Skip to main content
Erschienen in: Drug Safety 6/2013

01.06.2013 | Review Article

Tyrosine Kinase Inhibitors: Their On-Target Toxicities as Potential Indicators of Efficacy

verfasst von: Devron R. Shah, Rashmi R. Shah, Joel Morganroth

Erschienen in: Drug Safety | Ausgabe 6/2013

Einloggen, um Zugang zu erhalten

Abstract

Tyrosine kinase inhibitors (TKIs) have revolutionized the treatment of certain forms of cancers, raising hopes for many patients with otherwise unresponsive tumours. While these agents are generally well tolerated, clinical experience with them has highlighted their unexpected association with serious toxic effects on various organs such as the heart, lungs, liver, kidneys, thyroid, skin, blood coagulation, gastrointestinal tract and nervous system. Many of these toxic effects result from downstream inhibition of vascular endothelial growth factor or epidermal growth factor signalling in cells of normal organs. Many of these undesirable effects such as hypertension, hypothyroidism, skin reactions and possibly proteinuria are on-target effects. Since tyrosine kinases are widely distributed with specific functional roles in different organs, this association is not too surprising. Various studies suggest that the development of these on-target effects indicates clinically desirable and effective inhibition of the corresponding ligand-mediated receptor linked with oncogenesis. This is reflected as improved efficacy in the subgroup of patients who develop these on-target adverse effects compared with those who do not. Inevitably, issues arise with respect to the regulatory assessment of efficacy and risk/benefit of the TKIs as well as the clinical approach to managing patients who develop these effects. Routine subgroup analysis of efficacy data from clinical trials (patients with and without on-target toxicity) may enable more effective clinical use of TKIs since (i) discontinuing or reducing the dose of the TKI has a negative impact if the tumour is TKI-responsive; and (ii) it is usually possible to manage these undesirable on-target effects with conventional clinical approaches. Prospective studies are needed to investigate this proposition further.
Literatur
1.
Zurück zum Zitat Krause DS, Van Etten RA. Tyrosine kinases as targets for cancer therapy. N Engl J Med. 2005;353(2):172–87.PubMedCrossRef Krause DS, Van Etten RA. Tyrosine kinases as targets for cancer therapy. N Engl J Med. 2005;353(2):172–87.PubMedCrossRef
2.
Zurück zum Zitat Chen MH, Kerkela R, Force T. Mechanisms of cardiomyopathy associated with tyrosine kinase inhibitor cancer therapeutics. Circulation. 2008;118(1):84–95.PubMedCrossRef Chen MH, Kerkela R, Force T. Mechanisms of cardiomyopathy associated with tyrosine kinase inhibitor cancer therapeutics. Circulation. 2008;118(1):84–95.PubMedCrossRef
3.
Zurück zum Zitat Shah RR, Morganroth J, Shah DR. Cardiovascular safety of tyrosine kinase inhibitors: with a special focus on cardiac repolarization (QT interval). Drug Saf. doi:10.1007/s40264-013-0047-5 Shah RR, Morganroth J, Shah DR. Cardiovascular safety of tyrosine kinase inhibitors: with a special focus on cardiac repolarization (QT interval). Drug Saf. doi:10.​1007/​s40264-013-0047-5
4.
Zurück zum Zitat Keefe D, Bowen J, Gibson R, et al. Noncardiac vascular toxicities of vascular endothelial growth factor inhibitors in advanced cancer: a review. Oncologist. 2011;16(4):432–44.PubMedCrossRef Keefe D, Bowen J, Gibson R, et al. Noncardiac vascular toxicities of vascular endothelial growth factor inhibitors in advanced cancer: a review. Oncologist. 2011;16(4):432–44.PubMedCrossRef
5.
Zurück zum Zitat Cook KM, Figg WD. Angiogenesis inhibitors: current strategies and future prospects. CA Cancer J Clin. 2010;60(4):222–43. Cook KM, Figg WD. Angiogenesis inhibitors: current strategies and future prospects. CA Cancer J Clin. 2010;60(4):222–43.
6.
Zurück zum Zitat Gotlink KJ, Verheul HMW. Anti-angiogenic tyrosine kinase inhibitors: what is their mechanism of action? Angiogenesis. 2010;13(1):1–14. Gotlink KJ, Verheul HMW. Anti-angiogenic tyrosine kinase inhibitors: what is their mechanism of action? Angiogenesis. 2010;13(1):1–14.
7.
Zurück zum Zitat Laurent-Puig P, Lievre A, Blons H. Mutations and response to epidermal growth factor receptor inhibitors. Clin Cancer Res. 2009;15(4):1133–9.PubMedCrossRef Laurent-Puig P, Lievre A, Blons H. Mutations and response to epidermal growth factor receptor inhibitors. Clin Cancer Res. 2009;15(4):1133–9.PubMedCrossRef
8.
Zurück zum Zitat Elice F, Rodeghiero F, Falanga A, et al. Thrombosis associated with angiogenesis inhibitors. Best Pract Res Clin Haematol. 2009;22(1):115–28.PubMedCrossRef Elice F, Rodeghiero F, Falanga A, et al. Thrombosis associated with angiogenesis inhibitors. Best Pract Res Clin Haematol. 2009;22(1):115–28.PubMedCrossRef
9.
Zurück zum Zitat Sonpavde G, Bellmunt J, Schutz F, et al. The double edged sword of bleeding and clotting from VEGF inhibition in renal cancer patients. Curr Oncol Rep. 2012;14(4):295–306.PubMedCrossRef Sonpavde G, Bellmunt J, Schutz F, et al. The double edged sword of bleeding and clotting from VEGF inhibition in renal cancer patients. Curr Oncol Rep. 2012;14(4):295–306.PubMedCrossRef
10.
Zurück zum Zitat Amir E, Seruga B, Martinez-Lopez J, et al. Oncogenic targets, magnitude of benefit, and market pricing of antineoplastic drugs. J Clin Oncol. 2011;29(18):2543–9.PubMedCrossRef Amir E, Seruga B, Martinez-Lopez J, et al. Oncogenic targets, magnitude of benefit, and market pricing of antineoplastic drugs. J Clin Oncol. 2011;29(18):2543–9.PubMedCrossRef
11.
Zurück zum Zitat van Cruijsen H, van der Veldt A, Hoekman K. Tyrosine kinase inhibitors of VEGF receptors: clinical issues and remaining questions. Front Biosci. 2009;14(1):2248–68.PubMedCrossRef van Cruijsen H, van der Veldt A, Hoekman K. Tyrosine kinase inhibitors of VEGF receptors: clinical issues and remaining questions. Front Biosci. 2009;14(1):2248–68.PubMedCrossRef
12.
Zurück zum Zitat Roodhart JM, Langenberg MH, Witteveen E, et al. The molecular basis of class side effects due to treatment with inhibitors of the VEGF/VEGFR pathway. Curr Clin Pharmacol. 2008;3(2):132–43.PubMedCrossRef Roodhart JM, Langenberg MH, Witteveen E, et al. The molecular basis of class side effects due to treatment with inhibitors of the VEGF/VEGFR pathway. Curr Clin Pharmacol. 2008;3(2):132–43.PubMedCrossRef
13.
Zurück zum Zitat Eaby B, Culkin A, Lacouture ME. An interdisciplinary consensus on managing skin reactions associated with human epidermal growth factor receptor inhibitors. Clin J Oncol Nurs. 2008;12(2):283–90.PubMedCrossRef Eaby B, Culkin A, Lacouture ME. An interdisciplinary consensus on managing skin reactions associated with human epidermal growth factor receptor inhibitors. Clin J Oncol Nurs. 2008;12(2):283–90.PubMedCrossRef
14.
Zurück zum Zitat Asnacios A, Naveau S, Perlemuter G. Gastrointestinal toxicities of novel agents in cancer therapy. Eur J Cancer. 2009;45(Suppl. 1):332–42.PubMedCrossRef Asnacios A, Naveau S, Perlemuter G. Gastrointestinal toxicities of novel agents in cancer therapy. Eur J Cancer. 2009;45(Suppl. 1):332–42.PubMedCrossRef
15.
Zurück zum Zitat Steeghs N, Gelderblom H, Roodt JO, et al. Hypertension and rarefaction during treatment with telatinib, a small molecule angiogenesis inhibitor. Clin Cancer Res. 2008;14(11):3470–6.PubMedCrossRef Steeghs N, Gelderblom H, Roodt JO, et al. Hypertension and rarefaction during treatment with telatinib, a small molecule angiogenesis inhibitor. Clin Cancer Res. 2008;14(11):3470–6.PubMedCrossRef
33.
Zurück zum Zitat Nazer B, Humphreys BD, Moslehi J. Effects of novel angiogenesis inhibitors for the treatment of cancer on the cardiovascular system: focus on hypertension. Circulation. 2011;124(15):1687–91.PubMedCrossRef Nazer B, Humphreys BD, Moslehi J. Effects of novel angiogenesis inhibitors for the treatment of cancer on the cardiovascular system: focus on hypertension. Circulation. 2011;124(15):1687–91.PubMedCrossRef
34.
Zurück zum Zitat Qi WX, Shen Z, Lin F, et al. Incidence and risk of hypertension with vandetanib in cancer patients: a systematic review and meta-analysis of clinical trials. Br J Clin Pharmacol. 2013;75(4):919–30.PubMedCrossRef Qi WX, Shen Z, Lin F, et al. Incidence and risk of hypertension with vandetanib in cancer patients: a systematic review and meta-analysis of clinical trials. Br J Clin Pharmacol. 2013;75(4):919–30.PubMedCrossRef
35.
Zurück zum Zitat Rini BI, Cohen DP, Lu DR, et al. Hypertension as a biomarker of efficacy in patients with metastatic renal cell carcinoma treated with sunitinib. J Natl Cancer Inst. 2011;103(9):763–73.PubMedCrossRef Rini BI, Cohen DP, Lu DR, et al. Hypertension as a biomarker of efficacy in patients with metastatic renal cell carcinoma treated with sunitinib. J Natl Cancer Inst. 2011;103(9):763–73.PubMedCrossRef
36.
Zurück zum Zitat George S, Reichardt P, Lechner T, et al. Hypertension as a potential biomarker of efficacy in patients with gastrointestinal stromal tumor treated with sunitinib. Ann Oncol. 2012;23(12):3180–7.PubMedCrossRef George S, Reichardt P, Lechner T, et al. Hypertension as a potential biomarker of efficacy in patients with gastrointestinal stromal tumor treated with sunitinib. Ann Oncol. 2012;23(12):3180–7.PubMedCrossRef
37.
Zurück zum Zitat Rini BI, Schiller JH, Fruehauf JP, et al. Diastolic blood pressure as a biomarker of axitinib efficacy in solid tumors. Clin Cancer Res. 2011;17(11):3841–9.PubMedCrossRef Rini BI, Schiller JH, Fruehauf JP, et al. Diastolic blood pressure as a biomarker of axitinib efficacy in solid tumors. Clin Cancer Res. 2011;17(11):3841–9.PubMedCrossRef
38.
Zurück zum Zitat Estfan B, Byrne M, Kim R. Sorafenib in advanced hepatocellular carcinoma: hypertension as a potential surrogate marker for efficacy. Am J Clin Oncol (Epub 2012 Apr 27). Estfan B, Byrne M, Kim R. Sorafenib in advanced hepatocellular carcinoma: hypertension as a potential surrogate marker for efficacy. Am J Clin Oncol (Epub 2012 Apr 27).
39.
Zurück zum Zitat Kim JJ, Vaziri SA, Rini BI, et al. Association of VEGF and VEGFR2 single nucleotide polymorphisms with hypertension and clinical outcome in metastatic clear cell renal cell carcinoma patients treated with sunitinib. Cancer. 2012;118(7):1946–54.PubMedCrossRef Kim JJ, Vaziri SA, Rini BI, et al. Association of VEGF and VEGFR2 single nucleotide polymorphisms with hypertension and clinical outcome in metastatic clear cell renal cell carcinoma patients treated with sunitinib. Cancer. 2012;118(7):1946–54.PubMedCrossRef
40.
Zurück zum Zitat Li XS, Wu X, Zhao PJ, et al. Efficacy and safety of sunitinib in the treatment of metastatic renal cell carcinoma. Chin Med J (Engl). 2011;124(18):2920–4. Li XS, Wu X, Zhao PJ, et al. Efficacy and safety of sunitinib in the treatment of metastatic renal cell carcinoma. Chin Med J (Engl). 2011;124(18):2920–4.
41.
Zurück zum Zitat Clemons J, Gao D, Naam M, et al. Thyroid dysfunction in patients treated with sunitinib or sorafenib. Clin Genitourin Cancer. 2012;10(4):225–31.PubMedCrossRef Clemons J, Gao D, Naam M, et al. Thyroid dysfunction in patients treated with sunitinib or sorafenib. Clin Genitourin Cancer. 2012;10(4):225–31.PubMedCrossRef
42.
Zurück zum Zitat Daimon M, Kato T, Kaino W, et al. Thyroid dysfunction in patients treated with tyrosine kinase inhibitors, sunitinib, sorafenib and axitinib, for metastatic renal cell carcinoma. Jpn J Clin Oncol. 2012;42(8):742–7.PubMedCrossRef Daimon M, Kato T, Kaino W, et al. Thyroid dysfunction in patients treated with tyrosine kinase inhibitors, sunitinib, sorafenib and axitinib, for metastatic renal cell carcinoma. Jpn J Clin Oncol. 2012;42(8):742–7.PubMedCrossRef
43.
Zurück zum Zitat Torino F, Corsello SM, Longo R, et al. Hypothyroidism related to tyrosine kinase inhibitors: an emerging toxic effect of targeted therapy. Nat Rev Clin Oncol. 2009;6(4):219–28.PubMedCrossRef Torino F, Corsello SM, Longo R, et al. Hypothyroidism related to tyrosine kinase inhibitors: an emerging toxic effect of targeted therapy. Nat Rev Clin Oncol. 2009;6(4):219–28.PubMedCrossRef
44.
Zurück zum Zitat Sakurai K, Fukazawa H, Arihara Z, et al. Sunitinib-induced thyrotoxicosis followed by persistent hypothyroidism with shrinkage of thyroid volume. Tohoku J Exp Med. 2010;222(1):39–44.PubMedCrossRef Sakurai K, Fukazawa H, Arihara Z, et al. Sunitinib-induced thyrotoxicosis followed by persistent hypothyroidism with shrinkage of thyroid volume. Tohoku J Exp Med. 2010;222(1):39–44.PubMedCrossRef
45.
Zurück zum Zitat Krouse RS, Royal RE, Heywood G, et al. Thyroid dysfunction in 281 patients with metastatic melanoma or renal carcinoma treated with interleukin-2 alone. J Immunother Emphasis Tumor Immunol. 1995;18(4):272–8.PubMedCrossRef Krouse RS, Royal RE, Heywood G, et al. Thyroid dysfunction in 281 patients with metastatic melanoma or renal carcinoma treated with interleukin-2 alone. J Immunother Emphasis Tumor Immunol. 1995;18(4):272–8.PubMedCrossRef
46.
Zurück zum Zitat Schwartzentruber DJ, White DE, Zweig MH, et al. Thyroid dysfunction associated with immunotherapy for patients with cancer. Cancer. 1991;68(11):2384–90.PubMedCrossRef Schwartzentruber DJ, White DE, Zweig MH, et al. Thyroid dysfunction associated with immunotherapy for patients with cancer. Cancer. 1991;68(11):2384–90.PubMedCrossRef
47.
Zurück zum Zitat Wong E, Rosen LS, Mulay M, et al. Sunitinib induces hypothyroidism in advanced cancer patients and may inhibit thyroid peroxidase activity. Thyroid. 2007;17(4):351–5.PubMedCrossRef Wong E, Rosen LS, Mulay M, et al. Sunitinib induces hypothyroidism in advanced cancer patients and may inhibit thyroid peroxidase activity. Thyroid. 2007;17(4):351–5.PubMedCrossRef
48.
Zurück zum Zitat Mannavola D, Coco P, Vannucchi G, et al. A novel tyrosine-kinase selective inhibitor, sunitinib, induces transient hypothyroidism by blocking iodine uptake. J Clin Endocrinol Metab. 2007;92(9):3531–4.PubMedCrossRef Mannavola D, Coco P, Vannucchi G, et al. A novel tyrosine-kinase selective inhibitor, sunitinib, induces transient hypothyroidism by blocking iodine uptake. J Clin Endocrinol Metab. 2007;92(9):3531–4.PubMedCrossRef
49.
Zurück zum Zitat Abdulrahman RM, Verloop H, Hoftijzer H, et al. Sorafenib-induced hypothyroidism is associated with increased type 3 deiodination. J Clin Endocrinol Metab. 2010;95(8):3758–62.PubMedCrossRef Abdulrahman RM, Verloop H, Hoftijzer H, et al. Sorafenib-induced hypothyroidism is associated with increased type 3 deiodination. J Clin Endocrinol Metab. 2010;95(8):3758–62.PubMedCrossRef
50.
Zurück zum Zitat Kappers MH, van Esch JH, Smedts FM, et al. Sunitinib-induced hypothyroidism is due to induction of type 3 deiodinase activity and thyroidal capillary regression. J Clin Endocrinol Metab. 2011;96(10):3087–94.PubMedCrossRef Kappers MH, van Esch JH, Smedts FM, et al. Sunitinib-induced hypothyroidism is due to induction of type 3 deiodinase activity and thyroidal capillary regression. J Clin Endocrinol Metab. 2011;96(10):3087–94.PubMedCrossRef
51.
Zurück zum Zitat Vesely D, Astil J, Lastuvka P, et al. Serum levels of IGF-I, HGF, TGFβ1, bFGF and VEGF in thyroid gland tumors. Physiol Res. 2004;53(1):83–9.PubMed Vesely D, Astil J, Lastuvka P, et al. Serum levels of IGF-I, HGF, TGFβ1, bFGF and VEGF in thyroid gland tumors. Physiol Res. 2004;53(1):83–9.PubMed
52.
Zurück zum Zitat Makita N, Miyakawa M, Fujita T, et al. Sunitinib induces hypothyroidism with a markedly reduced vascularity. Thyroid. 2010;20(3):323–6.PubMedCrossRef Makita N, Miyakawa M, Fujita T, et al. Sunitinib induces hypothyroidism with a markedly reduced vascularity. Thyroid. 2010;20(3):323–6.PubMedCrossRef
53.
Zurück zum Zitat Sato S, Muraishi K, Tani J, et al. Clinical characteristics of thyroid abnormalities induced by sunitinib treatment in Japanese patients with renal cell carcinoma. Endocr J. 2010;57(10):873–80.PubMedCrossRef Sato S, Muraishi K, Tani J, et al. Clinical characteristics of thyroid abnormalities induced by sunitinib treatment in Japanese patients with renal cell carcinoma. Endocr J. 2010;57(10):873–80.PubMedCrossRef
54.
Zurück zum Zitat Kitajima K, Takahashi S, Maeda T, et al. Thyroid size change by CT monitoring after sorafenib or sunitinib treatment in patients with renal cell carcinoma: comparison with thyroid function. Eur J Radiol. 2012;81(9):2060–5.PubMedCrossRef Kitajima K, Takahashi S, Maeda T, et al. Thyroid size change by CT monitoring after sorafenib or sunitinib treatment in patients with renal cell carcinoma: comparison with thyroid function. Eur J Radiol. 2012;81(9):2060–5.PubMedCrossRef
55.
Zurück zum Zitat Riesenbeck LM, Bierer S, Hoffmeister I, et al. Hypothyroidism correlates with a better prognosis in metastatic renal cancer patients treated with sorafenib or sunitinib. World J Urol. 2011;29(6):807–13.PubMedCrossRef Riesenbeck LM, Bierer S, Hoffmeister I, et al. Hypothyroidism correlates with a better prognosis in metastatic renal cancer patients treated with sorafenib or sunitinib. World J Urol. 2011;29(6):807–13.PubMedCrossRef
56.
Zurück zum Zitat Schmidinger M, Vogl UM, Bojic M, et al. Hypothyroidism in patients with renal cell carcinoma: blessing or curse? Cancer. 2011;117(3):534–44.PubMedCrossRef Schmidinger M, Vogl UM, Bojic M, et al. Hypothyroidism in patients with renal cell carcinoma: blessing or curse? Cancer. 2011;117(3):534–44.PubMedCrossRef
57.
Zurück zum Zitat Robinson ES, Matulonis UA, Ivy P, et al. Rapid development of hypertension and proteinuria with cediranib, an oral vascular endothelial growth factor receptor inhibitor. Clin J Am Soc Nephrol. 2010;5(3):477–83.PubMedCrossRef Robinson ES, Matulonis UA, Ivy P, et al. Rapid development of hypertension and proteinuria with cediranib, an oral vascular endothelial growth factor receptor inhibitor. Clin J Am Soc Nephrol. 2010;5(3):477–83.PubMedCrossRef
58.
Zurück zum Zitat Eskens FA, de Jonge MJ, Bhargava P, et al. Biologic and clinical activity of tivozanib (AV-951, KRN-951), a selective inhibitor of VEGF receptor-1, -2, and -3 tyrosine kinases, in a 4-week-on, 2-week-off schedule in patients with advanced solid tumors. Clin Cancer Res. 2011;17(22):7156–63.PubMedCrossRef Eskens FA, de Jonge MJ, Bhargava P, et al. Biologic and clinical activity of tivozanib (AV-951, KRN-951), a selective inhibitor of VEGF receptor-1, -2, and -3 tyrosine kinases, in a 4-week-on, 2-week-off schedule in patients with advanced solid tumors. Clin Cancer Res. 2011;17(22):7156–63.PubMedCrossRef
59.
Zurück zum Zitat Eremina V, Jefferson JA, Kowalewska J, et al. VEGF inhibition and renal thrombotic microangiopathy. N Engl J Med. 2008;358(11):1129–36.PubMedCrossRef Eremina V, Jefferson JA, Kowalewska J, et al. VEGF inhibition and renal thrombotic microangiopathy. N Engl J Med. 2008;358(11):1129–36.PubMedCrossRef
60.
Zurück zum Zitat Eremina V, Quaggin SE. Biology of anti-angiogenc therapy-induced thrombotic microangiopathy. Semin Nephrol. 2010;30(6):582–90.PubMedCrossRef Eremina V, Quaggin SE. Biology of anti-angiogenc therapy-induced thrombotic microangiopathy. Semin Nephrol. 2010;30(6):582–90.PubMedCrossRef
61.
Zurück zum Zitat Izzedine H, Massard C, Spano JP, et al. VEGF signalling inhibition-induced proteinuria: Mechanisms, significance and management. Eur J Cancer. 2010;46(2):439–48.PubMedCrossRef Izzedine H, Massard C, Spano JP, et al. VEGF signalling inhibition-induced proteinuria: Mechanisms, significance and management. Eur J Cancer. 2010;46(2):439–48.PubMedCrossRef
63.
Zurück zum Zitat Bertuccio C, Veron D, Aggarwal PK, et al. Vascular endothelial growth factor receptor 2 direct interaction with nephrin links VEGF-A signals to actin in kidney podocytes. J Biol Chem. 2011;286(46):39933–44.PubMedCrossRef Bertuccio C, Veron D, Aggarwal PK, et al. Vascular endothelial growth factor receptor 2 direct interaction with nephrin links VEGF-A signals to actin in kidney podocytes. J Biol Chem. 2011;286(46):39933–44.PubMedCrossRef
64.
Zurück zum Zitat Sugimoto H, Hamano Y, Charytan D, et al. Neutralization of circulating vascular endothelial growth factor (VEGF) by anti-VEGF antibodies and soluble VEGF receptor 1 (sFlt-1) induces proteinuria. J Biol Chem. 2003;278(15):12605–8.PubMedCrossRef Sugimoto H, Hamano Y, Charytan D, et al. Neutralization of circulating vascular endothelial growth factor (VEGF) by anti-VEGF antibodies and soluble VEGF receptor 1 (sFlt-1) induces proteinuria. J Biol Chem. 2003;278(15):12605–8.PubMedCrossRef
65.
Zurück zum Zitat Blanco S, Bonet J, López D, et al. ACE inhibitors improve nephrin expression in Zucker rats with glomerulosclerosis. Kidney Int Suppl. 2005;67(S93):S10-4. Blanco S, Bonet J, López D, et al. ACE inhibitors improve nephrin expression in Zucker rats with glomerulosclerosis. Kidney Int Suppl. 2005;67(S93):S10-4.
66.
Zurück zum Zitat Agabiti-Rosei E. Structural and functional changes of the microcirculation in hypertension: influence of pharmacological therapy. Drugs. 2003;63(Spec No 1):19–29. Agabiti-Rosei E. Structural and functional changes of the microcirculation in hypertension: influence of pharmacological therapy. Drugs. 2003;63(Spec No 1):19–29.
67.
Zurück zum Zitat Rosen AC, Wu S, Damse A, et al. Risk of rash in cancer patients treated with vandetanib: systematic review and meta-analysis. J Clin Endocrinol Metab. 2012;97(4):1125–33.PubMedCrossRef Rosen AC, Wu S, Damse A, et al. Risk of rash in cancer patients treated with vandetanib: systematic review and meta-analysis. J Clin Endocrinol Metab. 2012;97(4):1125–33.PubMedCrossRef
68.
Zurück zum Zitat Lacouture ME, Laabs SM, Koehler M, et al. Analysis of dermatologic events in patients with cancer treated with lapatinib. Breast Cancer Res Treat. 2009;114(3):485–93.PubMedCrossRef Lacouture ME, Laabs SM, Koehler M, et al. Analysis of dermatologic events in patients with cancer treated with lapatinib. Breast Cancer Res Treat. 2009;114(3):485–93.PubMedCrossRef
69.
Zurück zum Zitat Choi NM. Chemotherapy-induced iatrogenic injury of skin: new drugs and new concepts Clin Dermatol. 2011;29(6):587–601. Choi NM. Chemotherapy-induced iatrogenic injury of skin: new drugs and new concepts Clin Dermatol. 2011;29(6):587–601.
70.
Zurück zum Zitat Hirsh V. Managing treatment-related adverse events associated with EGFR tyrosine kinase inhibitors in advanced non-small-cell lung cancer. Curr Oncol. 2011;18(3):126–38.PubMedCrossRef Hirsh V. Managing treatment-related adverse events associated with EGFR tyrosine kinase inhibitors in advanced non-small-cell lung cancer. Curr Oncol. 2011;18(3):126–38.PubMedCrossRef
71.
Zurück zum Zitat Suzumura T, Kimura T, Kudoh S, et al. Reduced CYP2D6 function is associated with gefitinib-induced rash in patients with non-small cell lung cancer. BMC Cancer. 2012;4(12):568.CrossRef Suzumura T, Kimura T, Kudoh S, et al. Reduced CYP2D6 function is associated with gefitinib-induced rash in patients with non-small cell lung cancer. BMC Cancer. 2012;4(12):568.CrossRef
72.
Zurück zum Zitat Li J, Karlsson MO, Brahmer J, et al. CYP3A phenotyping approach to predict systemic exposure to EGFR tyrosine kinase inhibitors. J Natl Cancer Inst. 2006;98(23):1714–23.PubMedCrossRef Li J, Karlsson MO, Brahmer J, et al. CYP3A phenotyping approach to predict systemic exposure to EGFR tyrosine kinase inhibitors. J Natl Cancer Inst. 2006;98(23):1714–23.PubMedCrossRef
73.
Zurück zum Zitat Pérez-Soler R, Zou Y, Li T, et al. The phosphatase inhibitor menadione (vitamin K3) protects cells from EGFR inhibition by erlotinib and cetuximab. Clin Cancer Res. 2011;17(21):6766–77.PubMedCrossRef Pérez-Soler R, Zou Y, Li T, et al. The phosphatase inhibitor menadione (vitamin K3) protects cells from EGFR inhibition by erlotinib and cetuximab. Clin Cancer Res. 2011;17(21):6766–77.PubMedCrossRef
74.
Zurück zum Zitat Mitra SS, Simcock R. Erlotinib induced skin rash spares skin in previous radiotherapy field. J Clin Oncol. 2006;24(16):e28–9.PubMedCrossRef Mitra SS, Simcock R. Erlotinib induced skin rash spares skin in previous radiotherapy field. J Clin Oncol. 2006;24(16):e28–9.PubMedCrossRef
75.
Zurück zum Zitat Pérez-Soler R. Can rash associated with HER1/EGFR inhibition be used as a marker of treatment outcome? Oncology (Williston Park). 2003;17(11 Suppl. 12):23–8. Pérez-Soler R. Can rash associated with HER1/EGFR inhibition be used as a marker of treatment outcome? Oncology (Williston Park). 2003;17(11 Suppl. 12):23–8.
76.
Zurück zum Zitat Pérez-Soler R. Rash as a surrogate marker for efficacy of epidermal growth factor receptor inhibitors in lung cancer. Clin Lung Cancer. 2006;8(Suppl. 1):S7–14.PubMedCrossRef Pérez-Soler R. Rash as a surrogate marker for efficacy of epidermal growth factor receptor inhibitors in lung cancer. Clin Lung Cancer. 2006;8(Suppl. 1):S7–14.PubMedCrossRef
77.
Zurück zum Zitat Wacker B, Nagrani T, Weinberg J, et al. Correlation between development of rash and efficacy in patients treated with the epidermal growth factor receptor tyrosine kinase inhibitor erlotinib in two large phase III studies. Clin Cancer Res. 2007;13(13):3913–21.PubMedCrossRef Wacker B, Nagrani T, Weinberg J, et al. Correlation between development of rash and efficacy in patients treated with the epidermal growth factor receptor tyrosine kinase inhibitor erlotinib in two large phase III studies. Clin Cancer Res. 2007;13(13):3913–21.PubMedCrossRef
78.
Zurück zum Zitat Liu G, Gurubhagavatula S, Zhou W, et al. Epidermal growth factor receptor polymorphisms and clinical outcomes in non-small-cell lung cancer patients treated with gefitinib. Pharmacogenomics J. 2008;8(2):129–38.PubMedCrossRef Liu G, Gurubhagavatula S, Zhou W, et al. Epidermal growth factor receptor polymorphisms and clinical outcomes in non-small-cell lung cancer patients treated with gefitinib. Pharmacogenomics J. 2008;8(2):129–38.PubMedCrossRef
79.
Zurück zum Zitat Vincenzi B, Santini D, Russo A, et al. Early skin toxicity as a predictive factor for tumor control in hepatocellular carcinoma patients treated with sorafenib. Oncologist. 2010;15(1):85–92.PubMedCrossRef Vincenzi B, Santini D, Russo A, et al. Early skin toxicity as a predictive factor for tumor control in hepatocellular carcinoma patients treated with sorafenib. Oncologist. 2010;15(1):85–92.PubMedCrossRef
80.
Zurück zum Zitat Petrelli F, Borgonovo K, Cabiddu M, et al. Relationship between skin rash and outcome in non-small-cell lung cancer patients treated with anti-EGFR tyrosine kinase inhibitors: a literature-based meta-analysis of 24 trials. Lung Cancer. 2012;78(1):8–15.PubMedCrossRef Petrelli F, Borgonovo K, Cabiddu M, et al. Relationship between skin rash and outcome in non-small-cell lung cancer patients treated with anti-EGFR tyrosine kinase inhibitors: a literature-based meta-analysis of 24 trials. Lung Cancer. 2012;78(1):8–15.PubMedCrossRef
81.
Zurück zum Zitat Stepanski EJ, Reyes C, Walker MS, et al. The association of rash severity with overall survival: findings from patients receiving erlotinib for pancreatic cancer in the community setting. Pancreas. 2013;42(1):32–6.PubMedCrossRef Stepanski EJ, Reyes C, Walker MS, et al. The association of rash severity with overall survival: findings from patients receiving erlotinib for pancreatic cancer in the community setting. Pancreas. 2013;42(1):32–6.PubMedCrossRef
82.
Zurück zum Zitat Fiala O, Pesek M, Finek J, et al. Skin rash as useful marker of erlotinib efficacy in NSCLC and its impact on clinical practice. Neoplasma. 2013;60(1):26–32.PubMedCrossRef Fiala O, Pesek M, Finek J, et al. Skin rash as useful marker of erlotinib efficacy in NSCLC and its impact on clinical practice. Neoplasma. 2013;60(1):26–32.PubMedCrossRef
83.
Zurück zum Zitat Mita AC, Papadopoulos K, de Jonge MJA, et al. Erlotinib ‘dosing-to-rash’: a phase II intrapatient dose escalation and pharmacologic study of erlotinib in previously treated advanced non-small cell lung cancer. Br J Cancer. 2011;105(7):938–44. Mita AC, Papadopoulos K, de Jonge MJA, et al. Erlotinib ‘dosing-to-rash’: a phase II intrapatient dose escalation and pharmacologic study of erlotinib in previously treated advanced non-small cell lung cancer. Br J Cancer. 2011;105(7):938–44.
84.
Zurück zum Zitat Liu HB, Wu Y, Lv TF, et al. Skin rash could predict the response to EGFR tyrosine kinase inhibitor and the prognosis for patients with non-small cell lung cancer: a systematic review and meta-analysis. PLoS One. 2013;8(1):e55128.PubMedCrossRef Liu HB, Wu Y, Lv TF, et al. Skin rash could predict the response to EGFR tyrosine kinase inhibitor and the prognosis for patients with non-small cell lung cancer: a systematic review and meta-analysis. PLoS One. 2013;8(1):e55128.PubMedCrossRef
85.
Zurück zum Zitat Jonker DJ, O’Callaghan CJ, Karapetis CS, et al. Cetuximab for the treatment of colorectal cancer. N Engl J Med. 2007;357(20):2040–8.PubMedCrossRef Jonker DJ, O’Callaghan CJ, Karapetis CS, et al. Cetuximab for the treatment of colorectal cancer. N Engl J Med. 2007;357(20):2040–8.PubMedCrossRef
86.
Zurück zum Zitat Karapetis CS, Khambata-Ford S, Jonker DJ, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med. 2008;359(17):1757–65.PubMedCrossRef Karapetis CS, Khambata-Ford S, Jonker DJ, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med. 2008;359(17):1757–65.PubMedCrossRef
87.
Zurück zum Zitat Van Cutsem E, Tejpar S, Vanbeckevoort D, et al. Intrapatient cetuximab dose escalation in metastatic colorectal cancer according to the grade of early skin reactions: the randomized EVEREST study. J Clin Oncol. 2012;30(23):2861–8.PubMedCrossRef Van Cutsem E, Tejpar S, Vanbeckevoort D, et al. Intrapatient cetuximab dose escalation in metastatic colorectal cancer according to the grade of early skin reactions: the randomized EVEREST study. J Clin Oncol. 2012;30(23):2861–8.PubMedCrossRef
88.
Zurück zum Zitat Lièvre A, Bachet JB, Boige V, et al. KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab. J Clin Oncol. 2008;26(3):374–9.PubMedCrossRef Lièvre A, Bachet JB, Boige V, et al. KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab. J Clin Oncol. 2008;26(3):374–9.PubMedCrossRef
92.
Zurück zum Zitat Marshall JL. Maximum-tolerated dose, optimum biologic dose, or optimum clinical value: dosing determination of cancer therapies. J Clin Oncol. 2012;30(23):2815–6.PubMedCrossRef Marshall JL. Maximum-tolerated dose, optimum biologic dose, or optimum clinical value: dosing determination of cancer therapies. J Clin Oncol. 2012;30(23):2815–6.PubMedCrossRef
93.
Zurück zum Zitat Mukohara T, Nakajima H, Mukai H, et al. Effect of axitinib (AG-013736) on fatigue, thyroid-stimulating hormone, and biomarkers: a phase I study in Japanese patients. Cancer Sci. 2010;101(4):963–8.PubMedCrossRef Mukohara T, Nakajima H, Mukai H, et al. Effect of axitinib (AG-013736) on fatigue, thyroid-stimulating hormone, and biomarkers: a phase I study in Japanese patients. Cancer Sci. 2010;101(4):963–8.PubMedCrossRef
94.
Zurück zum Zitat Fujiwara Y, Kiyota N, Chayahara N, et al. Management of axitinib (AG-013736)-induced fatigue and thyroid dysfunction, and predictive biomarkers of axitinib exposure: results from phase I studies in Japanese patients. Invest New Drugs. 2012;30(3):1055–64.PubMedCrossRef Fujiwara Y, Kiyota N, Chayahara N, et al. Management of axitinib (AG-013736)-induced fatigue and thyroid dysfunction, and predictive biomarkers of axitinib exposure: results from phase I studies in Japanese patients. Invest New Drugs. 2012;30(3):1055–64.PubMedCrossRef
97.
Zurück zum Zitat Girardi F, Franceschi E, Brandes AA. Cardiovascular safety of VEGF-targeting therapies: current evidence and handling strategies. Oncologist. 2010;15(7):683–94.PubMedCrossRef Girardi F, Franceschi E, Brandes AA. Cardiovascular safety of VEGF-targeting therapies: current evidence and handling strategies. Oncologist. 2010;15(7):683–94.PubMedCrossRef
98.
Zurück zum Zitat Franklin PH, Banfor PN, Tapang P, et al. Effect of the multitargeted receptor tyrosine kinase inhibitor, ABT-869 [N-(4-(3-amino-1H-indazol-4-yl)phenyl)-N’-(2-fluoro-5-methylphenyl)urea], on blood pressure in conscious rats and mice: reversal with antihypertensive agents and effect on tumor growth inhibition. J Pharmacol Exp Ther. 2009;329(3):928–37.PubMedCrossRef Franklin PH, Banfor PN, Tapang P, et al. Effect of the multitargeted receptor tyrosine kinase inhibitor, ABT-869 [N-(4-(3-amino-1H-indazol-4-yl)phenyl)-N’-(2-fluoro-5-methylphenyl)urea], on blood pressure in conscious rats and mice: reversal with antihypertensive agents and effect on tumor growth inhibition. J Pharmacol Exp Ther. 2009;329(3):928–37.PubMedCrossRef
99.
Zurück zum Zitat Izzedine H, Ederhy S, Goldwasser F, et al. Management of hypertension in angiogenesis inhibitor-treated patients. Ann Oncol. 2009;20(5):807–15.PubMedCrossRef Izzedine H, Ederhy S, Goldwasser F, et al. Management of hypertension in angiogenesis inhibitor-treated patients. Ann Oncol. 2009;20(5):807–15.PubMedCrossRef
100.
Zurück zum Zitat Molteni A, Heffelfinger S, Moulder JE, et al. Potential deployment of angiotensin I converting enzyme inhibitors and of angiotensin II type 1 and type 2 receptor blockers in cancer chemotherapy. Anticancer Agents Med Chem. 2006;6(5):451–60.PubMedCrossRef Molteni A, Heffelfinger S, Moulder JE, et al. Potential deployment of angiotensin I converting enzyme inhibitors and of angiotensin II type 1 and type 2 receptor blockers in cancer chemotherapy. Anticancer Agents Med Chem. 2006;6(5):451–60.PubMedCrossRef
101.
Zurück zum Zitat Wolter P, Stefan C, Decallonne B, et al. The clinical implications of sunitinib-induced hypothyroidism: a prospective evaluation. Br J Cancer. 2008;99(3):448–54.PubMedCrossRef Wolter P, Stefan C, Decallonne B, et al. The clinical implications of sunitinib-induced hypothyroidism: a prospective evaluation. Br J Cancer. 2008;99(3):448–54.PubMedCrossRef
102.
Zurück zum Zitat Garfield DH, Wolter P, Schöffski P, et al. Documentation of thyroid function in clinical studies with sunitinib: why does it matter? J Clin Oncol. 2008;26(31):5131–2.PubMedCrossRef Garfield DH, Wolter P, Schöffski P, et al. Documentation of thyroid function in clinical studies with sunitinib: why does it matter? J Clin Oncol. 2008;26(31):5131–2.PubMedCrossRef
103.
Zurück zum Zitat Lynch TJ Jr, Kim ES, Eaby B, et al. Epidermal growth factor receptor inhibitor-associated cutaneous toxicities: an evolving paradigm in clinical management. Oncologist. 2007;12(5):610–21.PubMedCrossRef Lynch TJ Jr, Kim ES, Eaby B, et al. Epidermal growth factor receptor inhibitor-associated cutaneous toxicities: an evolving paradigm in clinical management. Oncologist. 2007;12(5):610–21.PubMedCrossRef
104.
Zurück zum Zitat Thatcher N, Nicolson M, Groves RW, for the UK Erlotinib Skin Toxicity Management Consensus Group, et al. Expert consensus on the management of erlotinib-associated cutaneous toxicity in the UK. Oncologist. 2009;14(8):840–7. Thatcher N, Nicolson M, Groves RW, for the UK Erlotinib Skin Toxicity Management Consensus Group, et al. Expert consensus on the management of erlotinib-associated cutaneous toxicity in the UK. Oncologist. 2009;14(8):840–7.
105.
Zurück zum Zitat Potthoff K, Hofheinz R, Hassel JC, et al. Interdisciplinary management of EGFR-inhibitor-induced skin reactions: a German expert opinion. Ann Oncol. 2011;22(3):524–35.PubMedCrossRef Potthoff K, Hofheinz R, Hassel JC, et al. Interdisciplinary management of EGFR-inhibitor-induced skin reactions: a German expert opinion. Ann Oncol. 2011;22(3):524–35.PubMedCrossRef
106.
Zurück zum Zitat Abdullah SE, Haigentz M Jr, Piperdi B. Dermatologic toxicities from monoclonal antibodies and tyrosine kinase inhibitors against EGFR: pathophysiology and management. Chemother Res Pract. 2012;2012:351210.PubMed Abdullah SE, Haigentz M Jr, Piperdi B. Dermatologic toxicities from monoclonal antibodies and tyrosine kinase inhibitors against EGFR: pathophysiology and management. Chemother Res Pract. 2012;2012:351210.PubMed
107.
Zurück zum Zitat Robert C, Sibaud V, Mateus C, et al. Advances in the management of cutaneous toxicities of targeted therapies. Semin Oncol. 2012;39(2):227–40.PubMedCrossRef Robert C, Sibaud V, Mateus C, et al. Advances in the management of cutaneous toxicities of targeted therapies. Semin Oncol. 2012;39(2):227–40.PubMedCrossRef
108.
Zurück zum Zitat Hassel JC, Kripp M, Al-Batran S, et al. Treatment of epidermal growth factor receptor antagonist-induced skin rash: results of a survey among German oncologists. Onkologie. 2010;33(3):94–8.PubMedCrossRef Hassel JC, Kripp M, Al-Batran S, et al. Treatment of epidermal growth factor receptor antagonist-induced skin rash: results of a survey among German oncologists. Onkologie. 2010;33(3):94–8.PubMedCrossRef
109.
Zurück zum Zitat Bidoli P, Cortinovis DL, Colombo I, et al. Isotretinoin plus clindamycin seem highly effective against severe erlotinib-induced skin rash in advanced non-small cell lung cancer. J Thorac Oncol. 2010;5(10):1662–3.PubMedCrossRef Bidoli P, Cortinovis DL, Colombo I, et al. Isotretinoin plus clindamycin seem highly effective against severe erlotinib-induced skin rash in advanced non-small cell lung cancer. J Thorac Oncol. 2010;5(10):1662–3.PubMedCrossRef
110.
Zurück zum Zitat Requena C, Llombart B, Sanmartín O. Acneiform eruptions induced by epidermal growth factor receptor inhibitors: treatment with oral isotretinoin. Cutis. 2012;90(2):77–80.PubMed Requena C, Llombart B, Sanmartín O. Acneiform eruptions induced by epidermal growth factor receptor inhibitors: treatment with oral isotretinoin. Cutis. 2012;90(2):77–80.PubMed
111.
Zurück zum Zitat Blanchetot C, Tertoolen LG, den Hertog J. Regulation of receptor protein-tyrosine phosphatase alpha by oxidative stress. EMBO J. 2002;21(4):493–503.PubMedCrossRef Blanchetot C, Tertoolen LG, den Hertog J. Regulation of receptor protein-tyrosine phosphatase alpha by oxidative stress. EMBO J. 2002;21(4):493–503.PubMedCrossRef
112.
Zurück zum Zitat Talon Therapeutics, Inc. Safety, tolerability and systemic absorption of menadione topical lotion for epidermal-growth-factor-receptor (EGFR) inhibitor-associated rash [ClinicalTrials.gov identifier NCT00656786]. US National Institutes of Health, ClinicalTrials.gov. Available from URL: http://www.clinicaltrials.gov. Accessed 29 Oct 2012. Talon Therapeutics, Inc. Safety, tolerability and systemic absorption of menadione topical lotion for epidermal-growth-factor-receptor (EGFR) inhibitor-associated rash [ClinicalTrials.gov identifier NCT00656786]. US National Institutes of Health, ClinicalTrials.gov. Available from URL: http://​www.​clinicaltrials.​gov. Accessed 29 Oct 2012.
113.
Zurück zum Zitat Mayo Clinic. Menadione topical lotion in treating skin discomfort and psychological distress in patients with cancer receiving panitumumab, erlotinib hydrochloride, or cetuximab [ClinicalTrials.gov identifier NCT01393821]. Available from URL: http://www.clinicaltrials.gov. Accessed 29 Oct 2012. Mayo Clinic. Menadione topical lotion in treating skin discomfort and psychological distress in patients with cancer receiving panitumumab, erlotinib hydrochloride, or cetuximab [ClinicalTrials.gov identifier NCT01393821]. Available from URL: http://​www.​clinicaltrials.​gov. Accessed 29 Oct 2012.
Metadaten
Titel
Tyrosine Kinase Inhibitors: Their On-Target Toxicities as Potential Indicators of Efficacy
verfasst von
Devron R. Shah
Rashmi R. Shah
Joel Morganroth
Publikationsdatum
01.06.2013
Verlag
Springer International Publishing AG
Erschienen in
Drug Safety / Ausgabe 6/2013
Print ISSN: 0114-5916
Elektronische ISSN: 1179-1942
DOI
https://doi.org/10.1007/s40264-013-0050-x

Weitere Artikel der Ausgabe 6/2013

Drug Safety 6/2013 Zur Ausgabe