Trastuzumab Is Not a Tyrosine Kinase Inhibitor Nishant Mohan and Wen Jin Wu

Total Page:16

File Type:pdf, Size:1020Kb

Trastuzumab Is Not a Tyrosine Kinase Inhibitor Nishant Mohan and Wen Jin Wu CORRESPONDENCE Nature Reviews Cardiology published online 29 September 2015; doi:10.1038/nrcardio.2015.135 Trastuzumab is not a tyrosine kinase inhibitor Nishant Mohan and Wen Jin Wu We read with great interest the Review by cancer cells is associated with its capacity to Correspondence to: W.J.W. Ewer, M. S. & Ewer, S. M. (Cardiotoxicity of induce ERBB2 phosphorylation at Tyr1248.7 [email protected] anticancer treatments. Nat. Rev. Cardiol. 12, Gijsen and co-workers also reported that Acknowledgements 1 547–558; 2015), because it is highly relevant trastuzumab induced ERBB2 tyrosine phos- We thank J. Zhou and T. S. Stantchev from the to our research. We appreciate the thorough phorylation,8 and together with another Center for Drug Evaluation and Research, U.S. Food and Drug Administration, for critical review review of the heart disorders and the asso- group observed that trastuzumab treatment of the manuscript. ciated molecular mechanisms elicited by induced paradoxical activation of mitogen- receptor tyrosine-protein kinase erbB‑2 activated protein kinase 1/3 in breast cancer Competing interests (ERBB2)-targeted agents in this article. How­ cells.8,9 In 2014, we demonstrated that trastu- N.M. and W.J.W are employees of the US Food and Drug Administration. This Correspondence reflects ever, we believe that the authors’ description zumab upregulated ERBB2 tyrosine kinase the views of the authors and should not be of trastuzumab as “the first mono­clonal anti- activity in trastuzumab-sensitive breast construed to represent FDA’s views or policies. body tyrosine kinase inhibitor” is conceptu- cancer cells, and that this upregulated a­ctivity 9 1. Ewer, M. S. & Ewer, S. M. Cardiotoxicity of ally incorrect and does not reflect the current contributed to ERBB2 phosphorylation. anticancer treatments. Nat. Rev. Cardiol. 12, state of literature in this field. Despite different effects on ERBB2 phos- 547–558 (2015). Tyrosine kinase inhibitors (TKIs), such phorylation, both lapatinib and trastuzumab 2. Medina, P. J. & Goodin, S. Lapatinib: a dual as lapatinib, inhibit the kinase activity of have the capacity to downregulate Akt activ- inhibitor of human epidermal growth factor receptor tyrosine kinases. Clin. Ther. 8, epidermal growth factor receptor (EGFR), ity and to inhibit growth of breast cancer cells. 1426–1447 (2008). and ERBB2 and ligand-induced receptor Based on our data, we postulate that binding 3. Mendelsohn, J. & Baselga, J. Epidermal growth phosphorylation by reversibly binding to the of trastuzumab to ERBB2 could induce struc- factor receptor targeting in cancer. 2 Semin. Oncol. 33, 369–385 (2006). ATP-binding site of the kinase. Cetuximab, tural changes in the ERBB2 cytoplasmic 4. Mendelsohn, J. & Baselga, J. The EGF receptor a therapeutic monoclonal antibody directed kinase domain, leading to allosteric activa- family as targets for cancer therapy. Oncogene against the extracellular domain of EGFR, tion of ERBB2 kinase and trans­phosphory­ 19, 6550–6565 (2000). 9,10 5. Hudis, C. A. Trastuzumab—mechanism of acts as a TKI to block the activation of EGFR l­ation of ERBB2. Trastuzumab-induced action and use in clinical practice. N. Engl. J. tyrosine kinase by competing for ligand ERBB2 phosphorylation provides docking Med. 357, 39–51 (2007). binding.3,4 Trastuzumab is a therapeutic sites for downstream signalling molecules, 6. Scott, G. K. et al. p185HER2 signal monoclonal antibody directed against the which are different from those induced by transduction in breast cancer cells. J. Biol. 5 Chem. 266, 14300–14305 (1991). extracellular domain of ERBB2. However, a ligand (such as epidermal growth factor), 7. Diermeier, S. et al. Epidermal growth factor the effects of trastuzumab on ERBB2 kinase in that they recruit signalling molecules such receptor coexpression modulates susceptibility activity are different from those of TKIs and as Csk-homologous kinase, a non-receptor to Herceptin in HER2/neu overexpressing breast cancer cells via specific erbB-receptor cetuximab. An anti-ERBB2 mono­clonal tyrosine kinase. Negative signals downstream interaction and activation. Exp. Cell. Res. 304, antibody 4D5, which was later human- of ERBB2 subsequently contribute to growth 604–619 (2005). ized to generate trastuzumab, was shown inhibition of ERBB2-positive breast cancer 8. Gijsen, M. et al. HER2 phosphorylation is to exhibit agonistic signalling, resulting in cells.9,10 Taken together, these studies provide maintained by a PKB negative feedback loop in response to anti-HER2 herceptin in breast increased ERBB2 phosphorylation that cor- evidence that trastuzumab does not function cancer. PLoS Biol. 8, e1000563 (2010). related with its inhibitory effect on ERBB2- as a TKI. 9. Dokmanovic, M. et al. Trastuzumab-induced overexpressing breast cancer cells.6 Growing recruitment of Csk-homologous kinase (CHK) to ErbB2 receptor is associated with Division of Biotechnology Research and evidence indicates that targeting ERBB2 ErbB2‑Y1248 phosphorylation and ErbB2 using trastuzumab increases ERBB2 tyrosine Review I, Office of Biotechnology Products, degradation to mediate cell growth inhibition. phosphorylation in trastuzumab-sensitive Office of Pharmaceutical Quality, Center Cancer Biol. Ther. 8, 1029–1041 (2014). 7–9 for Drug Evaluation and Research, U.S. Food 10. Dokmanovic, M. & Wu, W. J. Trastuzumab- breast cancer cells. Diemeier and col- and Drug Administration, 10903 New induced HER2 phosphorylation: exploring the leagues reported that the inhibitory effect of Hampshire Avenue, Silver Spring, MD 20993, mechanisms and implications. Receptors Clin. trastuzumab on trastuzumab-sensitive breast USA (N.M., W.J.W.). Investig. 1, e340 (2014). NATURE REVIEWS | CARDIOLOGY www.nature.com/nrcardio © 2015 Macmillan Publishers Limited. All rights reserved.
Recommended publications
  • Pertuzumab and Trastuzumab: the Rationale Way to Synergy
    Anais da Academia Brasileira de Ciências (2016) 88(1 Suppl.): 565-577 (Annals of the Brazilian Academy of Sciences) Printed version ISSN 0001-3765 / Online version ISSN 1678-2690 http://dx.doi.org/10.1590/0001-3765201620150178 www.scielo.br/aabc Pertuzumab and trastuzumab: the rationale way to synergy SANDRINE RICHARD1, FRÉDÉRIC SELLE1, JEAN-PIERRE LOTZ1,2, AHMED KHALIL1, JOSEPH GLIGOROV1,2 and DANIELE G. SOARES1 1Medical Oncology Department, APREC (Alliance Pour la Recherche En Cancérologie), Tenon Hospital (Hôpitaux Universitaires de l’Est-Parisien, AP-HP), rue de la Chine, 75020 Paris, France 2Institut Universitaire de Cancérologie Université Pierre et Marie Curie (IUC-UPMC Univ Paris 06), Sorbonne Universités, 4 place Jussieu, 75005 Paris, France Manuscript received on March 13, 2015; accepted for publication on May 5, 2015 ABSTRACT It has now been 15 years since the HER2-targeted monoclonal antibody trastuzumab was introduced in clinical and revolutionized the treatment of HER2-positive breast cancer patients. Despite this achievement, most patients with HER2-positive metastatic breast cancer still show progression of their disease, highlighting the need for new therapies. The continuous interest in novel targeted agents led to the development of pertuzumab, the first in a new class of agents, the HER dimerization inhibitors. Pertuzumab is a novel recombinant humanized antibody directed against extracellular domain II of HER2 protein that is required for the heterodimerization of HER2 with other HER receptors, leading to the activation of downstream signalling pathways. Pertuzumab combined with trastuzumab plus docetaxel was approved for the first-line treatment of patients with HER2-positive metastatic breast cancer and is currently used as a standard of care in this indication.
    [Show full text]
  • Tyrosine Kinase – Role and Significance in Cancer
    Int. J. Med. Sci. 2004 1(2): 101-115 101 International Journal of Medical Sciences ISSN 1449-1907 www.medsci.org 2004 1(2):101-115 ©2004 Ivyspring International Publisher. All rights reserved Review Tyrosine kinase – Role and significance in Cancer Received: 2004.3.30 Accepted: 2004.5.15 Manash K. Paul and Anup K. Mukhopadhyay Published:2004.6.01 Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S Nagar, Mohali, Punjab, India-160062 Abstract Tyrosine kinases are important mediators of the signaling cascade, determining key roles in diverse biological processes like growth, differentiation, metabolism and apoptosis in response to external and internal stimuli. Recent advances have implicated the role of tyrosine kinases in the pathophysiology of cancer. Though their activity is tightly regulated in normal cells, they may acquire transforming functions due to mutation(s), overexpression and autocrine paracrine stimulation, leading to malignancy. Constitutive oncogenic activation in cancer cells can be blocked by selective tyrosine kinase inhibitors and thus considered as a promising approach for innovative genome based therapeutics. The modes of oncogenic activation and the different approaches for tyrosine kinase inhibition, like small molecule inhibitors, monoclonal antibodies, heat shock proteins, immunoconjugates, antisense and peptide drugs are reviewed in light of the important molecules. As angiogenesis is a major event in cancer growth and proliferation, tyrosine kinase inhibitors as a target for anti-angiogenesis can be aptly applied as a new mode of cancer therapy. The review concludes with a discussion on the application of modern techniques and knowledge of the kinome as means to gear up the tyrosine kinase drug discovery process.
    [Show full text]
  • Trastuzumab, Biosimilars, Trastuzumab-Hyaluronidase
    Clinical Policy: Trastuzumab, Biosimilars, Trastuzumab-Hyaluronidase Reference Number: CP.PHAR.228 Effective Date: 06.01.16 Last Review Date: 05.20 Coding Implications Line of Business: Commercial, HIM, Medicaid Revision Log See Important Reminder at the end of this policy for important regulatory and legal information. Description Trastuzumab (Herceptin®) is a human epidermal growth factor receptor 2 (HER2)/neu receptor antagonist. Trastuzumab-dkst (Ogivri™), trastuzumab-pkrb (Herzuma®), trastuzumab-dttb (Ontruzant®), trastuzumab-qyyp (Trazimera™), and trastuzumab-anns (Kanjinti™) are Herceptin biosimilars. Trastuzumab-hyaluronidase-oysk (Herceptin Hylecta™) is a combination of trastuzumab and hyaluronidase, an endoglycosidase. FDA Approved Indication(s) Indications* Description Herceptin, Herzuma Herceptin Ogivri, Hylecta Ontruzant, Trazimera, Kanjinti Adjuvant For adjuvant As part of a treatment X X X breast cancer treatment of regimen consisting of HER2- doxorubicin, overexpressing cyclophosphamide, node positive and either paclitaxel or node or docetaxel negative As part of a treatment X X X (ER/PR regimen with negative or docetaxel and with one high carboplatin risk feature) As a single agent X X X breast cancer: following multi- modality anthracycline based therapy Metastatic In combination with paclitaxel for first- X X X breast cancer line treatment of HER2-overexpressing metastatic breast cancer As a single agent for treatment of X X X HER2-overexpressing breast cancer in patients who have received one or more Page 1 of 13
    [Show full text]
  • Ephb3 Suppresses Non-Small-Cell Lung Cancer Metastasis Via a PP2A/RACK1/Akt Signalling Complex
    ARTICLE Received 7 Nov 2011 | Accepted 11 Jan 2012 | Published 7 Feb 2012 DOI: 10.1038/ncomms1675 EphB3 suppresses non-small-cell lung cancer metastasis via a PP2A/RACK1/Akt signalling complex Guo Li1, Xiao-Dan Ji1, Hong Gao1, Jiang-Sha Zhao1, Jun-Feng Xu1, Zhi-Jian Sun1, Yue-Zhen Deng1, Shuo Shi1, Yu-Xiong Feng1, Yin-Qiu Zhu1, Tao Wang2, Jing-Jing Li1 & Dong Xie1 Eph receptors are implicated in regulating the malignant progression of cancer. Here we find that despite overexpression of EphB3 in human non-small-cell lung cancer, as reported previously, the expression of its cognate ligands, either ephrin-B1 or ephrin-B2, is significantly downregulated, leading to reduced tyrosine phosphorylation of EphB3. Forced activation of EphB3 kinase in EphB3-overexpressing non-small-cell lung cancer cells inhibits cell migratory capability in vitro as well as metastatic seeding in vivo. Furthermore, we identify a novel EphB3-binding protein, the receptor for activated C-kinase 1, which mediates the assembly of a ternary signal complex comprising protein phosphatase 2A, Akt and itself in response to EphB3 activation, leading to reduced Akt phosphorylation and subsequent inhibition of cell migration. Our study reveals a novel tumour-suppressive signalling pathway associated with kinase-activated EphB3 in non-small-cell lung cancer, and provides a potential therapeutic strategy by activating EphB3 signalling, thus inhibiting tumour metastasis. 1 Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Graduate School of Chinese Academy of Sciences, Shanghai 200031, China. 2 The Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai 200433, China.
    [Show full text]
  • Erbb3 Is Involved in Activation of Phosphatidylinositol 3-Kinase by Epidermal Growth Factor STEPHEN P
    MOLECULAR AND CELLULAR BIOLOGY, June 1994, p. 3550-3558 Vol. 14, No. 6 0270-7306/94/$04.00+0 Copyright C 1994, American Society for Microbiology ErbB3 Is Involved in Activation of Phosphatidylinositol 3-Kinase by Epidermal Growth Factor STEPHEN P. SOLTOFF,l* KERMIT L. CARRAWAY III,1 S. A. PRIGENT,2 W. G. GULLICK,2 AND LEWIS C. CANTLEY' Division of Signal Transduction, Department ofMedicine, Beth Israel Hospital, Boston, Massachusetts 02115,1 and Molecular Oncology Laboratory, ICRF Oncology Group, Hammersmith Hospital, London W12 OHS, United Kingdom2 Received 11 October 1993/Returned for modification 11 November 1993/Accepted 24 February 1994 Conflicting results concerning the ability of the epidermal growth factor (EGF) receptor to associate with and/or activate phosphatidylinositol (Ptdlns) 3-kinase have been published. Despite the ability of EGF to stimulate the production of Ptdlns 3-kinase products and to cause the appearance of PtdIns 3-kinase activity in antiphosphotyrosine immunoprecipitates in several cell lines, we did not detect EGF-stimulated Ptdlns 3-kinase activity in anti-EGF receptor immunoprecipitates. This result is consistent with the lack of a phosphorylated Tyr-X-X-Met motif, the p85 Src homology 2 (SH2) domain recognition sequence, in this receptor sequence. The EGF receptor homolog, ErbB2 protein, also lacks this motif. However, the ErbB3 protein has seven repeats of the Tyr-X-X-Met motif in the carboxy-terminal unique domain. Here we show that in A431 cells, which express both the EGF receptor and ErbB3, Ptdlns 3-kinase coprecipitates with the ErbB3 protein (pl80eR3) in response to EGF. p180B3 is also shown to be tyrosine phosphorylated in response to EGF.
    [Show full text]
  • Trastuzumab Products
    1 Trastuzumab Products: Trastuzumab (Herceptin®), Trastuzumab-anns (Kanjinti®), Trastuzumab-dkst (Ogivri®), Trastuzumab-dttb (Ontruzant®), Trastuzumab-pkrb (Herzuma®), Trastuzumab-qyyp (Trazimera®) Prior Authorization Drug Coverage Policy Effective Date: 11/9/2020 Revision Date: n/a Review Date: 10/27/2020 Lines of Business: Commercial Policy type: Prior Authorization This Drug Coverage Policy provides parameters for the coverage of trastuzumab products, which includes Trastuzumab (Herceptin®), Trastuzumab-anns (Kanjinti ®), Trastuzumab-dkst (Ogivri ®), Trastuzumab-dttb (Ontruzant®), Trastuzumab-pakrb (Herzuma®), and Trastuzumab-qyyp (Trazimera ®). Consideration of medically necessary indications are based upon U.S. Food and Drug Administration (FDA) indications, recommended uses within the Centers of Medicare & Medicaid Services (CMS) five recognized compendia, including the National Comprehensive Cancer Network (NCCN) Drugs & Biologics Compendium (NCCN Category 1 or 2A recommendations), and peer-reviewed scientific literature eligible for coverage according to the CMS, Medicare Benefit Policy Manual, Chapter 15, section 50.4.5 titled, “Off-Label Use of Anti- Cancer Drugs and Biologics.” This policy evaluates whether the drug therapy is proven to be effective based on published evidence-based medicine. Drug Description 1 The human epidermal growth factor receptor 2 (HER2) proto-oncogene encodes a transmembrane receptor protein of 185 kDa, which is structurally related to the epidermal growth factor receptor. Trastuzumab products have been shown, in both in vitro assays and in animals, to inhibit the proliferation of human tumor cells that overexpress HER2. Trastuzumab products are mediators of antibody-dependent cellular cytotoxicity (ADCC). In vitro , trastuzumab product mediated ADCC has been shown to be preferentially exerted on HER2 overexpressing cancer cells compared with cancer cells that do not overexpress HER2.
    [Show full text]
  • Human Ephb3 Antibody Antigen Affinity-Purified Polyclonal Sheep Igg Catalog Number: AF5667
    Human EphB3 Antibody Antigen Affinity-purified Polyclonal Sheep IgG Catalog Number: AF5667 DESCRIPTION Species Reactivity Human Specificity Detects human EphB3 in direct ELISAs and Western blots. In direct ELISAs, approximately 3% cross­reactivity with recombinant mouse EphB3 is observed, and less than 1% cross­reactivity with recombinant rat EphB1, recombinant human (rh) EphB2 and rhEphB4 is observed. Source Polyclonal Sheep IgG Purification Antigen Affinity­purified Immunogen Mouse myeloma cell line NS0­derived recombinant human EphB3 Leu38­Ala550 Accession # P54753 Formulation Lyophilized from a 0.2 μm filtered solution in PBS with Trehalose. See Certificate of Analysis for details. *Small pack size (­SP) is supplied either lyophilized or as a 0.2 μm filtered solution in PBS. APPLICATIONS Please Note: Optimal dilutions should be determined by each laboratory for each application. General Protocols are available in the Technical Information section on our website. Recommended Sample Concentration Western Blot 1 µg/mL See Below DATA Western Blot Detection of Human EphB3 by Western Blot. Western blot shows lysates of SH­SY5Y human neuroblastoma cell line. PVDF Membrane was probed with 1 µg/mL of Sheep Anti­Human EphB3 Antigen Affinity­ purified Polyclonal Antibody (Catalog # AF5667) followed by HRP­ conjugated Anti­Sheep IgG Secondary Antibody (Catalog # HAF016). A specific band was detected for EphB3 at approximately 110 kDa (as indicated). This experiment was conducted under reducing conditions and using Immunoblot Buffer Group 8. PREPARATION AND STORAGE Reconstitution Sterile PBS to a final concentration of 0.2 mg/mL. Shipping The product is shipped at ambient temperature. Upon receipt, store it immediately at the temperature recommended below.
    [Show full text]
  • PDGFRB FISH for Gleevec Eligibility in Myelodysplastic Syndrome/Myeloproliferative Disease (MDS/MPD)
    PDGFRB FISH PRODUCT DATASHEET Proprietary Name: PDGFRB FISH for Gleevec Eligibility in Myelodysplastic Syndrome/Myeloproliferative Disease (MDS/MPD) Established Name: PDGFRB FISH for Gleevec in MDS/MPD INTENDED USE Humanitarian Device. Authorized by Federal law for use in the qualitative detection of PDGFRB gene rearrangement in patients with MDS/MPD. The effectiveness of this device for this use has not been demonstrated. Caution: Federal Law restricts this device to sale by or on the order of a licensed practitioner. PDGFRB FISH for Gleevec Eligibility in Myelodysplastic Syndrome/Myeloproliferative Disease (MDS/MPD) is an in vitro diagnostic test intended for the qualitative detection of PDGFRB gene rearrangement from fresh bone marrow samples of patients with MDS/MPD with a high index of suspicion based on karyotyping showing a 5q31~33 anomaly. The PDGFRB FISH assay is indicated as an aid in the selection of MDS/MPD patients for whom Gleevec® (imatinib mesylate) treatment is being considered. This assay is for professional use only and is to be performed at a single laboratory site. SUMMARY AND EXPLANATION OF THE TEST The PDGFRB FISH assay detects rearrangement of the PDGFRB locus at chromosome 5q31~33 in adult patients with myelodysplastic syndrome/myeloproliferative disease (MDS/MPD). The PDGFRB gene encodes a cell surface receptor tyrosine kinase that upon activation stimulates mesenchymal cell division. Rearrangement of PDGFRB has been demonstrated by classical cytogenetic analysis in an exceedingly small fraction of MDS/MPD patients (~1%), usually cases with a clinical phenotype of chronic myelomonocytic leukemia (CMML). In particular, these patients harbor a t(5:12)(q31;p12) translocation involving the PDGFRB and ETV6 genes.
    [Show full text]
  • Antibody–Drug Conjugates: the Last Decade
    pharmaceuticals Review Antibody–Drug Conjugates: The Last Decade Nicolas Joubert 1,* , Alain Beck 2 , Charles Dumontet 3,4 and Caroline Denevault-Sabourin 1 1 GICC EA7501, Equipe IMT, Université de Tours, UFR des Sciences Pharmaceutiques, 31 Avenue Monge, 37200 Tours, France; [email protected] 2 Institut de Recherche Pierre Fabre, Centre d’Immunologie Pierre Fabre, 5 Avenue Napoléon III, 74160 Saint Julien en Genevois, France; [email protected] 3 Cancer Research Center of Lyon (CRCL), INSERM, 1052/CNRS 5286/UCBL, 69000 Lyon, France; [email protected] 4 Hospices Civils de Lyon, 69000 Lyon, France * Correspondence: [email protected] Received: 17 August 2020; Accepted: 10 September 2020; Published: 14 September 2020 Abstract: An armed antibody (antibody–drug conjugate or ADC) is a vectorized chemotherapy, which results from the grafting of a cytotoxic agent onto a monoclonal antibody via a judiciously constructed spacer arm. ADCs have made considerable progress in 10 years. While in 2009 only gemtuzumab ozogamicin (Mylotarg®) was used clinically, in 2020, 9 Food and Drug Administration (FDA)-approved ADCs are available, and more than 80 others are in active clinical studies. This review will focus on FDA-approved and late-stage ADCs, their limitations including their toxicity and associated resistance mechanisms, as well as new emerging strategies to address these issues and attempt to widen their therapeutic window. Finally, we will discuss their combination with conventional chemotherapy or checkpoint inhibitors, and their design for applications beyond oncology, to make ADCs the magic bullet that Paul Ehrlich dreamed of. Keywords: antibody–drug conjugate; ADC; bioconjugation; linker; payload; cancer; resistance; combination therapies 1.
    [Show full text]
  • Targeting the Function of the HER2 Oncogene in Human Cancer Therapeutics
    Oncogene (2007) 26, 6577–6592 & 2007 Nature Publishing Group All rights reserved 0950-9232/07 $30.00 www.nature.com/onc REVIEW Targeting the function of the HER2 oncogene in human cancer therapeutics MM Moasser Department of Medicine, Comprehensive Cancer Center, University of California, San Francisco, CA, USA The year 2007 marks exactly two decades since human HER3 (erbB3) and HER4 (erbB4). The importance of epidermal growth factor receptor-2 (HER2) was func- HER2 in cancer was realized in the early 1980s when a tionally implicated in the pathogenesis of human breast mutationally activated form of its rodent homolog neu cancer (Slamon et al., 1987). This finding established the was identified in a search for oncogenes in a carcinogen- HER2 oncogene hypothesis for the development of some induced rat tumorigenesis model(Shih et al., 1981). Its human cancers. An abundance of experimental evidence human homologue, HER2 was simultaneously cloned compiled over the past two decades now solidly supports and found to be amplified in a breast cancer cell line the HER2 oncogene hypothesis. A direct consequence (King et al., 1985). The relevance of HER2 to human of this hypothesis was the promise that inhibitors of cancer was established when it was discovered that oncogenic HER2 would be highly effective treatments for approximately 25–30% of breast cancers have amplifi- HER2-driven cancers. This treatment hypothesis has led cation and overexpression of HER2 and these cancers to the development and widespread use of anti-HER2 have worse biologic behavior and prognosis (Slamon antibodies (trastuzumab) in clinical management resulting et al., 1989).
    [Show full text]
  • EGFR and HER-2 Antagonists in Breast Cancer
    ANTICANCER RESEARCH 27: 1285-1294 (2007) Review EGFR and HER-2 Antagonists in Breast Cancer NORMA O’DONOVAN1 and JOHN CROWN1,2 1National Institute for Cellular Biotechnology, Dublin City University, Dublin 9; 2Department of Medical Oncology, St. Vincent’s University Hospital, Dublin 4, Ireland Abstract. Both HER-2 and EGFR are expressed in breast tumorigenesis was obtained using transgenic mice expressing cancer and are implicated in its development and progression. an activated form of the HER-2/neu oncogene in the The discovery of the association between HER-2 gene mammary epithelium, which resulted in rapid induction of amplification and poor prognosis in breast cancer led to the mammary tumours in 100% of female transgenic carriers (4). development of HER-2 targeted therapies. Trastuzumab, a Recent microarray analysis of breast tumours has monoclonal antibody to HER-2, has significantly improved the identified four distinct sub-types of tumours (5, 6). The sub- prognosis for HER-2-positive breast cancer patients. It is now types are classified as (i) luminal (further sub-divided into approved for the treatment of both HER-2-positive metastatic A and B) (ii) HER-2-positive/estrogen receptor (ER) - breast cancer and early stage HER-2-positive breast cancer. negative, (iii) normal-like and (iv) basal-like. The basal-like Recent results from trials of the dual HER-2 and EGFR sub-type lacks expression of ER, progesterone receptor tyrosine kinase inhibitor, lapatinib, also show very promising (PR) and HER-2, but expression of EGFR is more results in HER-2-positive breast cancer. A number of EGFR frequently observed in this sub-type.
    [Show full text]
  • Changing the Destiny of HER2 Expressing Solid Tumors
    International Journal of Molecular Sciences Review Trastuzumab Deruxtecan: Changing the Destiny of HER2 Expressing Solid Tumors Alice Indini 1 , Erika Rijavec 1 and Francesco Grossi 2,* 1 Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; [email protected] (A.I.); [email protected] (E.R.) 2 Medical Oncology Unit, Department of Medicine and Surgery, University of Insubria, ASST dei Sette Laghi, 21100 Varese, Italy * Correspondence: [email protected] or [email protected] Abstract: HER2 targeted therapies have significantly improved prognosis of HER2-positive breast and gastric cancer. HER2 overexpression and mutation is the pathogenic driver in non-small cell lung cancer (NSCLC) and colorectal cancer, however, to date, there are no approved HER2-targeted therapies with these indications. Trastuzumab deruxtecan (T-DXd) is a novel HER2-directed antibody drug conjugate showing significant anti-tumor activity in heavily pre-treated HER2-positive breast and gastric cancer patients. Preliminary data have shown promising objective response rates in patients with HER2-positive NSCLC and colorectal cancer. T-DXd has an acceptable safety profile, however with concerns regarding potentially serious treatment-emergent adverse events. In this review we focus on the pharmacologic characteristics and toxicity profile of T-Dxd, and provide an update on the most recent results of clinical trials of T-DXd in solid tumors. The referenced papers were selected through a PubMed search performed on 16 March 2021 with the following searching terms: T-DXd and breast cancer, or gastric cancer, or non-small cell lung cancer (NSCLC), or colorectal cancer.
    [Show full text]