301
|
Sandoval-Sus J, Mahtani R, Glück S. HER2-positive metastatic breast cancer: a double-edged sword. BREAST CANCER MANAGEMENT 2012. [DOI: 10.2217/bmt.12.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Jose Sandoval-Sus
- Department of Medicine, Division of Hematology/Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Leonard M Miller School of Medicine, Miami, FL, USA
| | - Reshma Mahtani
- Department of Medicine, Division of Hematology/Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Leonard M Miller School of Medicine, Miami, FL, USA
| | - Stefan Glück
- Department of Medicine, Division of Hematology/Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Leonard M Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
302
|
Zardavas D, Pugliano L, Ades F, Bozovic-Spasojevic I, Capelan M, de Azambuja E. Targeted treatments of HER2-positive metastatic breast cancer: trastuzumab and beyond. BREAST CANCER MANAGEMENT 2012. [DOI: 10.2217/bmt.12.35] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY HER2 positivity defines a molecular subtype of breast cancer with aggressive biological behavior. HER2 has been clinically validated as a prominent therapeutic target in breast cancer, and an abundance of data from clinical trials proving the efficacy of trastuzumab, a humanized anti-HER2 monoclonal antibody, has changed the natural history of this disease. Despite positive results from many trials, resistance to anti-HER2 agents inevitably occurs in both the metastatic and adjuvant settings. This clinical reality has led to the development of various other targeted agents designed to block the HER2 receptor. Similarly, attempts to elucidate the molecular mechanisms of resistance to the already established anti-HER2 agents have opened new therapeutic avenues with numerous promising agents. This review presents a comprehensive overview of the data available on anti-HER2-targeted agents other than trastuzumab, and describes both challenges and directions for the future.
Collapse
Affiliation(s)
- Dimitrios Zardavas
- Institut Jules Bordet & l’Univeristé Libre de Bruxelles, Brussels, Belgium
| | - Lina Pugliano
- Institut Jules Bordet & l’Univeristé Libre de Bruxelles, Brussels, Belgium
| | - Felipe Ades
- Institut Jules Bordet & l’Univeristé Libre de Bruxelles, Brussels, Belgium
| | | | - Marta Capelan
- Institut Jules Bordet & l’Univeristé Libre de Bruxelles, Brussels, Belgium
| | - Evandro de Azambuja
- BrEAST Data Centre, Jules Bordet Institute, Boulevard de Waterloo, 121 (7th Floor), 1000 Brussels, Belgium
| |
Collapse
|
303
|
Abstract
The discovery of the molecular processes involved in cancer development has led to the design of an array of targeted agents. These agents, directed to specific proteins in the machinery of cancer cells, interfere with vital cascades involved in cell invasion, metastasis, apoptosis, cell-cycle control and angiogenesis. In breast cancer, the main pathways studied and targeted by drugs are the HER2 pathway, EGFR, VEGF, PI3K/Akt/mammalian target of rapamycin (PI3K-M-Tor), IGF/IGFR, poly(ADP ribose) polymerase 1, HDAC and many others. In this review, we present the most promising studies of these new targeted therapies and novel combination of targeted therapies with cytotoxic agents for the treatment of breast cancer patients.
Collapse
|
304
|
Geuna E, Montemurro F, Aglietta M, Valabrega G. Potential of afatinib in the treatment of patients with HER2-positive breast cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2012; 4:131-7. [PMID: 24367201 PMCID: PMC3846413 DOI: 10.2147/bctt.s25868] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the absence of treatment, overexpression of the human epidermal growth factor receptor 2 (HER2) predicts a poor prognosis in breast cancer. In the last decade, monoclonal antibodies and small molecule tyrosine kinase inhibitors have significantly improved the outcome of HER2-positive breast cancer patients. However, tumor resistance and toxicities often limit the use of these therapies. For this reason, there is a compelling need for further investigation of new targeted therapies, such as afatinib, an oral irreversible pan inhibitor of the epidermal growth factor receptor (EGFR) family. This compound covalently interacts with tyrosine kinase domains, which are deeply involved in signal transduction leading to cell proliferation and protection from apoptosis. Afatinib has been studied in several Phase I clinical trials in advanced solid tumors. These trials have shown encouraging clinical activity and manageable side effects when afatinib is used either as a single agent or in combination with chemotherapy, with cutaneous adverse events and diarrhea being the most frequently observed toxicities. This review will focus on afatinib's clinical activity and will discuss ongoing clinical studies in HER2-positive breast cancer patients. In the scenario of the different HER2-targeted therapies, it will be important to define the best specific clinical and "molecular" setting for afatinib use, trying to identify predictors of resistance and response. Moreover, afatinib, which has the ability to cross the blood-brain barrier, could play a role in patients with brain metastases from breast cancer.
Collapse
Affiliation(s)
- Elena Geuna
- Division of Medical Oncology, Institute for Cancer Research and Treatment, Candiolo, Turin, Italy
| | - Filippo Montemurro
- Institute for Cancer Research, Candiolo, Turin, Italy
- Foundation of Piedmont Oncology, Candiolo, Turin, Italy
- Unit of Investigative Clinical Oncology, Candiolo, Turin, Italy
| | - Massimo Aglietta
- Division of Medical Oncology, Institute for Cancer Research and Treatment, Candiolo, Turin, Italy
- Institute for Cancer Research, Candiolo, Turin, Italy
- Foundation of Piedmont Oncology, Candiolo, Turin, Italy
- University Medical School of Turin, Turin, Italy
| | - Giorgio Valabrega
- Division of Medical Oncology, Institute for Cancer Research and Treatment, Candiolo, Turin, Italy
- Institute for Cancer Research, Candiolo, Turin, Italy
- Foundation of Piedmont Oncology, Candiolo, Turin, Italy
- University Medical School of Turin, Turin, Italy
| |
Collapse
|
305
|
Abstract
HER2-positive tumors comprise 15% to 20% of all breast cancers (BC) and are associated with worse clinical outcomes [Slamon et al., Science 1987;235:177-82]. Trastuzumab is a humanized monoclonal antibody designed to target the extracellular domain of the HER2 receptor, and is the foundation of care of women with early and advanced HER2-positive BC. However, a significant proportion of patients with this type of BC display either primary or secondary resistance to trastuzumab. Therefore, in an effort to overcome such resistance and further improve the outcome of patients with HER2-positive disease, several new anti-HER2 agents are currently being developed. These include small molecules that inhibit the HER2 tyrosine kinase activity (lapatinib, neratinib), monoclonal antibodies directed at other epitopes of the HER2 extracellular domain (pertuzumab), antibody-drug conjugates (trastuzumab-DMl), and heat shock protein 90 inhibitors (tanespimycin). A great deal of interest has been generated by recent data from the randomized neo-adjuvant studies NeoALTTO and NeoSphere, which have shown that dual blockade of the HER2 receptor with anti-HER2 agents is significantly superior to using one agent alone. If these results are validated in larger ongoing and planned phase III studies in early BC, they could lead to a paradigm shift in treatment strategy. Therefore, to avoid unnecessary toxicities and costs, it is critical to intensify the research for biomarkers that can identify those patients most likely to benefit from specific targeted therapies.
Collapse
|
306
|
Clinical perspectives for irreversible tyrosine kinase inhibitors in cancer. Biochem Pharmacol 2012; 84:1388-99. [PMID: 22885287 DOI: 10.1016/j.bcp.2012.07.031] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 07/27/2012] [Accepted: 07/30/2012] [Indexed: 11/21/2022]
Abstract
Irreversible inhibitors provide potent and selective inhibition of tyrosine kinase enzymes. Use of such inhibitors has proved promising in overcoming the tumor resistance encountered with reversible tyrosine kinase inhibitors. Irreversible inhibitors inactivate their protein target through covalent interaction with a nucleophilic cysteine residue within the nucleotide binding pocket of the kinase domain. Different irreversible tyrosin kinase inhibitors directed against epidermal growth factor receptor (EGFR), Bruton's tyrosine kinase (BTK), vascular endothelial growth factor receptor (VEGFR) and fibroblast growth factor receptor tyrosine kinase (FGFR) have been developed and some of them have been employed clinically as anticancer agents. This review focuses on recent preclinical and clinical progress with currently available irreversible tyrosine kinase inhibitors. The chemical structures of the candidates, structure-activity relationships, biological activities and results of current clinical investigations are described.
Collapse
|
307
|
SCHNUTE MARKE, HUANG ADRIAN, SAIAH EDDINE. Bruton's Tyrosine Kinase (Btk). ANTI-INFLAMMATORY DRUG DISCOVERY 2012. [DOI: 10.1039/9781849735346-00297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Bruton's tyrosine kinase (Btk) is a non-receptor tyrosine kinase belonging to the Tec family of kinases. Btk is critical for B-cell development, differentiation and signalling through the B-cell antigen receptor (BCR) as is evident by its genetic association to a human primary immunodeficiency disease known as X-linked Agammaglobulinemia (XLA). Btk is also present in specific cells of the myeloid lineage and contributes to the activation of the FcγR and FcεR signalling pathways in macrophages, neutrophils and mast cells. Because of its key role in these pathways, Btk is considered a promising target for therapeutic intervention in autoimmune and inflammatory disease. Numerous research groups are actively working to identify Btk inhibitors through the targeting of inactive kinase conformations or covalent active site inhibition. Both strategies have benefited from the rapid growth in structural biology insight for the target. Recently discovered potent and orally bioavailable Btk inhibitors have shown promising efficacy in several pre-clinical animal models of rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). These results, coupled with promising initial findings from the study of Btk inhibitors in human clinical trials for oncology, strongly suggest Btk intervention offers significant potential as a treatment strategy in inflammatory disease.
Collapse
Affiliation(s)
- MARK E. SCHNUTE
- Medicinal Chemistry, Pfizer Inc. 200 Cambridge Park Drive Cambridge, MA 02140 USA
| | - ADRIAN HUANG
- Department of Chemistry Wellesley College, 106 Central Street, Wellesley, MA 02481 USA
| | - EDDINE SAIAH
- Medicinal Chemistry, Pfizer Inc. 200 Cambridge Park Drive Cambridge, MA 02140 USA
| |
Collapse
|
308
|
Orphanos G, Kountourakis P. Targeting the HER2 Receptor in Metastatic Breast Cancer. Hematol Oncol Stem Cell Ther 2012; 5:127-37. [DOI: 10.5144/1658-3876.2012.127] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
309
|
Zhao XQ, Xie JD, Chen XG, Sim HM, Zhang X, Liang YJ, Singh S, Talele TT, Sun Y, Ambudkar SV, Chen ZS, Fu LW. Neratinib reverses ATP-binding cassette B1-mediated chemotherapeutic drug resistance in vitro, in vivo, and ex vivo. Mol Pharmacol 2012; 82:47-58. [PMID: 22491935 PMCID: PMC3382829 DOI: 10.1124/mol.111.076299] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 04/04/2012] [Indexed: 01/29/2023] Open
Abstract
Neratinib, an irreversible inhibitor of epidermal growth factor receptor and human epidermal receptor 2, is in phase III clinical trials for patients with human epidermal receptor 2-positive, locally advanced or metastatic breast cancer. The objective of this study was to explore the ability of neratinib to reverse tumor multidrug resistance attributable to overexpression of ATP-binding cassette (ABC) transporters. Our results showed that neratinib remarkably enhanced the sensitivity of ABCB1-overexpressing cells to ABCB1 substrates. It is noteworthy that neratinib augmented the effect of chemotherapeutic agents in inhibiting the growth of ABCB1-overexpressing primary leukemia blasts and KBv200 cell xenografts in nude mice. Furthermore, neratinib increased doxorubicin accumulation in ABCB1-overexpressing cell lines and Rhodamine 123 accumulation in ABCB1-overexpressing cell lines and primary leukemia blasts. Neratinib stimulated the ATPase activity of ABCB1 at low concentrations but inhibited it at high concentrations. Likewise, neratinib inhibited the photolabeling of ABCB1 with [(125)I]iodoarylazidoprazosin in a concentration-dependent manner (IC(50) = 0.24 μM). Neither the expression of ABCB1 at the mRNA and protein levels nor the phosphorylation of Akt was affected by neratinib at reversal concentrations. Docking simulation results were consistent with the binding conformation of neratinib within the large cavity of the transmembrane region of ABCB1, which provides computational support for the cross-reactivity of tyrosine kinase inhibitors with human ABCB1. In conclusion, neratinib can reverse ABCB1-mediated multidrug resistance in vitro, ex vivo, and in vivo by inhibiting its transport function.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP-Binding Cassette Transporters/genetics
- ATP-Binding Cassette Transporters/metabolism
- Adenosine Triphosphatases/genetics
- Adenosine Triphosphatases/metabolism
- Animals
- Antineoplastic Agents/pharmacology
- Cell Line, Tumor
- Doxorubicin/pharmacology
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Multiple/genetics
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- HEK293 Cells
- HL-60 Cells
- Humans
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Oncogene Protein v-akt/genetics
- Oncogene Protein v-akt/metabolism
- Phosphorylation/drug effects
- Quinolines/pharmacology
- RNA, Messenger/genetics
- Rhodamines/pharmacology
- rho-Associated Kinases/genetics
- rho-Associated Kinases/metabolism
Collapse
Affiliation(s)
- Xiao-qin Zhao
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
310
|
Criscitiello C, Metzger-Filho O, Saini KS, de Castro Jr G, Diaz M, La Gerche A, de Azambuja E, Piccart-Gebhart MJ. Targeted therapies in breast cancer: are heart and vessels also being targeted? Breast Cancer Res 2012; 14:209. [PMID: 22713170 PMCID: PMC3446327 DOI: 10.1186/bcr3142] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The concept of 'targeted' therapies implies that such drugs only act on cells that specifically express the particular target, therefore giving rise to a low incidence of side effects. However, targeted therapies currently approved for the treatment of breast cancer have demonstrated a relatively high incidence of cardiovascular events. The anti-HER2 agents trastuzumab and lapatinib may cause left ventricular dysfunction or even congestive heart failure. Bevacizumab, an antiangiogenic drug, has been shown to increase the risk of hypertension, cardiovascular dysfunction and thromboembolic events. In addition, several anti-human epidermal growth factor receptor 2 (HER2) and antiangiogenic agents plus their combinations are currently being developed and evaluated for the treatment of breast cancer. In this review, we aim to assess the incidence of cardiac adverse events associated with targeted therapies designed to block HER2 and angiogenic pathways.
Collapse
Affiliation(s)
- Carmen Criscitiello
- Department of Medical Oncology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Otto Metzger-Filho
- Department of Medical Oncology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Kamal S Saini
- Department of Medical Oncology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium
- Breast International Group, 1000 Brussels, Belgium
| | - Gilberto de Castro Jr
- Department of Clinical Oncology, Instituto do Câncer do Estado de São Paulo and Faculdade de Medicina da Universidade de São Paulo, São Paulo, 01246-000 Brazil
| | - Marie Diaz
- Department of Medical Oncology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - André La Gerche
- Department of Cardiovascular Medicine, University Hospital Leuven, B-3000 Belgium and Department of Cardiology, St Vincent's Hospital, University of Melbourne, Melbourne, Victoria, 3065 Australia
| | - Evandro de Azambuja
- Department of Medical Oncology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Martine J Piccart-Gebhart
- Department of Medical Oncology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium
- Breast International Group, 1000 Brussels, Belgium
| |
Collapse
|
311
|
Stopeck AT, Brown-Glaberman U, Wong HY, Park BH, Barnato SE, Gradishar WJ, Hudis CA, Rugo HS. The role of targeted therapy and biomarkers in breast cancer treatment. Clin Exp Metastasis 2012; 29:807-19. [PMID: 22692561 DOI: 10.1007/s10585-012-9496-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 05/28/2012] [Indexed: 12/24/2022]
Abstract
Breast cancer is the most prevalent life-threatening cancer in women and the second leading cause of cancer associated deaths. Consequently, optimizing breast cancer therapy to increase cure rates in early stage disease and improve life expectancy and palliation for patients with metastasis is a critical need and major area of research in medical oncology. This article focuses on the development of personalized therapy in breast cancer though the use of targeted therapies and their associated biomarkers. Topics reviewed include the historic advances in targeting the HER2 receptor and the current avenues being studied to circumvent resistance; new developments in the management of triple negative and metastatic breast cancer; and the challenges and pitfalls associated with targeting angiogenesis. Using these as examples, many of the innovations and challenges in the treatment of women with breast cancer are explored.
Collapse
Affiliation(s)
- Alison T Stopeck
- University of Arizona Cancer Center, 1515N. Campbell Avenue, Tucson, AZ 85724, USA.
| | | | | | | | | | | | | | | |
Collapse
|
312
|
Gori S, Montemurro F, Spazzapan S, Metro G, Foglietta J, Bisagni G, Ferzi A, Silva R, Gamucci T, Clavarezza M, Stocchi L, Fabi A, Cognetti F, Torrisi E, Crivellari D. Retreatment with trastuzumab-based therapy after disease progression following lapatinib in HER2-positive metastatic breast cancer. Ann Oncol 2012; 23:1436-41. [DOI: 10.1093/annonc/mdr474] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
313
|
Curigliano G. New drugs for breast cancer subtypes: Targeting driver pathways to overcome resistance. Cancer Treat Rev 2012; 38:303-10. [DOI: 10.1016/j.ctrv.2011.06.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 06/21/2011] [Indexed: 02/03/2023]
|
314
|
Krop IE, LoRusso P, Miller KD, Modi S, Yardley D, Rodriguez G, Guardino E, Lu M, Zheng M, Girish S, Amler L, Winer EP, Rugo HS. A phase II study of trastuzumab emtansine in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer who were previously treated with trastuzumab, lapatinib, an anthracycline, a taxane, and capecitabine. J Clin Oncol 2012; 30:3234-41. [PMID: 22649126 DOI: 10.1200/jco.2011.40.5902] [Citation(s) in RCA: 274] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE To determine whether the antibody-drug conjugate trastuzumab emtansine (T-DM1), which combines human epidermal growth factor receptor 2 (HER2) -targeted delivery of the potent antimicrotubule agent DM1 with the antitumor activity of trastuzumab, is effective in patients with HER2-positive metastatic breast cancer (MBC) who have previously received all standard HER2-directed therapies. PATIENTS AND METHODS In this single-arm phase II study, T-DM1 3.6 mg/kg was administered intravenously every 3 weeks to patients with HER2-positive MBC who had prior treatment with trastuzumab, lapatinib, an anthracycline, a taxane, and capecitabine. The primary objectives were overall response rate (ORR) by independent review and safety. RESULTS Among 110 pretreated patients (median, seven prior agents for MBC; median follow-up, 17.4 months), the ORR was 34.5% (95% CI, 26.1% to 43.9%), clinical benefit rate was 48.2% (95% CI, 38.8% to 57.9%), median progression-free survival (PFS) was 6.9 months (95% CI, 4.2 to 8.4 months), and median duration of response was 7.2 months (95% CI, 4.6 months to not estimable). In patients with confirmed HER2-positive tumors (n = 80 by retrospective central testing), the response rate was 41.3% (95% CI, 30.4% to 52.8%), and median PFS was 7.3 months (95% CI, 4.6 to 12.3 months). Most adverse events were grades 1 to 2; the most frequent grade ≥ 3 events were thrombocytopenia (9.1%), fatigue (4.5%), and cellulitis (3.6%). CONCLUSION T-DM1 is well tolerated and has single-agent activity in patients with HER2-positive MBC who have previously received both approved HER2-directed therapies and multiple chemotherapy agents. T-DM1 may be an effective new treatment for this patient population.
Collapse
Affiliation(s)
- Ian E Krop
- Dana-Farber Cancer Institute, 450 Brookline Ave, Boston MA 02215, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
315
|
Rexer BN, Arteaga CL. Intrinsic and acquired resistance to HER2-targeted therapies in HER2 gene-amplified breast cancer: mechanisms and clinical implications. Crit Rev Oncog 2012; 17:1-16. [PMID: 22471661 DOI: 10.1615/critrevoncog.v17.i1.20] [Citation(s) in RCA: 263] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Approximately 25% of human breast cancers overexpress the HER2 (ErbB2) proto-oncogene, which confers a more aggressive tumor phenotype and associates with a poor prognosis in patients with this disease. Two approved therapies targeting HER2, the monoclonal antibody trastuzumab and the tyrosine kinase inhibitor lapatinib, are clinically active against this type of breast cancer. However, a significant fraction of patients with HER2+ breast cancer treated with these agents eventually relapse or develop progressive disease. This suggests that tumors acquire or possess intrinsic mechanisms of resistance that allow escape from HER2 inhibition. This review focuses on mechanisms of intrinsic and/or acquired resistance to HER2-targeted therapies that have been identified in preclinical and clinical studies. These mechanisms involve alterations to HER2 itself, coexpression or acquisition of bypass signaling through other receptor or intracellular signaling pathways, defects in mechanisms of cell cycle regulation or apoptosis, and host factors that may modulate drug response. Emerging clinical evidence already suggests that combinations of therapies targeting HER2 as well as these resistance pathways will be effective in overcoming or preventing resistance.
Collapse
Affiliation(s)
- Brent N Rexer
- Departments of Medicine and Cancer Biology, Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37212, USA
| | | |
Collapse
|
316
|
Wong ALA, Lee SC. Mechanisms of Resistance to Trastuzumab and Novel Therapeutic Strategies in HER2-Positive Breast Cancer. Int J Breast Cancer 2012; 2012:415170. [PMID: 22649737 PMCID: PMC3357513 DOI: 10.1155/2012/415170] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 02/03/2012] [Indexed: 12/20/2022] Open
Abstract
HER2-positive breast cancers have poorer prognosis and are prime candidates for molecular-targeted therapy because they are driven by the unique mechanism of HER2 oncogene addiction. While anti-HER2 agents such as trastuzumab and lapatinib are integral to the treatment of HER2-positive breast cancer, intrinsic and secondary resistance pose a significant challenge, underscoring the need to develop novel anti-HER2 therapies. In recent years, an array of promising and novel anti-HER2 therapeutic agents and their combinations have entered various stages of clinical development. However, questions remain on the optimal sequences of HER2-directed therapies and selection of patients for the most appropriate drug or combinations; incompletely defined mechanisms of trastuzumab action and resistance have also dampened the progress of more successful biomarker-driven treatment approaches. This paper summarizes existing preclinical and clinical evidence on the mechanisms of trastuzumab action and resistance and provides an up-to-date overview of novel HER2-directed therapies in clinical development.
Collapse
Affiliation(s)
- Andrea L. A. Wong
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore 119228
| | - Soo-Chin Lee
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore 119228
- Cancer Science Institute, National University of Singapore, Singapore 117599
| |
Collapse
|
317
|
Davies DM, Foster J, van der Stegen SJC, Parente-Pereira AC, Chiapero-Stanke L, Delinassios GJ, Burbridge SE, Kao V, Liu Z, Bosshard-Carter L, van Schalkwyk MCI, Box C, Eccles SA, Mather SJ, Wilkie S, Maher J. Flexible targeting of ErbB dimers that drive tumorigenesis by using genetically engineered T cells. Mol Med 2012; 18:565-76. [PMID: 22354215 PMCID: PMC3388141 DOI: 10.2119/molmed.2011.00493] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 02/16/2012] [Indexed: 11/06/2022] Open
Abstract
Pharmacological targeting of individual ErbB receptors elicits antitumor activity, but is frequently compromised by resistance leading to therapeutic failure. Here, we describe an immunotherapeutic approach that exploits prevalent and fundamental mechanisms by which aberrant upregulation of the ErbB network drives tumorigenesis. A chimeric antigen receptor named T1E28z was engineered, in which the promiscuous ErbB ligand, T1E, is fused to a CD28 + CD3ζ endodomain. Using a panel of ErbB-engineered 32D hematopoietic cells, we found that human T1E28z⁺ T cells are selectively activated by all ErbB1-based homodimers and heterodimers and by the potently mitogenic ErbB2/3 heterodimer. Owing to this flexible targeting capability, recognition and destruction of several tumor cell lines was achieved by T1E28⁺ T cells in vitro, comprising a wide diversity of ErbB receptor profiles and tumor origins. Furthermore, compelling antitumor activity was observed in mice bearing established xenografts, characterized either by ErbB1/2 or ErbB2/3 overexpression and representative of insidious or rapidly progressive tumor types. Together, these findings support the clinical development of a broadly applicable immunotherapeutic approach in which the propensity of solid tumors to dysregulate the extended ErbB network is targeted for therapeutic gain.
Collapse
Affiliation(s)
- David M Davies
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Julie Foster
- Centre for Molecular Oncology and Imaging, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Sjoukje J C van der Stegen
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Ana C Parente-Pereira
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Laura Chiapero-Stanke
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - George J Delinassios
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Sophie E Burbridge
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Vincent Kao
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Zhe Liu
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Leticia Bosshard-Carter
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - May C I van Schalkwyk
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - Carol Box
- Tumour Biology and Metastasis, Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Suzanne A Eccles
- Tumour Biology and Metastasis, Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Stephen J Mather
- Centre for Molecular Oncology and Imaging, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Scott Wilkie
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
| | - John Maher
- King’s College London, King’s Health Partners Integrated Cancer Center, Department of Research Oncology, Guy’s Hospital Campus, London, UK
- Department of Immunology, Barnet and Chase Farm National Health Service (NHS) Trust, Barnet, Hertfordshire, UK
- Department of Clinical Immunology and Allergy, King’s College Hospital NHS Foundation Trust, Denmark Hill, London, UK
| |
Collapse
|
318
|
Nahta R. New developments in the treatment of HER2-positive breast cancer. BREAST CANCER-TARGETS AND THERAPY 2012; 4:53-64. [PMID: 23869176 DOI: 10.2147/bctt.s24976] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Approximately 20%-30% of metastatic breast cancers show increased expression of the human epidermal growth factor receptor-2 (HER2) tyrosine kinase. Two HER2-specific therapies are currently approved for clinical treatment of patients with HER2-overexpressing metastatic breast cancer. Trastuzumab is a monoclonal antibody against HER2 and is approved for first-line treatment of HER2-positive metastatic breast cancer. Lapatinib is a small molecule dual inhibitor of epidermal growth factor receptor and HER2 tyrosine kinases, and is approved for trastuzumab-refractory disease. Although trastuzumab is a highly effective therapy for patients with HER2-overexpressing metastatic breast cancer, a significant number of patients in the initial clinical trials of trastuzumab monotherapy showed resistance to trastuzumab-based therapy. Further, among those who did respond, the initial trials indicated that the median time to progression was less than 1 year. Similarly, lapatinib is effective in a subset of trastuzumab-refractory cases, but the majority of patients display resistance. This review discusses the multiple molecular mechanisms of resistance that have been proposed in the literature. In addition, novel agents that are being tested for efficacy against HER2-positive breast cancer, including the antibodies pertuzumab and trastuzumab-DM1 and the immunotoxin affitoxin, are reviewed. The introduction of trastuzumab has revolutionized the clinical care of patients with HER2-positive metastatic breast cancer and has resulted in dramatic reductions in recurrences of early-stage HER2-positive breast cancer. The development and implementation of gene- and protein-based assays that measure potential molecular predictors of trastuzumab resistance will allow individualization of HER2-targeted therapeutic approaches, and may ultimately improve treatment of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Rita Nahta
- Departments of Pharmacology and Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
319
|
De Mattos-Arruda L, Cortes J. Advances in first-line treatment for patients with HER-2+ metastatic breast cancer. Oncologist 2012; 17:631-44. [PMID: 22523199 DOI: 10.1634/theoncologist.2011-0187] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The prognosis for breast cancer patients overexpressing human epidermal growth factor receptor (HER)-2 has changed with anti-HER-2-targeted therapy. Although anti-HER-2 therapy with trastuzumab and chemotherapy is the standard first-line treatment, the best therapeutic regimen has yet to be defined, and new strategies are evolving. METHODS A literature review of well-established and recently published trials, reviews, and ongoing clinical trials addressing first-line treatment for HER-2(+) metastatic breast cancer patients was performed. RESULTS Taxanes are the agents most commonly used in combination with trastuzumab, but other chemotherapy drugs, such as anthracyclines, vinorelbine, and gemcitabine and triple-combination therapies including platinum compounds, capecitabine, and taxanes have been studied. The combination of aromatase inhibitors with anti-HER-2 therapies is a new therapeutic option for some patients who coexpress HER-2 and hormone receptors, although its activity observed in randomized clinical trials seems to be inferior to that of chemotherapy plus anti-HER-2 therapies. In addition, new anti-HER-2 therapies have shown activity in HER-2(+) tumors, both alone and in combination with trastuzumab. CONCLUSIONS Trastuzumab plus chemotherapy is the current standard of care for the upfront treatment of HER-2(+) breast cancer patients, though other anti-HER-2-targeting agents may appear as new standards in the upcoming years.
Collapse
|
320
|
Trastuzumab and Gemcitabine in Pretreated HER2 Overexpressing Metastatic Breast Cancer Patients: Retrospective Analysis of Our Series. JOURNAL OF ONCOLOGY 2012; 2012:198412. [PMID: 22536237 PMCID: PMC3321447 DOI: 10.1155/2012/198412] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 01/05/2012] [Accepted: 01/20/2012] [Indexed: 11/17/2022]
Abstract
Trastuzumab-based regimes improved clinical outcome in women with overexpressing HER2 metastatic breast cancer, mainly due to the availability of different combination therapies, clinically active and well tolerated. In this study we retrospectively evaluated clinical activity and toxicity of trastuzuamb plus gemcitabine regimen in heavily pretreated HER2 positive metastatic breast cancer patients. Although the observed population was heavily pretreated, the evaluated regimen was notably effective in terms of response rate, time to progression and survival, with very mild toxicity. These data suggest that in over expressing HER2 metastatic breast cancer patients, sequential trastuzumab based chemotherapeutic regimens can achieve good response rate with prolonged TTP in responding patients, even after other target therapy such as lapatinib based combinations.
Collapse
|
321
|
Seyhan AA, Varadarajan U, Choe S, Liu W, Ryan TE. A genome-wide RNAi screen identifies novel targets of neratinib resistance leading to identification of potential drug resistant genetic markers. MOLECULAR BIOSYSTEMS 2012; 8:1553-70. [PMID: 22446932 DOI: 10.1039/c2mb05512k] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Neratinib (HKI-272) is a small molecule tyrosine kinase inhibitor of the ErbB receptor family currently in Phase III clinical trials. Despite its efficacy, the mechanism of potential cellular resistance to neratinib and genes involved with it remains unknown. We have used a pool-based lentiviral genome-wide functional RNAi screen combined with a lethal dose of neratinib to discover chemoresistant interactions with neratinib. Our screen has identified a collection of genes whose inhibition by RNAi led to neratinib resistance including genes involved in oncogenesis (e.g. RAB33A, RAB6A and BCL2L14), transcription factors (e.g. FOXP4, TFEC, ZNF), cellular ion transport (e.g. CLIC3, TRAPPC2P1, P2RX2), protein ubiquitination (e.g. UBL5), cell cycle (e.g. CCNF), and genes known to interact with breast cancer-associated genes (e.g. CCNF, FOXP4, TFEC, several ZNF factors, GNA13, IGFBP1, PMEPA1, SOX5, RAB33A, RAB6A, FXR1, DDO, TFEC, OLFM2). The identification of novel mediators of cellular resistance to neratinib could lead to the identification of new or neoadjuvant drug targets. Their use as patient or treatment selection biomarkers could make the application of anti-ErbB therapeutics more clinically effective.
Collapse
Affiliation(s)
- Attila A Seyhan
- Systems Biology, Global Biotherapeutics, Pfizer Inc., 200 Cambridgepark Drive, Cambridge, MA 02140, USA.
| | | | | | | | | |
Collapse
|
322
|
Lin NU, Winer EP, Wheatley D, Carey LA, Houston S, Mendelson D, Munster P, Frakes L, Kelly S, Garcia AA, Cleator S, Uttenreuther-Fischer M, Jones H, Wind S, Vinisko R, Hickish T. A phase II study of afatinib (BIBW 2992), an irreversible ErbB family blocker, in patients with HER2-positive metastatic breast cancer progressing after trastuzumab. Breast Cancer Res Treat 2012; 133:1057-65. [PMID: 22418700 PMCID: PMC3387495 DOI: 10.1007/s10549-012-2003-y] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 02/16/2012] [Indexed: 12/12/2022]
Abstract
Afatinib is an oral, ErbB family blocker, which covalently binds and irreversibly blocks all kinase-competent ErbB family members. This phase II, open-label, single-arm study explored afatinib activity in human epidermal growth factor receptor 2 (HER2)-positive breast cancer patients progressing after trastuzumab treatment. Patients had stage IIIB/IV HER2-positive metastatic breast cancer, with progression following trastuzumab or trastuzumab intolerance and an Eastern Cooperative Oncology Group (ECOG) performance status of 0–2. Patients received 50 mg afatinib once-daily until disease progression. Primary endpoint was objective response rate (Response Evaluation Criteria in Solid Tumors 1.0), with tumor assessments every 8 weeks. Forty-one patients were treated. Patients had received a median of three prior chemotherapy lines (range, 0–15) and 68.3% had received trastuzumab for >1 year. Four patients (10% of 41 treated; 11% of evaluable patients) had partial response. Fifteen patients (37% of 41) had stable disease as best response and 19 (46% of 41) achieved clinical benefit. Median progression-free survival was 15.1 weeks (95% confidence interval [CI]: 8.1–16.7); median overall survival was 61.0 weeks (95% CI: 56.7–not evaluable). Most frequent common terminology criteria for adverse events grade 3 treatment-related adverse events were diarrhea (24.4%) and rash (9.8%). Afatinib monotherapy was associated with promising clinical activity in extensively pretreated HER2-positive breast cancer patients who had progressed following trastuzumab treatment.
Collapse
Affiliation(s)
- Nancy U Lin
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
323
|
Abbas R, Hug BA, Leister C, Sonnichsen D. A double-blind, randomized, multiple-dose, parallel-group study to characterize the occurrence of diarrhea following two different dosing regimens of neratinib, an irreversible pan-ErbB receptor tyrosine kinase inhibitor. Cancer Chemother Pharmacol 2012; 70:191-9. [PMID: 22418773 DOI: 10.1007/s00280-012-1857-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 02/26/2012] [Indexed: 10/28/2022]
Abstract
PURPOSE Neratinib, a potent, low-molecular-weight, orally administered, irreversible, pan-ErbB receptor tyrosine kinase inhibitor has antitumor activity in ErbB2 + breast cancer. The objective of this study was to characterize the onset, severity, and duration of diarrhea after administration of neratinib 240 mg once daily (QD) and 120 mg twice daily (BID) for ≤14 days in healthy subjects. METHODS A randomized, double-blind, parallel-group, inpatient study was conducted in 50 subjects given oral neratinib either 240 mg QD or 120 mg BID with food for ≤14 days. The primary endpoint was the proportion of subjects with diarrhea of at least moderate severity (grade 2; 5-7 loose stools/day). In subjects with grade 2 diarrhea, fecal analytes were determined. Pharmacokinetic profiles were characterized for neratinib on Days 1 and 7. RESULTS No severe (grade 3) diarrhea was reported. By Day 4, all subjects had grade 1 diarrhea. Grade 2 diarrhea occurred in 11/22 evaluable subjects (50 % [90 % confidence interval (CI): 28-72 %]) in the QD group and 17/23 evaluable subjects (74 % [90 % CI: 52-90 %]) in the BID group (P = 0.130). In fecal analyses, 18 % tested positive for hemoglobin and 46 % revealed fecal lactoferrin. Specimen pH was neutral to slightly alkaline. In pharmacokinetic analyses, Day 1 peak plasma concentration and Day 7 steady-state exposure were higher with the QD regimen than the BID regimen. In an exploratory analysis, ABCG2 genotype showed no correlation with severity or onset of diarrhea. CONCLUSIONS Incidences and onsets of at least grade 1 and at least grade 2 diarrhea were not improved on BID dosing compared with QD dosing.
Collapse
Affiliation(s)
- Richat Abbas
- Pfizer Inc, 500 Arcola Road, Collegeville, PA 19426, USA.
| | | | | | | |
Collapse
|
324
|
Rana P, Sridhar SS. Efficacy and tolerability of lapatinib in the management of breast cancer. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2012; 6:67-77. [PMID: 22438669 PMCID: PMC3306225 DOI: 10.4137/bcbcr.s6374] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Approximately 20%–25% of all breast cancers over express a key cell surface growth factor receptor known as HER2. HER2 plays a key role in cell growth and proliferation and is linked to worse clinical outcomes, making it a logical therapeutic target. The first HER2 targeted drug to be approved by the FDA, was the humanized monoclonal antibody trastuzumab, after it showed improvements in survival in the adjuvant setting, and delayed time to progression in the metastatic setting. Although highly effective, for reasons that are not clear, some patients display resistance to trastuzumab. Lapatinib is an oral, small molecule tyrosine kinase inhibitor, that inhibits both the HER1 ahd HER2 receptors and may be able to overcome trastuzumab resistance. Lapatinib is approved in the second line setting for use in combination with capecitabine or with letrozole. In this review, we will discuss the indications, concerns or any issues with regards to the drug.
Collapse
Affiliation(s)
- Punam Rana
- Department of Medical Oncology and Hematology, Princess Margaret Hospital, Toronto, Ontario, Canada
| | | |
Collapse
|
325
|
Olson EM. Maximizing human epidermal growth factor receptor 2 inhibition: a new oncologic paradigm in the era of targeted therapy. J Clin Oncol 2012; 30:1712-4. [PMID: 22393085 DOI: 10.1200/jco.2011.40.2545] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Erin M Olson
- Richard J. Solove Research Institute, Columbus, OH 43210, USA.
| |
Collapse
|
326
|
Kim HJ, Kim HP, Yoon YK, Kim MS, Lee GS, Han SW, Im SA, Kim TY, Oh DY, Bang YJ. Antitumor activity of HM781-36B, a pan-HER tyrosine kinase inhibitor, in HER2-amplified breast cancer cells. Anticancer Drugs 2012; 23:288-297. [PMID: 23422737 DOI: 10.1097/cad.0b013e32834e7d9b] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
HM781-36B is an orally administered pan-human epidermal growth factor receptor (HER) inhibitor. To explore the role of pan-HER inhibitor in breast cancer, we investigated the antitumor effect and mechanisms of HM781-36B in breast cancer cell lines. Six breast cancer cell lines (BT474, MDA-MB-453, SK-BR-3, T47D, MCF-7, and MDA-MB-231) were tested. The growth inhibitory effect was assessed using the tetrazolium bromide [3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyl-tetrazolium bromide] assay. The cell cycle at various concentrations of HM781-36B was analyzed by flow cytometry, and analysis of downstream molecules was performed by western blot analysis. Interaction of HM781-36B with cytotoxic chemotherapeutic agents was analyzed by combination index using CalcuSyn. The HER2-amplified cells (SK-BR-3, BT474, and MDA-MB-453) were sensitive to HM781-36B (IC50=0.001 μmol/l, 0.0012 μmol/l, and 0.0095 μmol/l, respectively). HM781-36B induced G1 arrest and resulted in apoptosis. It reduced the level of p-HER2, p-AKT, p-ERK, and p-STAT3. HM781-36B combined with 5-fluorouracil, cisplatin, paclitaxel, or gemcitabine showed a synergistic inhibitory effect on the HER2-amplified and on some of the HER2-nonamplified breast cancer cells. HM781-36B could be a promising treatment for HER2-amplified breast cancer as a single agent or in combination with cytotoxic agents and can be a candidate for treatment of HER2-nonamplified breast cancer in combination with cytotoxic agents.
Collapse
|
327
|
What Are the Most Important New Targets in Breast Cancer? CURRENT BREAST CANCER REPORTS 2012. [DOI: 10.1007/s12609-011-0068-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
328
|
Ito Y, Suenaga M, Hatake K, Takahashi S, Yokoyama M, Onozawa Y, Yamazaki K, Hironaka S, Hashigami K, Hasegawa H, Takenaka N, Boku N. Safety, efficacy and pharmacokinetics of neratinib (HKI-272) in Japanese patients with advanced solid tumors: a Phase 1 dose-escalation study. Jpn J Clin Oncol 2012; 42:278-86. [PMID: 22371427 DOI: 10.1093/jjco/hys012] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE Neratinib (HKI-272), a potent, irreversible, small-molecule, orally administered, pan-ErbB inhibitor that blocks signal transduction via inhibition of three epidermal growth factor receptors [ErbB1, ErbB2 (Her2) and ErbB4], is being developed for the treatment of solid tumors, including breast cancer. This Phase 1 dose-escalation study assessed the safety, tolerability, maximum-tolerated dose, antitumor activity and pharmacokinetics of neratinib in Japanese patients with advanced solid tumors. METHODS Patients received neratinib 80, 160, 240 or 320 mg orally; each patient enrolled in only one dose cohort. Patients received a single dose in week 1, followed by daily continuous doses. Blood samples collected were on days 1 and 21 for pharmacokinetic analyses. RESULTS Twenty-one patients were enrolled (3 breast cancer; 17 colorectal cancer; 1 gastric cancer). Neratinib-related adverse events (all grades) included diarrhea (20 patients), fatigue (14 patients), nausea and abdominal pain (9 patients each) and anorexia (8 patients). Grade ≥3 neratinib-related adverse events in two or more patients were diarrhea and anorexia (two patients each). Dose-limiting toxicities were diarrhea and anorexia (two patients, 320 mg dose). The maximum-tolerated dose and recommended dose was neratinib 240 mg once daily. Of 21 evaluable patients, 2 with breast cancer had partial response, 3 had stable disease ≥24 weeks, 7 had stable disease ≥16 weeks and 9 had progressive disease. Pharmacokinetic analyses indicated that neratinib exposures increased with dose. CONCLUSIONS The safety, efficacy and pharmacokinetic profiles of neratinib are consistent with those reported for non-Japanese patients and warrant further investigation of neratinib in Japanese patients with solid tumors.
Collapse
Affiliation(s)
- Yoshinori Ito
- Breast Cancer Division, Department of Medical Oncology, The Cancer Institute of the Japanese Foundation for Cancer Research, 3-8-31 Ariake Koto-ku, Tokyo 135-8550, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
329
|
Wong H, Yau T. Targeted therapy in the management of advanced gastric cancer: are we making progress in the era of personalized medicine? Oncologist 2012; 17:346-58. [PMID: 22334453 PMCID: PMC3316920 DOI: 10.1634/theoncologist.2011-0311] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 12/19/2011] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Gastric cancer is one of the leading causes of cancer death. With greater understanding of the molecular basis of carcinogenesis, targeted agents have led to a modest improvement in the outcome of advanced gastric cancer (AGC) patients. METHODS AND RESULTS We conducted an overview of the published evidence regarding the use of targeted therapy in AGC patients. Thus far, the human epidermal growth factor receptor (HER) pathway, angiogenic pathway, and phosphatidylinositol-3-kinase (PI3K)-Akt-mammalian target of rapamycin pathway have emerged as potential avenues for targeted therapy in AGC patients. The promising efficacy results of the Trastuzumab for Gastric Cancer trial led to the approved use of trastuzumab-based therapy as first-line treatment for patients with HER-2+ AGC. On the other hand, the Avastin® in Gastric Cancer trial evaluating bevacizumab in combination with chemotherapy did not meet its primary endpoint of a longer overall survival duration despite a significantly higher response rate and longer progression-free survival time in patients in the bevacizumab arm. Phase III data are awaited for other targeted agents, including cetuximab, panitumumab, lapatinib, and everolimus. CONCLUSION Recent progress in targeted therapy development for AGC has been modest. Further improvement in the outcome of AGC patients will depend on the identification of biomarkers in different patient populations to facilitate the understanding of gastric carcinogenesis, combining different targeted agents with chemotherapy, and unraveling new molecular targets for therapeutic intervention.
Collapse
Affiliation(s)
- Hilda Wong
- Division of Hematology and Medical Oncology, Department of Medicine, and
| | - Thomas Yau
- Division of Hematology and Medical Oncology, Department of Medicine, and
- Department of Surgery, Queen Mary Hospital, Hong Kong
- Centre for Cancer Research, The University of Hong Kong, Hong Kong
| |
Collapse
|
330
|
Abstract
The use of trastuzumab, a monoclonal antibody that targets the human epidermal growth factor receptor 2 (HER2) alteration present in 25 to 30% of breast cancers, has been associated with improved survival outcomes in both the adjuvant and metastatic settings. However, despite the robust clinical efficacy of trastuzumab in HER2-positive metastatic breast cancer (MBC), primary and secondary resistance remains a clinical challenge. Although lapatinib has demonstrated modest activity in this setting, trials reported to date have yet to demonstrate improvements in overall survival with its use. Novel therapeutic strategies to circumvent trastuzumab resistance are warranted, and agents targeting the HER, vascular endothelial growth factor, heat shock protein 90, phosphoinositide 3 kinase/Akt/mammalian target of rapamycin, and insulin-like growth factor-1 receptor pathways represent rational approaches in the management of HER2-positive disease. In this review, early-phase and emerging trial data surrounding the use of these promising agents in HER2-positive MBC will be discussed.
Collapse
|
331
|
New therapies in HER2-positive breast cancer: a major step towards a cure of the disease? Cancer Treat Rev 2012; 38:494-504. [PMID: 22305205 DOI: 10.1016/j.ctrv.2012.01.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 01/02/2012] [Accepted: 01/03/2012] [Indexed: 01/22/2023]
Abstract
Overexpression of the human epidermal growth factor receptor 2 (HER2) predicts a poor prognosis in metastatic breast cancer. While the introduction of HER2-targeted therapies, such as the monoclonal antibody trastuzumab and the small-molecule tyrosine kinase inhibitor lapatinib, has significantly improved outcomes in HER2+ breast cancer compared with previously available therapies, use of these targeted therapies is often limited by the development of drug resistance and tolerability issues. These limitations create the need for further development and investigation of new targeted therapies that show potent and selective inhibition of these targets or closely connected molecular pathways. Recently, several agents have demonstrated promising activity in HER2+ metastatic breast cancer, either as monotherapy or in combination therapy, including the tyrosine-kinase inhibitors neratinib (HKI-272) and afatinib (BIBW-2992) and the anti-HER2 monoclonal antibodies pertuzumab and trastuzumab-DM1 (T-DM1). Agents that target other molecular pathways, such as the vascular endothelial growth factor receptor, mammalian target of rapamycin, PI3-kinases, insulin-like growth factor (IGFR), HSP-90, and other kinases also have potential, in combination with anti-HER2 and/or other systemic therapies, to be active in this subtype of breast cancer. Innovative clinical studies are required in well-characterized patient populations to define the true clinical value of these emerging new approaches.
Collapse
|
332
|
Curigliano G, Locatelli M, Fumagalli L, Brollo J, Munzone E, Nolé F, Criscitiello C, Goldhirsch A. Targeting the subtypes of breast cancer: rethinking investigational drugs. Expert Opin Investig Drugs 2012; 21:191-204. [DOI: 10.1517/13543784.2012.651456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
333
|
|
334
|
Noe MC, Gilbert AM. Targeted Covalent Enzyme Inhibitors. ANNUAL REPORTS IN MEDICINAL CHEMISTRY VOLUME 47 2012. [DOI: 10.1016/b978-0-12-396492-2.00027-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
335
|
Gersch M, Kreuzer J, Sieber SA. Electrophilic natural products and their biological targets. Nat Prod Rep 2012; 29:659-82. [DOI: 10.1039/c2np20012k] [Citation(s) in RCA: 193] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
336
|
Olson EM, Lin NU, DiPiro PJ, Najita JS, Krop IE, Winer EP, Burstein HJ. Responses to subsequent anti-HER2 therapy after treatment with trastuzumab-DM1 in women with HER2-positive metastatic breast cancer. Ann Oncol 2012; 23:93-97. [PMID: 21531783 PMCID: PMC3276325 DOI: 10.1093/annonc/mdr061] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 02/08/2011] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Women with human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer (MBC) can respond to multiple lines of anti-HER2 therapy. It is unknown whether these patients will derive further clinical benefit following treatment with trastuzumab-MCC-DM1 (T-DM1). PATIENTS AND METHODS We retrospectively identified HER2-positive MBC patients treated with T-DM1 and characterized outcomes during subsequent lines of anti-HER2 therapy. Response was determined by a blinded radiology review. Time-dependent analyses were carried out using Kaplan-Meier estimates. RESULTS We identified 23 patients treated with single-agent T-DM1 and report on the 20 patients who discontinued protocol therapy. All patients received trastuzumab-based metastatic therapy before initiation of T-DM1 [median 7 regimens (range 3-14)]. Of these 20 patients, 75% (15 of 20) received further therapy with or without anti-HER2 agents after discontinuing T-DM1. Partial response to either first- or second-subsequent line(s) of therapy was seen in 5 of 15 (33%) treated patients, including 33% (4 of 12) who received a regimen containing trastuzumab and/or lapatinib. Median durations of therapy to first- and second-subsequent regimens after T-DM1 were 5.5 and 6.4 months, respectively. CONCLUSIONS In heavily pretreated HER2-positive MBC patients, prior exposure to T-DM1 does not exhaust the potential benefit of ongoing anti-HER2 therapy with trastuzumab- and/or lapatinib-based regimens.
Collapse
Affiliation(s)
| | - N U Lin
- Departments of Medical Oncology
| | | | - J S Najita
- Departments of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | | | | | | |
Collapse
|
337
|
Incorporating Agents that Target HER2 in the Neoadjuvant Setting. CURRENT BREAST CANCER REPORTS 2011. [DOI: 10.1007/s12609-011-0054-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
338
|
Saunus JM, Momeny M, Simpson PT, Lakhani SR, Da Silva L. Molecular aspects of breast cancer metastasis to the brain. GENETICS RESEARCH INTERNATIONAL 2011; 2011:219189. [PMID: 22567347 PMCID: PMC3335473 DOI: 10.4061/2011/219189] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Revised: 07/15/2011] [Accepted: 08/14/2011] [Indexed: 02/05/2023]
Abstract
Our knowledge of the biology underlying the development of brain metastases (BM) from breast cancer has improved over the last decade due to large clinical epidemiological studies, animal models of metastasis, and the use of high-resolution gene expression profiling technologies. However, there are still major gaps in our understanding of the mechanisms utilized by breast cancer cells to colonize the brain microenvironment, thus our arsenal of therapies remains relatively nonspecific, and the prognosis for breast cancer patients with BM remains poor. Additional insights into these mechanisms are necessary to facilitate the development of new preventive and curative therapeutic regimens to block this fatal disease. This paper aims to provide a general overview for the readers of what has been achieved in this field of research and its translation into clinical practice to date and to highlight exciting new areas of research that promise to inform the development of new targeted therapies for BM.
Collapse
Affiliation(s)
- Jodi M Saunus
- UQ Centre for Clinical Research, The University of Queensland, Herston, QLD 4029, Australia
| | | | | | | | | |
Collapse
|
339
|
Abstract
The advent of HER2-directed therapies has significantly improved the outlook for patients with HER2-positive early stage breast cancer. However, a significant proportion of these patients still relapse and die of breast cancer. Trials to define, refine and optimize the use of the two approved HER2-targeted agents (trastuzumab and lapatinib) in patients with HER2-positive early stage breast cancer are ongoing. In addition, promising new approaches are being developed including monoclonal antibodies and small-molecule tyrosine kinase inhibitors targeting HER2 or other HER family members, antibodies linked to cytotoxic moieties or modified to improve their immunological function, immunostimulatory peptides, and targeting the PI3K and IGF-1R pathways. Improved understanding of the HER2 signaling pathway, its relationship with other signaling pathways and mechanisms of resistance has also led to the development of rational combination therapies and to a greater insight into treatment response in patients with HER2-positive breast cancer. Based on promising results with new agents in HER2-positive advanced-stage disease, a series of large trials in the adjuvant and neoadjuvant settings are planned or ongoing. This Review focuses on current treatment for patients with HER2-positive breast cancer and aims to update practicing clinicians on likely future developments in the treatment for this disease according to ongoing clinical trials and translational research.
Collapse
|
340
|
Barton S, Swanton C. Recent Developments in Treatment Stratification for Metastatic Breast Cancer. Drugs 2011; 71:2099-113. [DOI: 10.2165/11594480-000000000-00000] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
341
|
Wong H, Leung R, Kwong A, Chiu J, Liang R, Swanton C, Yau T. Integrating molecular mechanisms and clinical evidence in the management of trastuzumab resistant or refractory HER-2⁺ metastatic breast cancer. Oncologist 2011; 16:1535-46. [PMID: 22020213 DOI: 10.1634/theoncologist.2011-0165] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Human epidermal growth factor receptor (HER)-2(+) breast cancer is a distinct molecular and clinical entity, the prognosis of which is improved by trastuzumab. However, primary resistance to trastuzumab is observed in >50% of patients with HER-2(+) advanced breast cancer, and the majority of patients who initially respond to treatment eventually develop disease progression. To facilitate crosstrial comparisons and the understanding of resistance mechanisms, we propose a unifying definition of trastuzumab resistance as progression at first radiological reassessment at 8-12 weeks or within 3 months after first-line trastuzumab in the metastatic setting or new recurrences diagnosed during or within 12 months after adjuvant trastuzumab. In contrast, we define trastuzumab-refractory breast cancer as disease progression after two or more lines of trastuzumab-containing regimens that initially achieved disease response or stabilization at first radiological assessment. We review mechanisms of trastuzumab resistance mediated by p95HER-2 overexpression, phosphoinositide 3-kinase pathway activation, and signaling pathway activation driven by HER-3, epidermal growth factor receptor, and insulin-like growth factor 1 receptor. We distinguish in vitro from in vivo evidence, highlighting that most data describing trastuzumab resistance are derived from preclinical studies or small retrospective patient cohorts, and discuss targeted therapeutic approaches to overcome resistance. Prospective analysis through clinical trials with robust tissue collection procedures, prior to and following acquisition of resistance, integrated with next-generation tumor genome sequencing technologies, is identified as a priority area for development. The identification of predictive biomarkers is of paramount importance to optimize health economic costs and enhance stratification of anti-HER-2 targeted therapies.
Collapse
Affiliation(s)
- Hilda Wong
- Division of Hematology and Medical Oncology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | |
Collapse
|
342
|
Kwak E. The role of irreversible HER family inhibition in the treatment of patients with non-small cell lung cancer. Oncologist 2011; 16:1498-507. [PMID: 22016476 DOI: 10.1634/theoncologist.2011-0087] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Small-molecule tyrosine kinase inhibitors (TKIs) of the human epidermal growth factor receptor (HER) include the reversible epidermal growth factor receptor (EGFR/HER-1) inhibitors gefitinib and erlotinib. EGFR TKIs have demonstrated activity in the treatment of patients with non-small cell lung cancer (NSCLC) harboring activating EGFR mutations; however, multiple mechanisms of resistance limit the benefit of these drugs. Although resistance to EGFR TKIs can be intrinsic and correlated with molecular lesions such as in Kirsten rat sarcoma viral oncogene homolog (KRAS; generally observed in a wild-type EGFR background), acquired resistance to EGFR TKIs can evolve in the setting of activating EGFR mutations, such as in the case of EGFR T790M mutations. Several irreversible inhibitors that target multiple members of the HER family simultaneously are currently in clinical development for NSCLC and may have a role in the treatment of TKI-sensitive and TKI-resistant disease. These include PF00299804, an inhibitor of EGFR/HER-1, HER-2, and HER-4, and afatinib (BIBW 2992), an inhibitor of EGFR/HER-1, HER-2, and HER-4. Results of large, randomized trials of these agents may help to determine their potential for the treatment of NSCLC.
Collapse
Affiliation(s)
- Eunice Kwak
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts 02114, USA.
| |
Collapse
|
343
|
Perez EA, Spano JP. Current and emerging targeted therapies for metastatic breast cancer. Cancer 2011; 118:3014-25. [DOI: 10.1002/cncr.26356] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 04/27/2011] [Accepted: 05/10/2011] [Indexed: 11/09/2022]
|
344
|
Abstract
The HER family of receptors consists of four closely related type 1 transmembrane TK receptors: HER1 (EGFR), HER2, HER3 and HER4. Signalling via the HER family of receptors underpins the majority of the intricate array of cellular activities on which cell survival and functionality depend. Aberrant HER2 expression and/or functionality have been implicated in the evolution of breast cancer and this receptor has proved to be a potent target for anticancer therapies, including antibody-based therapies to prevent ligand binding, dimer formation or the recruitment of antibody-dependent cell-mediated cytotoxicity, and direct kinase inhibition to prevent molecular activation and recruitment of downstream signalling partners. Novel strategies against HER2 include HER tyrosine kinase inhibitors, HSP90 inhibitors and antibody-chemotherapy conjugates. This latter approach is exemplified by T-DM1, a potent antibody that has a good safety profile and that has shown remarkable activity in patients with advanced disease. In addition, pertuzumab, an mAb that directly inhibits the formation of HER2 dimers including the HER2:HER3 dimer, offers a unique mechanism of HER3 inhibition. All these approaches have shown substantial clinical activity in patients refractory to trastuzumab. It is anticipated that with the increased availability of novel anti-HER2 agents together with a better understanding of the mechanisms of resistance to anti-HER2 agents we should be able to further improve the outcome of patients with HER2 breast cancer. There will also be an increasing tendency towards moving the study of these agents to earlier stages of the disease, namely in the adjuvant and neoadjuvant setting.
Collapse
Affiliation(s)
- J Baselga
- Massachusetts General Hospital Cancer Center, Boston, MA, USA.
| |
Collapse
|
345
|
LoRusso PM, Weiss D, Guardino E, Girish S, Sliwkowski MX. Trastuzumab emtansine: a unique antibody-drug conjugate in development for human epidermal growth factor receptor 2-positive cancer. Clin Cancer Res 2011; 17:6437-47. [PMID: 22003071 DOI: 10.1158/1078-0432.ccr-11-0762] [Citation(s) in RCA: 336] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Trastuzumab emtansine (T-DM1) is a human epidermal growth factor receptor (HER2)-targeted antibody-drug conjugate, composed of trastuzumab, a stable thioether linker, and the potent cytotoxic agent DM1 (derivative of maytansine), in phase III development for HER2-positive cancer. Extensive analysis of T-DM1 in preclinical studies has shown that T-DM1 combines the distinct mechanisms of action of both DM1 and trastuzumab, and has antitumor activity in trastuzumab- and lapatinib-refractory experimental models. Clinically, T-DM1 has a consistent pharmacokinetics profile and minimal systemic exposure to free DM1, with no evidence of DM1 accumulation following repeated T-DM1 doses. Although a few covariates were shown to affect interindividual variability in T-DM1 exposure and clearance in population-pharmacokinetics analyses, the magnitude of their effect on T-DM1 exposure was not clinically relevant. Phase I and phase II clinical trials of T-DM1 as a single agent and in combination with paclitaxel, docetaxel, and pertuzumab have shown clinical activity and a favorable safety profile in patients with HER2-positive metastatic breast cancer. Two randomized phase III trials of T-DM1 are recruiting patients: EMILIA (NCT00829166) is evaluating T-DM1 compared with lapatinib plus capecitabine, and MARIANNE (NCT01120184) is evaluating T-DM1 plus placebo versus T-DM1 plus pertuzumab versus trastuzumab plus a taxane. Additional combinations of T-DM1 (for example, with GDC-0941) and additional disease settings (early-stage HER2-positive breast cancer) are also under investigation. Data from the phase III trials and other studies of T-DM1-containing agents are eagerly awaited.
Collapse
|
346
|
Kaur A, Dasanu CA. Targeting the HER2 pathway for the therapy of lower esophageal and gastric adenocarcinoma. Expert Opin Pharmacother 2011; 12:2493-503. [PMID: 21967344 DOI: 10.1517/14656566.2011.605354] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The mysteries of complex molecular pathways of tumorigenesis are only beginning to be unraveled. Overexpression of HER2 receptors has been associated with adverse outcomes in certain malignant solid tumors. AREAS COVERED The authors give a focused review of the HER2 pathway and its importance for cancer cell survival. Similar to the situation in breast cancer, HER2 overexpression is seen in up to one-quarter of all gastric and gastroesophageal junction adenocarcinomas. The audience will also be familiarized with the existing HER2 targeted agents (both at the bench and at the bedside) for the therapy of gastric and gastroesophageal cancers. EXPERT OPINION Despite recent advances, treatment of upper gastrointestinal malignancies remains a significant challenge. Trastuzumab in combination with chemotherapy is the current standard of therapy for patients with metastatic HER2-overexpressing esophageal and gastric cancers. The activity of lapatinib, an active agent in advanced HER2-positive breast cancer, is now being tested in HER2-overexpressing esophageal and gastric adenocarcinomas. A variety of monoclonal antibodies and tyrosine kinase inhibitors with affinity for HER2 are in development and may improve further the outcomes of these malignancies.
Collapse
Affiliation(s)
- Antarpreet Kaur
- University of Connecticut Medical Center, Department of Internal Medicine, Farmington, CT 06030, USA.
| | | |
Collapse
|
347
|
Jørgensen JT. A challenging drug development process in the era of personalized medicine. Drug Discov Today 2011; 16:891-897. [PMID: 21945860 DOI: 10.1016/j.drudis.2011.09.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 09/08/2011] [Accepted: 09/13/2011] [Indexed: 11/15/2022]
Abstract
After years of hesitation a larger number of pharmaceutical and biotech companies are now supporting the idea of a more individualized pharmacotherapy. The companies that are now involved in this research-demanding area will have to face several challenges. The key factor for success will be an in-depth molecular understanding of the pathophysiology and the mechanism of the drug under development. The one-disease-one-drug-one-target paradigm that has been prevailing for decades is history. Most diseases are heterogeneous and based on molecular profiling they can be divided into biological subgroups that each requires a specific pharmacological intervention.
Collapse
|
348
|
Ahn ER, Vogel CL. Dual HER2-targeted approaches in HER2-positive breast cancer. Breast Cancer Res Treat 2011; 131:371-83. [PMID: 21956210 DOI: 10.1007/s10549-011-1781-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 09/13/2011] [Indexed: 01/24/2023]
Abstract
Approximately 15-20% of all breast cancers are human epidermal growth factor receptor 2 (HER2) positive, with clinical studies having validated the HER2 receptor tyrosine kinase pathway as an important therapeutic target. Presently, two HER2-targeted therapies are approved by the Food and Drug Administration for treatment of HER2-positive breast cancer: the HER2-targeted humanized monoclonal antibody trastuzumab and the small-molecule tyrosine kinase inhibitor lapatinib. Despite use of these HER2-targeted agents, many patients still experience disease progression. For this reason, numerous new agents and therapeutic strategies are under investigation. Based on preclinical data suggesting synergistic effects from dual therapy targeting HER2, clinical trials that test the effects of combining anti-HER2 agents have been conducted and are ongoing. Here, we review recently presented data from several clinical trials, which indicate that the strategy of combining HER2 blockade therapies can offer greater clinical efficacy, with adverse effects of varying degrees. Specifically, we review new data reported at the 2010 San Antonio Breast Cancer Symposium (SABCS 2010), including the phase II NeoSphere and phase III NeoALTTO clinical trials, and data from three clinical trials reported at the 2011 American Society of Clinical Oncology (ASCO 2011) meeting. Together these trials elucidate the potential role of combining trastuzumab with lapatinib or pertuzumab. We also discuss additional ongoing studies that will help further define the role of dual HER2 blockade therapies and its impact on clinical practice.
Collapse
Affiliation(s)
- Eugene R Ahn
- Sylvester Comprehensive Cancer Center, 1475 NW 12th Avenue Suite 3300, Miami, FL 33136, USA.
| | | |
Collapse
|
349
|
Sachdev JC, Jahanzeb M. Blockade of the HER family of receptors in the treatment of HER2-positive metastatic breast cancer. Clin Breast Cancer 2011; 12:19-29. [PMID: 21903480 DOI: 10.1016/j.clbc.2011.07.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 06/29/2011] [Accepted: 07/08/2011] [Indexed: 11/29/2022]
Abstract
Breast cancer is the most common type of cancer among women and the second leading cause of cancer death in the United States. Metastatic breast cancer is considered incurable, and treatment is aimed at palliating symptoms, achieving remission, and prolonging survival. Treatment options for metastatic disease vary based on tumor surface markers and clinical factors in an individual patient and include cytotoxic chemotherapy, hormonal therapy, biological therapy, or some combination of these. An important molecular determinant of therapy is the human epidermal growth factor receptor 2 (HER2) positivity of the tumor, which affects response to HER2-targeted treatment. HER2 is a member of the human epidermal growth factor receptor family of receptor tyrosine kinases, also known as the HER family, which activates signaling that promotes tumorigenic cellular processes such as proliferation and evasion of apoptosis. Several targeted agents, including monoclonal antibodies and tyrosine kinase inhibitors that inhibit one or more HER family receptors have been developed that affect signaling through this pathway. Some of these, such as trastuzumab and lapatinib, have been approved for breast cancer treatment. Resistance to therapy is a challenge that limits the duration of benefit achieved with these agents. Therefore, combinations of HER family-targeted agents with other therapies such as cytotoxic agents, hormonal therapy, or inhibitors of other cellular pathways, are being developed to exploit synergy and overcome resistance mechanisms. Here we review the HER family-targeted agents currently approved or in development for HER2-positive metastatic breast cancer with a focus on strategies to overcome tumor resistance.
Collapse
|
350
|
Coleman S, Peethambaram PP, Jatoi A. Consumer beware: a systematic assessment of potential bias in the lay electronic media to examine the portrayal of "PARP" inhibitors for cancer treatment. JOURNAL OF CANCER EDUCATION : THE OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER EDUCATION 2011; 26:474-7. [PMID: 20857352 PMCID: PMC3081418 DOI: 10.1007/s13187-010-0166-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
This study examined how the lay electronic media covers poly-ADP-ribose polymerase, or "PARP," inhibitors, a class of cancer agents currently under clinical investigation. Of 771 internet links, 51 targeted the lay public. Independent review by two investigators yielded the following categorizations: 36 (71%) were "overly positive", 15 (29%) "neutral", and none "overly negative". "Overly positive" articles used: (l) overstated benefit, (2) included quotations from enthusiastic scientists, and (3) discussed single or small patient subsets. They used such phrases as "the holy grail of cancer research", "the most exciting development in cancer research in a decade or more…. it could save thousands of lives", and "we were surprised and delighted…. it's the kind of thing you don't really think will happen". Healthcare providers should be aware of the foregoing when discussing PARP inhibitors-and perhaps other novel therapies-with cancer patients.
Collapse
Affiliation(s)
- Shawnta Coleman
- Department of Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | - Aminah Jatoi
- Department of Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|