1
|
Raghav KP, Moasser MM. Molecular Pathways and Mechanisms of HER2 in Cancer Therapy. Clin Cancer Res 2023; 29:2351-2361. [PMID: 36574481 PMCID: PMC10293474 DOI: 10.1158/1078-0432.ccr-22-0283] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 09/18/2022] [Accepted: 12/16/2022] [Indexed: 12/28/2022]
Abstract
The oncogene ERBB2 encoding the receptor tyrosine-protein kinase erbB-2 (HER2) is frequently overexpressed or amplified and occasionally mutated in a variety of human cancers. The early discovery of this oncogene, its established oncogenic relevance in diverse cancers, its substantial expression on the surface of cancer cells, and its druggable catalytic activity have made it one of the most pursued targets in the history of cancer drug development. Initiatives targeting HER2 provided the early stimulus for several transformational pharmaceutical technologies, including mAbs, tyrosine kinase inhibitors, antibody-drug conjugates, and others. The seismic impact of these efforts has been felt in treatment of many cancers, including breast, gastroesophageal, lung, colorectal, and others. This impact continues to broaden with increasing indications on the horizon and a plethora of novel agents in development. However, implementation of these therapeutic strategies has been complex. The clinical translation of every one of these classes of agents has been notable for underperformance or overperformance characteristics that have informed new lines of research providing deeper insights into the mechanistic complexities and unrealized opportunities provided by this molecular target. Despite all the successes to date, the preponderance of scientific evidence indicates that the full potential of HER2 as a target for cancer therapeutics is far greater than currently realized, and numerous lines of investigation are ongoing to deepen and broaden the scope of impact of HER2 as a signaling, homing, or immunologic target. In this review, we explore the existing data and evolving paradigms surrounding this remarkable target for cancer therapy.
Collapse
Affiliation(s)
- Kanwal P.S. Raghav
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, TX, USA
| | - Mark M. Moasser
- Department of Medicine (Hematology/Oncology), Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (UCSF), San Francisco, CA, USA
| |
Collapse
|
2
|
Yang T, Kang L, Li D, Song Y. Immunotherapy for HER-2 positive breast cancer. Front Oncol 2023; 13:1097983. [PMID: 37007133 PMCID: PMC10061112 DOI: 10.3389/fonc.2023.1097983] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
Immunotherapy is a developing treatment for advanced breast cancer. Immunotherapy has clinical significance for the treatment of triple-negative breast cancers and human epidermal growth factor receptor-2 positive (HER2+) breast cancers. As a proved effective passive immunotherapy, clinical application of the monoclonal antibodies trastuzumab, pertuzumab and T-DM1 (ado-trastuzumab emtansine) has significantly improved the survival of patients with HER2+ breast cancers. Immune checkpoint inhibitors that block programmed death receptor-1 and its ligand (PD-1/PD-L1) have also shown benefits for breast cancer in various clinical trials. Adoptive T-cell immunotherapies and tumor vaccines are emerging as novel approaches to treating breast cancer, but require further study. This article reviews recent advances in immunotherapy for HER2+ breast cancers.
Collapse
|
3
|
Moasser MM. Inactivating amplified HER2: challenges, dilemmas, and future directions. Cancer Res 2022; 82:2811-2820. [PMID: 35731927 DOI: 10.1158/0008-5472.can-22-1121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/10/2022] [Accepted: 06/15/2022] [Indexed: 11/16/2022]
Abstract
The pharmaceutical inactivation of driver oncogenes has revolutionized the treatment of cancer replacing cytotoxic chemotherapeutic approaches with kinase inhibitor therapies for many types of cancers. This approach has not yet been realized for the treatment of HER2-amplified cancers. The monotherapy activities associated with HER2-targeting antibodies and kinase inhibitors are modest, and their clinical use has been in combination with, and not in replacement of cytotoxic chemotherapies. This stands in sharp contrast to achievements in the treatment of many other oncogene-driven cancers. The mechanism-based treatment hypothesis regarding the inactivation of HER2 justifies expectations far beyond what is currently realized. Overcoming this barrier requires mechanistic insights that can fuel new directions for pursuit, but scientific investigation of this treatment hypothesis, particularly with regards to trastuzumab, has been complicated by conflicting and confusing data sets, ironclad dogma, and mechanistic conclusions that have repeatedly failed to translate clinically. We are now approaching a point of convergence regarding the challenges and resiliency in this tumor driver, and I will provide here a review and opinion to inform where we currently stand with this treatment hypothesis and where the future potential lies.
Collapse
Affiliation(s)
- Mark M Moasser
- University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
4
|
Campbell MR, Ruiz-Saenz A, Zhang Y, Peterson E, Steri V, Oeffinger J, Sampang M, Jura N, Moasser MM. Extensive conformational and physical plasticity protects HER2-HER3 tumorigenic signaling. Cell Rep 2022; 38:110285. [PMID: 35108526 PMCID: PMC8865943 DOI: 10.1016/j.celrep.2021.110285] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 08/30/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022] Open
Abstract
Surface-targeting biotherapeutic agents have been successful in treating HER2-amplified cancers through immunostimulation or chemodelivery but have failed to produce effective inhibitors of constitutive HER2-HER3 signaling. We report an extensive structure-function analysis of this tumor driver, revealing complete uncoupling of intracellular signaling and tumorigenic function from regulation or constraints from their extracellular domains (ECDs). The canonical HER3 ECD conformational changes and exposure of the dimerization interface are nonessential, and the entire ECDs of HER2 and HER3 are redundant for tumorigenic signaling. Restricting the proximation of partner ECDs with bulk and steric clash through extremely disruptive receptor engineering leaves tumorigenic signaling unperturbed. This is likely due to considerable conformational flexibilities across the span of these receptor molecules and substantial undulations in the plane of the plasma membrane, none of which had been foreseen as impediments to targeting strategies. The massive overexpression of HER2 functionally and physically uncouples intracellular signaling from extracellular constraints.
Collapse
Affiliation(s)
- Marcia R Campbell
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ana Ruiz-Saenz
- Departments of Cell Biology & Medical Oncology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Yuntian Zhang
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Elliott Peterson
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Veronica Steri
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Julie Oeffinger
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Maryjo Sampang
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Natalia Jura
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mark M Moasser
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
5
|
Mandó P, Rivero SG, Rizzo MM, Pinkasz M, Levy EM. Targeting ADCC: A different approach to HER2 breast cancer in the immunotherapy era. Breast 2021; 60:15-25. [PMID: 34454323 PMCID: PMC8399304 DOI: 10.1016/j.breast.2021.08.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/06/2021] [Accepted: 08/16/2021] [Indexed: 11/28/2022] Open
Abstract
The clinical outcome of patients with human epidermal growth factor receptor 2 (HER2) amplified breast carcinoma (BC) has improved with the development of anti-HER2 targeted therapies. However, patients can experience disease recurrence after curative intent and disease progression in the metastatic setting. In the current era of evolving immunotherapy agents, the understanding of the immune response against HER2 tumor cells developed by anti-HER2 antibodies (Abs) is rapidly evolving. Trastuzumab therapy promotes Natural Killer (NK) cell activation in patients with BC overexpressing HER2, indicating that the efficacy of short-term trastuzumab monotherapy, albeit direct inhibition of HER, could also be related with antibody-dependent cell-mediated cytotoxicity (ADCC). Currently, dual HER2 blockade using trastuzumab and pertuzumab is the standard of care in early and advanced disease as this combination could confer an additive effect in ADCC. In patients with disease relapse or progression, ADCC may be hampered by several factors such as FcγRIIIa polymorphism and an immunosuppressive environment, among others. Hence, new drug development strategies are being investigated aiming to boost the ADCC response triggered by anti-HER2 therapy. In this review, we summarize these strategies and the rationale, through mAbs engineering and combinatorial strategies, focusing on clinical results and ongoing trials.
Collapse
Affiliation(s)
- Pablo Mandó
- Fundación Cáncer, Ciudad Autónoma de Buenos Aires, Argentina; Centro de Educación Médica e Investigaciones Clínicas "Norberto Quirno" (CEMIC), Ciudad Autónoma de Buenos Aires, Argentina.
| | - Sergio G Rivero
- Instituto Alexander Fleming, Ciudad Autónoma de Buenos Aires, Argentina
| | - Manglio M Rizzo
- Cancer Immunobiology, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET, Universidad Austral, Derqui-Pilar, Argentina; Department of Medical Oncology, Hospital Universitario Austral, Derqui-Pilar, Argentina
| | - Marina Pinkasz
- Centro de Investigaciones Oncológicas, Fundación Cáncer, Ciudad Autónoma de Buenos Aires, Argentina
| | - Estrella M Levy
- Centro de Investigaciones Oncológicas, Fundación Cáncer, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
6
|
Affiliation(s)
- Ana Ruiz-Saenz
- Ana Ruiz-Saenz, University of California at San Francisco, San Francisco, CA; and Mark M. Moasser, University of California at San Francisco, San Francisco, CA
| | - Mark M Moasser
- Ana Ruiz-Saenz, University of California at San Francisco, San Francisco, CA; and Mark M. Moasser, University of California at San Francisco, San Francisco, CA
| |
Collapse
|
7
|
Bang YJ, Giaccone G, Im SA, Oh DY, Bauer TM, Nordstrom JL, Li H, Chichili GR, Moore PA, Hong S, Stewart SJ, Baughman JE, Lechleider RJ, Burris HA. First-in-human phase 1 study of margetuximab (MGAH22), an Fc-modified chimeric monoclonal antibody, in patients with HER2-positive advanced solid tumors. Ann Oncol 2017; 28:855-861. [PMID: 28119295 PMCID: PMC6246722 DOI: 10.1093/annonc/mdx002] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Margetuximab is an anti-HER2 antibody that binds with elevated affinity to both the lower and higher affinity forms of CD16A, an Fc-receptor important for antibody dependent cell-mediated cytotoxicity (ADCC) against tumor cells. A Phase 1 study was initiated to evaluate the toxicity profile, maximum tolerated dose (MTD), pharmacokinetics, and antitumor activity of margetuximab in patients with HER2-overexpressing carcinomas. PATIENTS AND METHODS Patients with HER2-positive breast or gastric cancer, or other carcinomas that overexpress HER2, for whom no standard therapy was available, were treated with margetuximab by intravenous infusion at doses of 0.1-6.0 mg/kg for 3 of every 4 weeks (Regimen A) or once every 3 weeks (10-18 mg/kg) (Regimen B). RESULTS Sixty-six patients received margetuximab (34 patients for Regimen A and 32 patients for Regimen B). The MTD was not reached for either regimen. Treatment was well-tolerated, with mostly Grade 1 and 2 toxicities consisting of constitutional symptoms such as pyrexia, nausea, anemia, diarrhea, and fatigue. Among 60 response-evaluable patients, confirmed partial responses and stable disease were observed in 7 (12%) and 30 (50%) patients, respectively; 26 (70%) of these patients had received prior HER2-targeted therapy. Tumor reductions were observed in over half (18/23, 78%) of response-evaluable patients with breast cancer including durable (>30 weeks) responders. Ex vivo analyses of patient peripheral blood mononuclear cell samples confirmed the ability of margetuximab to support enhanced ADCC compared with trastuzumab. CONCLUSIONS Margetuximab was well-tolerated and has promising single-agent activity. Further development efforts of margetuximab as single agent and in combination with other therapeutic agents are ongoing. TRIAL REGISTRATION ID NCT01148849.
Collapse
Affiliation(s)
- Y. J. Bang
- Department of Internal Medicine, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - G. Giaccone
- Department of Medical Oncology, National Cancer Institute, Bethesda
| | - S. A. Im
- Department of Internal Medicine, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - D. Y. Oh
- Department of Internal Medicine, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - T. M. Bauer
- Department of Drug Development, Sarah Cannon Research Institute and Tennessee Oncology, Nashville
| | | | - H. Li
- MacroGenics, Inc, Rockville, and South San Francisco, USA
| | - G. R. Chichili
- MacroGenics, Inc, Rockville, and South San Francisco, USA
| | - P. A. Moore
- MacroGenics, Inc, Rockville, and South San Francisco, USA
| | - S. Hong
- MacroGenics, Inc, Rockville, and South San Francisco, USA
| | - S. J. Stewart
- MacroGenics, Inc, Rockville, and South San Francisco, USA
| | - J. E. Baughman
- MacroGenics, Inc, Rockville, and South San Francisco, USA
| | | | - H. A. Burris
- Department of Drug Development, Sarah Cannon Research Institute and Tennessee Oncology, Nashville
| |
Collapse
|
8
|
Amin DN, Ahuja D, Yaswen P, Moasser MM. A TORC2-Akt Feed-Forward Topology Underlies HER3 Resiliency in HER2-Amplified Cancers. Mol Cancer Ther 2015; 14:2805-17. [PMID: 26438156 PMCID: PMC4674361 DOI: 10.1158/1535-7163.mct-15-0403] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/18/2015] [Indexed: 11/16/2022]
Abstract
The requisite role of HER3 in HER2-amplified cancers is beyond what would be expected as a dimerization partner or effector substrate and it exhibits a substantial degree of resiliency that mitigates the effects of HER2-inhibitor therapies. To better understand the roots of this resiliency, we conducted an in-depth chemical-genetic interrogation of the signaling network downstream of HER3. A unique attribute of these tumors is the deregulation of TORC2. The upstream signals that ordinarily maintain TORC2 signaling are lost in these tumors, and instead TORC2 is driven by Akt. We find that in these cancers HER3 functions as a buffering arm of an Akt-TORC2 feed-forward loop that functions as a self-perpetuating module. This network topology alters the role of HER3 from a conditionally engaged ligand-driven upstream physiologic signaling input to an essential component of a concentric signaling throughput highly competent at preservation of homeostasis. The competence of this signaling topology is evident in its response to perturbation at any of its nodes. Thus, a critical pathophysiologic event in the evolution of HER2-amplified cancers is the loss of the input signals that normally drive TORC2 signaling, repositioning it under Akt dependency, and fundamentally altering the role of HER3. This reprogramming of the downstream network topology is a key aspect in the pathogenesis of HER2-amplified cancers and constitutes a formidable barrier in the targeted therapy of these cancers.
Collapse
Affiliation(s)
- Dhara N Amin
- Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Deepika Ahuja
- Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Paul Yaswen
- Lawrence Berkeley National Laboratory, Berkeley, California
| | - Mark M Moasser
- Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.
| |
Collapse
|
9
|
Miolo G, Muraro E, Martorelli D, Lombardi D, Scalone S, Spazzapan S, Massarut S, Perin T, Viel E, Comaro E, Talamini R, Bidoli E, Turchet E, Crivellari D, Dolcetti R. Anthracycline-free neoadjuvant therapy induces pathological complete responses by exploiting immune proficiency in HER2+ breast cancer patients. BMC Cancer 2014; 14:954. [PMID: 25512030 PMCID: PMC4302069 DOI: 10.1186/1471-2407-14-954] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 12/11/2014] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Neoadjuvant Chemotherapy (NC) including trastuzumab induces a high rate of pathological Complete Responses (pCR) in patients with locally advanced HER2-overexpressing Breast Cancer (BC), but is penalized by a severe cardiotoxicity when combined with anthracyclines. A phase II study was designed to assess whether an anthracycline-free NC regimen based on the early addition of trastuzumab to paclitaxel may increase the pCR rate without inducing severe cardiotoxicity in patients with locally advanced HER2-overexpressing BC. Immunomonitoring was performed to assess the contribution of patients' immunological background to the induction of clinical responses. METHODS Stage II-III HER2-positive BC patients received 24 weeks paclitaxel and trastuzumab NC, followed by 1 year adjuvant trastuzumab ± hormonal and/or radio-therapy. Assessment of pCR rate was the primary endpoint. A group of HER2-negative BC patients treated with neoadjuvant taxanes and anthracyclines was included. Serum levels of 10 cytokines and the efficiency of trastuzumab-mediated antibody-dependent cell cytotoxicity (ADCC) were monitored in vitro every 3 months. RESULTS From July 2006 to February 2013, we enrolled 109 patients including 46 evaluable HER2-positive cases. A pCR rate of 50% was reached and no severe cardiotoxicity occurred. Serum cytokine profiling revealed only an IL-10 decrease (P = 0.02) in patients achieving a partial response, while HER2-negative patients disclosed marked cytokines changes. Compared to the unfavourable F/F genotype, patients carrying the V allele in the FcγRIIIa-158 polymorphism showed a higher efficacy of trastuzumab-ADCC throughout treatment (P ≤0.05). CONCLUSIONS In the absence of anthracyclines, trastuzumab and paclitaxel induced a high rate of pCR, exploiting the synergy between the immunomodulating properties of these drugs and the retained immunological proficiency of patients with HER2-overexpressing BC. TRIAL REGISTRATION Trial registration number: NCT02307227, registered on ClinicalTrials.gov (http://www.clinicaltrials.gov, November 26, 2014).
Collapse
Affiliation(s)
- Gianmaria Miolo
- />Department of Medical Oncology, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Elena Muraro
- />Cancer Bio-Immunotherapy Unit, Department of Medical Oncology, C.R.O, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Debora Martorelli
- />Cancer Bio-Immunotherapy Unit, Department of Medical Oncology, C.R.O, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Davide Lombardi
- />Department of Medical Oncology, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Simona Scalone
- />Department of Medical Oncology, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Simon Spazzapan
- />Department of Medical Oncology, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Samuele Massarut
- />Division of Breast Surgical Oncology, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Tiziana Perin
- />Pathology Unit, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Elda Viel
- />Cardiology, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Elisa Comaro
- />Cancer Bio-Immunotherapy Unit, Department of Medical Oncology, C.R.O, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Renato Talamini
- />Unit of Epidemiology and Biostatistics, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Ettore Bidoli
- />Unit of Epidemiology and Biostatistics, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Elisa Turchet
- />Scientific Direction, CRO Aviano, IRCCS, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Diana Crivellari
- />Department of Medical Oncology, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Riccardo Dolcetti
- />Cancer Bio-Immunotherapy Unit, Department of Medical Oncology, C.R.O, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| |
Collapse
|
10
|
|
11
|
Del Mastro L, Lambertini M, Bighin C, Levaggi A, D'Alonzo A, Giraudi S, Pronzato P. Trastuzumab as first-line therapy in HER2-positive metastatic breast cancer patients. Expert Rev Anticancer Ther 2012; 12:1391-405. [PMID: 23072512 DOI: 10.1586/era.12.107] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Trastuzumab is a humanized monoclonal antibody against the extracellular domain of hEGF receptor-2 (HER2). Trastuzumab in combination with chemotherapy has proven efficacy in treating both early and metastatic HER2-positive breast cancer. In the metastatic setting, the addition of trastuzumab to chemotherapy is associated with a statistically significant longer time to disease progression, higher rate of objective response and improvement in overall survival. Trastuzumab efficacy is not influenced by hormone receptor status, but differences in median overall survival exist between HER2-positive and HER2-negative states. Reassessment of the benefit of re-exposing patients with metastatic breast cancer to trastuzumab following relapse in the adjuvant setting is necessary. Ongoing research into new HER2-targeted therapies and trials involving combination anti-HER2 drug therapy without chemotherapy show promise. This review is focused on the available results obtained with the use of trastuzumab in the subset of HER2-positive breast cancer patients with metastatic disease.
Collapse
Affiliation(s)
- Lucia Del Mastro
- Istituto Di Ricovero e Cura a Carattere Scientifico Azienda Ospedaliera Universitaria San Martini-IST, Largo Rosanna Benzi, 10, 16132, Genova, Italy.
| | | | | | | | | | | | | |
Collapse
|
12
|
Puglisi F, Minisini AM, De Angelis C, Arpino G. Overcoming treatment resistance in HER2-positive breast cancer: potential strategies. Drugs 2012; 72:1175-93. [PMID: 22686613 DOI: 10.2165/11634000-000000000-00000] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Human epidermal growth factor receptor (HER)-2 overexpression or amplification occurs in about 20% of all breast cancers and results in a worse prognosis. Nevertheless, anti-HER2 treatments have recently been developed, resulting in dramatic improvements in the clinical outcome of patients with HER2-positive breast cancer. Trastuzumab has shown efficacy in early and advanced breast cancer treatment and lapatinib is currently approved for the treatment of advanced disease. Other anti-HER2 agents are being investigated. Mechanisms of resistance to trastuzumab treatment include crosstalk with heterologous receptors and amplification of HER2 signalling; amplification of the phosphoinositide 3-kinase (PI3K)/AKT pathway; alteration in binding of trastuzumab to HER2; and loss of HER2 expression. Proposed mechanisms of resistance to lapatinib involve derepression and/or activation of compensatory survival pathways through increased PI3K/AKT or estrogen receptor (ER) signalling. Several strategies to overcome resistance to anti-HER2 treatment are in different phases of development and include treatment with pertuzumab, T-DM1 and mammalian target of rapamycin (mTOR) inhibitors.
Collapse
Affiliation(s)
- Fabio Puglisi
- Department of Oncology, University Hospital of Udine, Udine, Italy.
| | | | | | | |
Collapse
|
13
|
Bartusik D, Tomanek B. Detection of fluorine labeled Herceptin using cellular 19F MRI ex vivo. J Pharm Biomed Anal 2010; 51:894-900. [DOI: 10.1016/j.jpba.2009.10.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2009] [Revised: 10/06/2009] [Accepted: 10/06/2009] [Indexed: 11/30/2022]
|
14
|
Pikó B. [Lapatinib treatment-option in trastuzumab-resistant breast cancer]. Magy Onkol 2010; 53:369-75. [PMID: 20071309 DOI: 10.1556/monkol.53.2009.4.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
HER2 is overexpressed in 20-25% of breast cancers and is associated with an aggressive phenotype and poor prognosis. Lapatinib is a dual tyrosine kinase inhibitor selective for inhibition of epidermal growth factor receptor (EGFR1/ErbB1) and HER2/ErbB2. Having more targets, probably its antitumor activity could be more efficient. Preclinical data reveal that lapatinib has activity in trastuzumab-resistant cell lines as well as synergistic activity with trastuzumab. Phase I clinical trials have also shown that lapatinib is well tolerated, with mild diarrhea and skin rush as common toxic effects and low incidence of cardiotoxicity. Lapatinib can cross the blood-brain barrier and might therefore have a role in preventing central-nervous-system progression. In a pivotal phase III trial, a combination of lapatinib and capecitabine almost doubled time to disease progression when compared to capecitabine alone (8.4 vs. 4.1 months) in women with HER2/ErbB2-positive advanced or metastatic breast cancer previously treated with anthracyclin, taxanes and trastuzumab. The overall survival was 15.6 vs. 15.4 months. Several clinical trials that explore the efficacy of lapatinib in combination with conventional chemotherapeutic agents, hormone therapy and other target therapies are ongoing in advanced breast cancer or in neo-adjuvant and adjuvant settings.
Collapse
Affiliation(s)
- Béla Pikó
- Békés Megyei Képviselôtestület Pándy Kálmán Kórháza Megyei Onkológiai Központ 5700 Gyula Semmelweis u. 1.
| |
Collapse
|
15
|
Spector NL, Blackwell KL. Understanding the mechanisms behind trastuzumab therapy for human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol 2009; 27:5838-47. [PMID: 19884552 DOI: 10.1200/jco.2009.22.1507] [Citation(s) in RCA: 372] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
PURPOSE Targeted therapy with the humanized monoclonal antibody trastuzumab has become a mainstay for human epidermal growth factor receptor 2 (HER2) -positive breast cancer (BC). The mechanisms of action of trastuzumab have not been fully elucidated, and data available to date are reviewed here. The impact of the mechanisms of action on clinical benefit also is discussed. METHODS An extensive literature review of trastuzumab and proposed mechanisms of action was performed. RESULTS At least five potential extracellular and intracellular antitumor mechanisms of trastuzumab have been identified in the preclinical setting. These include activation of antibody-dependent cellular cytotoxicity, inhibition of extracellular domain cleavage, abrogation of intracellular signaling, reduction of angiogenesis, and decreased DNA repair. These effects lead to tumor cell stasis and/or death. Clinical benefit from trastuzumab-based therapy in both early and advanced BC has been demonstrated. The benefit of trastuzumab use beyond progression has also been shown, which indicates the need for continuous suppression of the HER2 pathway. Targeting both HER2, with various approaches, and other pathways may enhance the clinical benefit observed with trastuzumab and overcome potential resistance. Novel combinations include pertuzumab (a HER2 dimerization inhibitor), lapatinib (a HER1/HER2 tyrosine kinase inhibitor), bevacizumab (an antiangiogenic agent), tanespimycin (a heat shock protein inhibitor), antiestrogen therapies, and an antibody-drug conjugate (trastuzumab-DM1). CONCLUSION Trastuzumab is the foundation of care for patients with HER2-positive BC. Emerging data from studies of other targeted agents may provide alternative treatment combinations to maximize the clinical benefit from trastuzumab and prevent or delay resistance. The continued development of trastuzumab highlights promising treatment approaches for the future.
Collapse
Affiliation(s)
- Neil L Spector
- Duke Translational Research Oncology Program, Duke University Hospital, Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC 27710, USA.
| | | |
Collapse
|
16
|
Trastuzumab as adjuvant systemic therapy for HER2-positive breast cancer. ACTA ACUST UNITED AC 2008; 6:93-104. [DOI: 10.1038/ncponc1298] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2007] [Accepted: 05/21/2008] [Indexed: 01/24/2023]
|
17
|
de la Cruz-Merino L, Grande-Pulido E, Albero-Tamarit A, Codes-Manuel de Villena ME. Cancer and immune response: old and new evidence for future challenges. Oncologist 2008; 13:1246-54. [PMID: 19056856 DOI: 10.1634/theoncologist.2008-0166] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cancer may occur as a result of abnormal host immune system tolerance. Recent studies have confirmed the occurrence of spontaneous and induced antitumor immune responses expressed as the presence of tumor-infiltrating T cells in the tumor microenvironment in some cancer models. This finding has been recognized as a good prognostic factor in several types of tumors. Some chemotherapy agents, such as anthracyclines and gemcitabine, are effective boosters of the immune response through tumor-specific antigen overexpression after apoptotic tumor cell destruction. Other strategies, such as GM-CSF or interleukin-2, are pursued to increase immune cell availability in the tumor vicinity, and thus improve both antigen presentation and T-cell activation and proliferation. In addition, cytotoxic T lymphocyte antigen 4-blocking monoclonal antibodies enhance immune activity by prolonging T-cell activation. Strategies to stimulate the dormant immune system against tumors are varied and warrant further investigation of their applications to cancer therapy in the future.
Collapse
|
18
|
Antibodies targeted to TRAIL receptor-2 and ErbB-2 synergize in vivo and induce an antitumor immune response. Proc Natl Acad Sci U S A 2008; 105:16254-9. [PMID: 18838682 DOI: 10.1073/pnas.0806849105] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Despite the development of human epidermal growth factor receptor-2 (ErbB-2/HER2)-targeted therapies, there remains an unmet medical need for breast cancer patients with ErbB-2 overexpression. We investigated the therapeutic activity of an agonist mAb to mouse tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor-2 (DR5) against ErbB2-driven breast cancer. Established tumors in BALB/c transgenic mice expressing a constitutively active ErbB-2/neuT were treated with anti-DR5 mAb and/or anti-ErbB-2 mAb and monitored for tumor progression. Treatment with anti-DR5 or anti-ErbB2 mAb as single agents significantly delayed tumor growth, although all tumors eventually progressed. Remarkably, treatment with a combination of anti-DR5 and anti-ErbB-2 mAbs induced complete response in a majority of mice. In vivo blockade of CD11b(+) cells, but not natural killer cell depletion, significantly abrogated the early antitumor response. Notably, depletion of CD8(+) T cells provoked primary and secondary tumor relapse, revealing the induction of antitumor immunity by the combination treatment. Combined therapy with anti-DR5 and anti-ErbB-2 mAbs further significantly suppressed the growth of advanced spontaneous tumors in ErbB-2/neuT transgenic mice, even when treatment was delayed until tumors were palpable. We thus demonstrated that the combination of anti-DR5 and anti-ErbB2 mAbs might be an effective form of treatment for ErbB-2-overexpressing breast cancer.
Collapse
|
19
|
Abstract
During the carcinogenesis of colorectal cancer in about half of the cases K-RAS, while much less frequently B-RAF mutation occur in early adenomas. While K-RAS mutant tumors are more likely present in male patients, B-RAF mutant tumors develop more frequently in females and are independent of the microsatellite status. Colorectal cancers are characterized by EGFR expression; the gene is not mutated, rarely amplified and increased copy number is due to chromosomal polysomy. This phenotype/genotype of colorectal cancer lent support to the introduction of anti-EGFR antibody therapies. For a while positive EGFR expression status of the tumor was the basis of patient selection for these targeted therapies in colorectal cancer. Monotherapies with the two available anti-EGFR antibodies of chemoresistant colorectal cancers resulted in appr. 10% objective response rate, which was independent of the level of EGFR expression. In case of panitumumab it was discovered that the efficacy of this targeted therapy depends on the K-RAS mutant status of the tumors. Furthermore, preliminary data suggest that cetuximab combined with chemotherapy is effective also exclusively in K-RAS wild-type tumors. Based on these data it is safe to say that K-RAS mutant status of colorectal cancer is a negative predictor for EGFR-targeted therapies of colorectal cancer. Accordingly, it is necessary to determine the K-RAS status of colorectal cancer before making therapeutic decisions.
Collapse
Affiliation(s)
- József Tímár
- Országos Onkológiai Intézet 1122 Budapest Ráth György u. 7-9.
| | | |
Collapse
|