Trastuzumab emtansine: a review of its use in patients with HER2-positive advanced breast cancer previously treated with trastuzumab-based therapy
- PMID: 24659374
- DOI: 10.1007/s40265-014-0201-0
Trastuzumab emtansine: a review of its use in patients with HER2-positive advanced breast cancer previously treated with trastuzumab-based therapy
Abstract
Trastuzumab emtansine (Kadcyla™) is an antibody-drug conjugate consisting of the humanized anti-human epidermal growth factor receptor (HER) 2 antibody trastuzumab covalently linked to the highly potent microtubule inhibitory drug DM1 (a cytotoxic derivative of maytansine) via a stable thioether linker. Intravenous trastuzumab emtansine was recently approved for use in patients with HER2-positive, unresectable, locally advanced (in the EU) or metastatic (in the USA and EU) breast cancer who had previously received trastuzumab and a taxane (separately or in combination), making it the first antibody-drug conjugate approved in this indication. This article reviews the efficacy and tolerability of trastuzumab emtansine in these patients and summarizes its pharmacology. In the well-designed EMILIA study, trastuzumab emtansine significantly prolonged progression-free survival and overall survival, relative to treatment with lapatinib plus capecitabine, in patients with HER2-positive, unresectable, locally advanced or metastatic breast cancer who were previously treated with trastuzumab and a taxane. Trastuzumab emtansine was generally well tolerated in this study, with <6% of patients discontinuing treatment because of adverse events. Based on its efficacy and favourable tolerability, the US National Comprehensive Cancer Network guidelines recommend trastuzumab emtansine as the preferred option in patients with HER2-positive metastatic breast cancer who have received previous trastuzumab-based therapy.
Similar articles
-
Trastuzumab emtansine versus capecitabine plus lapatinib in patients with previously treated HER2-positive advanced breast cancer (EMILIA): a descriptive analysis of final overall survival results from a randomised, open-label, phase 3 trial.Lancet Oncol. 2017 Jun;18(6):732-742. doi: 10.1016/S1470-2045(17)30312-1. Epub 2017 May 16. Lancet Oncol. 2017. PMID: 28526536 Free PMC article. Clinical Trial.
-
Trastuzumab emtansine for HER2-positive advanced breast cancer.N Engl J Med. 2012 Nov 8;367(19):1783-91. doi: 10.1056/NEJMoa1209124. Epub 2012 Oct 1. N Engl J Med. 2012. PMID: 23020162 Free PMC article. Clinical Trial.
-
A multicenter Phase II study evaluating the efficacy, safety and pharmacokinetics of trastuzumab emtansine in Japanese patients with heavily pretreated HER2-positive locally recurrent or metastatic breast cancer.Jpn J Clin Oncol. 2016 May;46(5):407-14. doi: 10.1093/jjco/hyw013. Epub 2016 Feb 24. Jpn J Clin Oncol. 2016. PMID: 26917603 Clinical Trial.
-
Trastuzumab emtansine: first global approval.Drugs. 2013 May;73(7):755-65. doi: 10.1007/s40265-013-0050-2. Drugs. 2013. PMID: 23620199 Review.
-
Ado-trastuzumab emtansine: a HER2-positive targeted antibody-drug conjugate.Ann Pharmacother. 2014 Nov;48(11):1484-93. doi: 10.1177/1060028014545354. Epub 2014 Jul 31. Ann Pharmacother. 2014. PMID: 25082874 Review.
Cited by
-
Molecular Mechanism of HER2 Rapid Internalization and Redirected Trafficking Induced by Anti-HER2 Biparatopic Antibody.Antibodies (Basel). 2020 Sep 18;9(3):49. doi: 10.3390/antib9030049. Antibodies (Basel). 2020. PMID: 32961882 Free PMC article.
-
Targeting Strategies for Renal Cell Carcinoma: From Renal Cancer Cells to Renal Cancer Stem Cells.Front Pharmacol. 2016 Nov 10;7:423. doi: 10.3389/fphar.2016.00423. eCollection 2016. Front Pharmacol. 2016. PMID: 27891093 Free PMC article. Review.
-
Challenges and opportunities in metastatic breast cancer treatments: Nano-drug combinations delivered preferentially to metastatic cells may enhance therapeutic response.Pharmacol Ther. 2022 Aug;236:108108. doi: 10.1016/j.pharmthera.2022.108108. Epub 2022 Jan 6. Pharmacol Ther. 2022. PMID: 34999182 Free PMC article. Review.
-
Simultaneous inhibition of deubiquitinating enzymes (DUBs) and autophagy synergistically kills breast cancer cells.Oncotarget. 2015 Feb 28;6(6):4159-70. doi: 10.18632/oncotarget.2904. Oncotarget. 2015. PMID: 25784654 Free PMC article.
-
Breaking the Bottleneck in Anticancer Drug Development: Efficient Utilization of Synthetic Biology.Molecules. 2022 Nov 2;27(21):7480. doi: 10.3390/molecules27217480. Molecules. 2022. PMID: 36364307 Free PMC article. Review.
References
Publication types
MeSH terms
Substances
LinkOut - more resources
Full Text Sources
Other Literature Sources
Medical
Research Materials
Miscellaneous