al. Emerging multitarget tyrosine kinase inhibitors in the treatment of neuroendocrine neoplasms. Endocr Relat Cancer. 2018;25(9):R453–R466.
  • Xu JM, Wang Y, Chen YL, et al. Sulfatinib, a novel kinase inhibitor, in patients with advanced solid tumors: results from a phase I study. Oncotarget. 2017;8(26):42076–42086.
  • Xu J, Li J, Bai C, et al. Surufatinib in advanced well-differentiated neuroendocrine tumors: a multicenter, single-arm, open-label, phase Ib/II trial. Clin Cancer Res. 2019;25(12):3486–3494.
  • Xua JM, Li J, Bai C. An open-label phase Ib/II study of sulfatinib in patients with advanced neuroendocrine tumors (NCT02267967). 14th Annual ENETS Conference, Barcelona, Spain; 2017 Mar 8-10.
  • Hutchison China MediTech (Chi-Med). Surufatinib phase III SANET-ep study has met its primary endpoint at interim analysis in advanced non-pancreatic neuroendocrine tumors in china and will stop early. (AIM/Nasdaq: HCM)Innovation Platform, Press Releases, RNS Announcements; 2019.
  • Manjulika D. Surufatinib in neuroendocrine tumours for the study by Xu. Lancet Oncol. 2019;20(4):e196.
  • Hu-Lowe DD, Zou HY, Grazzini ML, et al. Nonclinical antiangiogenesis and antitumor activities of axitinib (AG-013736), an oral, potent, and selective inhibitor of vascular endothelial growth factor receptor tyrosine kinases 1, 2, 3. Clin Cancer Res. 2008;14:7272–7283.
  • Kroll J, Waltenberger J. A novel function of VEGF receptor-2 (KDR): rapid release of nitric oxide in response to VEGF-A stimulation in endothelial cells. Biochem Biophys Res Commun. 1999;265:636–639.
  • Shiojima I, Walsh K. Role of Akt signaling in vascular homeostasis and angiogenesis. Circ Res. 2002;90:1243–1250.
  • Kelly RJ, Rixe O. Axitinib–a selective inhibitor of the vascular endothelial growth factor (VEGF) receptor. Target Oncol. 2009;4:297–305.
  • Escudier B, Gore M. Axitinib for the management of metastatic renal cell carcinoma. Drugs R D. 2011;11:113–126.
  • Wilmes LJ, Pallavicini MG, Fleming LM, et al. AG-013736, a novel inhibitor of VEGF receptor tyrosine kinases, inhibits breast cancer growth and decreases vascular permeability as detected by dynamic contrast-enhanced magnetic resonance imaging. Magn Reson Imaging. 2007;25:319–327.
  • Liu G, Rugo HS, Wilding G, et al. Dynamic contrast-enhanced magnetic resonance imaging as a pharmacodynamic measure of response after acute dosing of AG-013736, an oral angiogenesis inhibitor, in patients with advanced solid tumors: results from a phase I study. J Clin Oncol. 2005;23:5464–5473.
  • Gilbert JA, Adhikari LJ, Lloyd RV, et al. Molecular markers for novel therapeutic strategies in pancreatic endocrine tumors. Pancreas. 2013 Apr;42(3):411–421.
  • Wang Z, Chen H, Zhou N, et al. H727 multicellular spheroids and its resistance to antitumor drugs sunitinib and axitinib. J Nanosci Nanotechnol. 2018;18(12):8078–8084.
  • Strosberg J, Cives M, Hwang J, et al. A phase II study of axitinib in advanced neuroendocrine tumors. Endocr Related Cancer. 2016 May;23(5):411–418.
  • Rini BI, Escudier B, Tomczak P, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet. 2011 Dec 3;378(9807):1931–1939.
  • Loges S, Schmidt T, Carmeliet P. Mechanisms of resistance to anti-angiogenic therapy and development of third-generation anti-angiogenic drug candidates. Genes Cancer. 2010;1:12–25.
  • Casanovas O, Hicklin DJ, Bergers G, et al. Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell. 2005;8:299–309.
  • Van Belle E, Witzenbichler B, Chen D, et al. Potentiated angiogenic effect of scatter factor/hepatocyte growth factor via induction of vascular endothelial growth factor: the case for paracrine ampli?cation of angiogenesis. Circulation. 1998;97:381–390.
  • Rini BI, Plimack ER, Stus V, et al. Pembrolizumab plus Axitinib versus Sunitinib in advanced renal-cell carcinoma. New Engl J Med. 2019;380:1116–1127.

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.